Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
PLoS Pathog ; 20(1): e1011640, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38215165

ABSTRACT

Retroviral reverse transcription starts within the capsid and uncoating and reverse transcription are mutually dependent. There is still debate regarding the timing and cellular location of HIV's uncoating and reverse transcription and whether it occurs solely in the cytoplasm, nucleus or both. HIV can infect non-dividing cells because there is active transport of the preintegration complex (PIC) across the nuclear membrane, but Murine Leukemia Virus (MLV) is thought to depend on cell division for replication and whether MLV uncoating and reverse transcription is solely cytoplasmic has not been studied. Here, we used NIH3T3 and primary mouse dendritic cells to determine where the different stages of reverse transcription occur and whether cell division is needed for nuclear entry. Our data strongly suggest that in both NIH3T3 cells and dendritic cells (DCs), the initial step of reverse transcription occurs in the cytoplasm. However, we detected MLV RNA/DNA hybrid intermediates in the nucleus of dividing NIH3T3 cells and non-dividing DCs, suggesting that reverse transcription can continue after nuclear entry. We also confirmed that the MLV PIC requires cell division to enter the nucleus of NIH3T3 cells. In contrast, we show that MLV can infect non-dividing primary DCs, although integration of MLV DNA in DCs still required the viral p12 protein. Knockdown of several nuclear pore proteins dramatically reduced the appearance of integrated MLV DNA in DCs but not NIH3T3 cells. Additionally, MLV capsid associated with the nuclear pore proteins NUP358 and NUP62 during infection. These findings suggest that simple retroviruses, like the complex retrovirus HIV, gain nuclear entry by traversing the nuclear pore complex in non-mitotic cells.


Subject(s)
HIV Infections , Nuclear Pore Complex Proteins , Animals , Mice , Nuclear Pore Complex Proteins/genetics , NIH 3T3 Cells , Leukemia Virus, Murine/genetics , Viral Proteins , Capsid Proteins , Retroviridae , DNA , Dendritic Cells
2.
Eur J Immunol ; 53(12): e2350546, 2023 12.
Article in English | MEDLINE | ID: mdl-37751619

ABSTRACT

Cryopreservation of mouse thymus depletes donor thymocytes but preserves thymus function when transplanted after thawing into athymic mice. No differences in immune reconstitution were observed between fresh and frozen/thawed transplants suggesting that donor thymocyte depletion does not affect outcome. Thus, cryopreservation of thymus may improve outcomes in thymus transplant patients.


Subject(s)
Immune Reconstitution , Thymocytes , Humans , Animals , Mice , Thymus Gland , Cryopreservation
3.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34323272

ABSTRACT

During positive selection at the transition from CD4+CD8+ double-positive (DP) to single-positive (SP) thymocyte, TCR signalling results in appropriate MHC restriction and signals for survival and progression. We show that the pioneer transcription factors Foxa1 and Foxa2 are required to regulate RNA splicing during positive selection of mouse T cells and that Foxa1 and Foxa2 have overlapping/compensatory roles. Conditional deletion of both Foxa1 and Foxa2 from DP thymocytes reduced positive selection and development of CD4SP, CD8SP and peripheral naïve CD4+ T cells. Foxa1 and Foxa2 regulated the expression of many genes encoding splicing factors and regulators, including Mbnl1, H1f0, Sf3b1, Hnrnpa1, Rnpc3, Prpf4b, Prpf40b and Snrpd3. Within the positively selecting CD69+DP cells, alternative RNA splicing was dysregulated in the double Foxa1/Foxa2 conditional knockout, leading to >850 differentially used exons. Many genes important for this stage of T-cell development (Ikzf1-3, Ptprc, Stat5a, Stat5b, Cd28, Tcf7) and splicing factors (Hnrnpab, Hnrnpa2b1, Hnrnpu, Hnrnpul1, Prpf8) showed multiple differentially used exons. Thus, Foxa1 and Foxa2 are required during positive selection to regulate alternative splicing of genes essential for T-cell development, and, by also regulating splicing of splicing factors, they exert widespread control of alternative splicing.


Subject(s)
Alternative Splicing/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , RNA Splicing/genetics , Thymocytes/physiology , Animals , Exons/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Splicing Factors/genetics , T-Lymphocytes/physiology , Thymus Gland/physiology
4.
Blood ; 140(1): 25-37, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35507686

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of immature T lymphocytes, associated with higher rates of induction failure compared with those in B cell acute lymphoblastic leukemia. The potent immunotherapeutic approaches applied in B cell acute lymphoblastic leukemia, which have revolutionized the treatment paradigm, have proven more challenging in T-ALL, largely due to a lack of target antigens expressed on malignant but not healthy T cells. Unlike B cell depletion, T-cell aplasia is highly toxic. Here, we show that the chemokine receptor CCR9 is expressed in >70% of cases of T-ALL, including >85% of relapsed/refractory disease, and only on a small fraction (<5%) of normal T cells. Using cell line models and patient-derived xenografts, we found that chimeric antigen receptor (CAR) T-cells targeting CCR9 are resistant to fratricide and have potent antileukemic activity both in vitro and in vivo, even at low target antigen density. We propose that anti-CCR9 CAR-T cells could be a highly effective treatment strategy for T-ALL, avoiding T cell aplasia and the need for genome engineering that complicate other approaches.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Antigens, CD19 , Humans , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell , T-Lymphocytes
5.
Development ; 147(19)2020 10 07.
Article in English | MEDLINE | ID: mdl-32907850

ABSTRACT

Pre-T-cell receptor (TCR) signal transduction is required for developing thymocytes to differentiate from CD4-CD8- double-negative (DN) cell to CD4+CD8+ double-positive (DP) cell. Notch signalling is required for T-cell fate specification and must be maintained throughout ß-selection, but inappropriate Notch activation in DN4 and DP cells is oncogenic. Here, we show that pre-TCR signalling leads to increased expression of the transcriptional repressor Bcl6 and that Bcl6 is required for differentiation to DP. Conditional deletion of Bcl6 from thymocytes reduced pre-TCR-induced differentiation to DP cells, disrupted expansion and enrichment of intracellular TCRß+ cells within the DN population and increased DN4 cell death. Deletion also increased Notch1 activation and Notch-mediated transcription in the DP population. Thus, Bcl6 is required in thymocyte development for efficient differentiation from DN3 to DP and to attenuate Notch1 activation in DP cells. Given the importance of inappropriate NOTCH1 signalling in T-cell acute lymphoblastic leukaemia (T-ALL), and the involvement of BCL6 in other types of leukaemia, this study is important to our understanding of T-ALL.


Subject(s)
Receptor, Notch1/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Flow Cytometry , Genotype , Mice , Receptor, Notch1/genetics , Signal Transduction/genetics , Signal Transduction/physiology
6.
PLoS Pathog ; 17(6): e1009662, 2021 06.
Article in English | MEDLINE | ID: mdl-34097709

ABSTRACT

Signal-regulatory protein alpha (SIRPA) is a well-known inhibitor of phagocytosis when it complexes with CD47 expressed on target cells. Here we show that SIRPA decreased in vitro infection by a number of pathogenic viruses, including New World and Old World arenaviruses, Zika virus, vesicular stomatitis virus and pseudoviruses bearing the Machupo virus, Ebola virus and SARS-CoV-2 glycoproteins, but not HSV-1, MLV or mNoV. Moreover, mice with targeted mutation of the Sirpa gene that renders it non-functional were more susceptible to infection with the New World arenaviruses Junín virus vaccine strain Candid 1 and Tacaribe virus, but not MLV or mNoV. All SIRPA-inhibited viruses have in common the requirement for trafficking to a low pH endosomal compartment. This was clearly demonstrated with SARS-CoV-2 pseudovirus, which was only inhibited by SIRPA in cells in which it required trafficking to the endosome. Similar to its role in phagocytosis inhibition, SIRPA decreased virus internalization but not binding to cell surface receptors. We also found that increasing SIRPA levels via treatment with IL-4 led to even greater anti-viral activity. These data suggest that enhancing SIRPA's activity could be a target for anti-viral therapies.


Subject(s)
Endocytosis , RNA Viruses/immunology , Receptors, Immunologic/physiology , Virus Internalization , Animals , Antiviral Agents/pharmacology , Cell Line , Cell Membrane/virology , Chlorocebus aethiops , Drug Delivery Systems , Integrins/immunology , Interleukin-4/pharmacology , Mice , Mice, Knockout , Protein Domains , Receptors, Immunologic/genetics , Vero Cells
7.
Proc Natl Acad Sci U S A ; 117(32): 19497-19506, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719120

ABSTRACT

Understanding the genetics of susceptibility to infectious agents is of great importance to our ability to combat disease. Here, we show that voltage-gated calcium channels (VGCCs) are critical for cellular binding and entry of the New World arenaviruses Junín and Tacaribe virus, suggesting that zoonosis via these receptors could occur. Moreover, we demonstrate that α1s haploinsufficiency renders cells and mice more resistant to infection by these viruses. In addition to being more resistant to infection, haploinsufficient cells and mice required a lower dosage of VGCC antagonists to block infection. These studies underscore the importance of genetic variation in susceptibility to both viruses and pharmaceutics.


Subject(s)
Arenaviridae Infections/genetics , Calcium Channels, L-Type/genetics , Disease Resistance/genetics , Animals , Arenaviridae Infections/drug therapy , Arenaviruses, New World/physiology , Calcium Channel Agonists/pharmacology , Calcium Channel Agonists/therapeutic use , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Heterozygote , Humans , Mice , Mice, Mutant Strains , Mutation , Virus Attachment/drug effects , Virus Internalization/drug effects
8.
J Virol ; 95(22): e0124421, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34468176

ABSTRACT

Apolipoprotein B mRNA editing enzyme catalytic subunit 3 (APOBEC3) proteins are critical for the control of infection by retroviruses. These proteins deaminate cytidines in negative-strand DNA during reverse transcription, leading to G-to-A changes in coding strands. Uracil DNA glycosylase (UNG) is a host enzyme that excises uracils in genomic DNA, which the base excision repair machinery then repairs. Whether UNG removes uracils found in retroviral DNA after APOBEC3-mediated mutation is not clear, and whether this occurs in vivo has not been demonstrated. To determine if UNG plays a role in the repair of retroviral DNA, we used APOBEC3G (A3G) transgenic mice which we showed previously had extensive deamination of murine leukemia virus (MLV) proviruses. The A3G transgene was crossed onto an Ung and mouse Apobec3 knockout background (UNG-/-APO-/-), and the mice were infected with MLV. We found that virus infection levels were decreased in A3G UNG-/-APO-/- compared with A3G APO-/- mice. Deep sequencing of the proviruses showed that there were significantly higher levels of G-to-A mutations in proviral DNA from A3G transgenic UNG-/-APO-/- than A3G transgenic APO-/- mice, suggesting that UNG plays a role in the repair of uracil-containing proviruses. In in vitro studies, we found that cytoplasmic viral DNA deaminated by APOBEC3G was uracilated. In the absence of UNG, the uracil-containing proviruses integrated at higher levels into the genome than those made in the presence of UNG. Thus, UNG also functions in the nucleus prior to integration by nicking uracil-containing viral DNA, thereby blocking integration. These data show that UNG plays a critical role in the repair of the damage inflicted by APOBEC3 deamination of reverse-transcribed DNA. IMPORTANCE While APOBEC3-mediated mutation of retroviruses is well-established, what role the host base excision repair enzymes play in correcting these mutations is not clear. This question is especially difficult to address in vivo. Here, we use a transgenic mouse developed by our lab that expresses human APOBEC3G and also lacks the endogenous uracil DNA glycosylase (Ung) gene and show that UNG removes uracils introduced by this cytidine deaminase in MLV reverse transcripts, thereby reducing G-to-A mutations in proviruses. Furthermore, our data suggest that UNG removes uracils at two stages in infection-first, in unintegrated nuclear viral reverse-transcribed DNA, resulting in its degradation; and second, in integrated proviruses, resulting in their repair. These data suggest that retroviruses damaged by host cytidine deaminases take advantage of the host DNA repair system to overcome this damage.


Subject(s)
APOBEC-3G Deaminase/immunology , DNA, Viral/immunology , Retroviridae Infections , Retroviridae , Uracil-DNA Glycosidase/immunology , Animals , DNA Repair , HEK293 Cells , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Retroviridae/genetics , Retroviridae/immunology , Retroviridae Infections/immunology , Retroviridae Infections/virology
9.
PLoS Biol ; 17(2): e3000137, 2019 02.
Article in English | MEDLINE | ID: mdl-30726215

ABSTRACT

Tripartite motif (TRIM) proteins belong to a large family with many roles in host biology, including restricting virus infection. Here, we found that TRIM2, which has been implicated in cases of Charcot-Marie-Tooth disease (CMTD) in humans, acts by blocking hemorrhagic fever New World arenavirus (NWA) entry into cells. We show that Trim2-knockout mice, as well as primary fibroblasts from a CMTD patient with mutations in TRIM2, are more highly infected by the NWAs Junín and Tacaribe virus than wild-type mice or cells are. Using mice with different Trim2 gene deletions and TRIM2 mutant constructs, we demonstrate that its antiviral activity is uniquely independent of the RING domain encoding ubiquitin ligase activity. Finally, we show that one member of the TRIM2 interactome, signal regulatory protein α (SIRPA), a known inhibitor of phagocytosis, also restricts NWA infection and conversely that TRIM2 limits phagocytosis of apoptotic cells. In addition to demonstrating a novel antiviral mechanism for TRIM proteins, these studies suggest that the NWA entry and phagocytosis pathways overlap.


Subject(s)
Antigens, Differentiation/genetics , Arenaviruses, New World/genetics , Charcot-Marie-Tooth Disease/genetics , Host-Pathogen Interactions/genetics , Nuclear Proteins/genetics , Receptors, Immunologic/genetics , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Apoptosis , Arenaviruses, New World/growth & development , Arenaviruses, New World/pathogenicity , Brain/immunology , Brain/metabolism , Brain/virology , Cell Line, Tumor , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Chlorocebus aethiops , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Neurofilament Proteins/genetics , Neurofilament Proteins/immunology , Neurofilament Proteins/metabolism , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoblasts/virology , Primary Cell Culture , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Vero Cells , Virus Internalization
10.
Development ; 145(3)2018 01 31.
Article in English | MEDLINE | ID: mdl-29361554

ABSTRACT

Gli3 is a Hedgehog (Hh)-responsive transcription factor that can function as a transcriptional repressor or activator. We show that Gli3 activity in mouse thymic epithelial cells (TECs) promotes positive selection and differentiation from CD4+ CD8+ to CD4+ CD8- single-positive (SP4) cells in the fetal thymus and that Gli3 represses Shh Constitutive deletion of Gli3, and conditional deletion of Gli3 from TECs, reduced differentiation to SP4, whereas conditional deletion of Gli3 from thymocytes did not. Conditional deletion of Shh from TECs increased differentiation to SP4, and expression of Shh was upregulated in the Gli3-deficient thymus. Use of a transgenic Hh reporter showed that the Hh pathway was active in thymocytes, and increased in the Gli3-deficient fetal thymus. Neutralisation of endogenous Hh proteins in the Gli3-/- thymus restored SP4 differentiation, indicating that Gli3 in TECs promotes SP4 differentiation by repression of Shh Transcriptome analysis showed that Hh-mediated transcription was increased whereas TCR-mediated transcription was decreased in Gli3-/- thymocytes compared with wild type.


Subject(s)
Hedgehog Proteins/metabolism , Nerve Tissue Proteins/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Epithelial Cells/cytology , Female , Gene Expression Profiling , Hedgehog Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Pregnancy , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/metabolism , Zinc Finger Protein Gli3/deficiency , Zinc Finger Protein Gli3/genetics
11.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641479

ABSTRACT

Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) family members are cytidine deaminases that play important roles in intrinsic responses to retrovirus infection. Complex retroviruses like human immunodeficiency virus type 1 (HIV-1) encode the viral infectivity factor (Vif) protein to counteract APOBEC3 proteins. Vif induces degradation of APOBEC3G and other APOBEC3 proteins and thereby prevents their packaging into virions. It is not known if murine leukemia virus (MLV) encodes a Vif-like protein. Here, we show that the MLV P50 protein, produced from an alternatively spliced gag RNA, interacts with the C terminus of mouse APOBEC3 and prevents its packaging without causing its degradation. By infecting APOBEC3 knockout (KO) and wild-type (WT) mice with Friend or Moloney MLV P50-deficient viruses, we found that APOBEC3 restricts the mutant viruses more than WT viruses in vivo Replication of P50-mutant viruses in an APOBEC3-expressing stable cell line was also much slower than that of WT viruses, and overexpressing P50 in this cell line enhanced mutant virus replication. Thus, MLV encodes a protein, P50, that overcomes APOBEC3 restriction by preventing its packaging into virions.IMPORTANCE MLV has existed in mice for at least a million years, in spite of the existence of host restriction factors that block infection. Although MLV is considered a simple retrovirus compared to lentiviruses, it does encode proteins generated from alternatively spliced RNAs. Here, we show that P50, generated from an alternatively spliced RNA encoded in gag, counteracts APOBEC3 by blocking its packaging. MLV also encodes a protein, glycoGag, that increases capsid stability and limits APOBEC3 access to the reverse transcription complex (RTC). Thus, MLV has evolved multiple means of preventing APOBEC3 from blocking infection, explaining its survival as an infectious pathogen in mice.


Subject(s)
Cytidine Deaminase/genetics , Gene Expression Regulation, Viral , Gene Products, gag/genetics , Leukemia, Experimental/genetics , Moloney murine leukemia virus/genetics , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Alternative Splicing , Animals , Capsid/metabolism , Cytidine Deaminase/deficiency , Gene Products, gag/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Mice , Mice, Knockout , Moloney murine leukemia virus/metabolism , Moloney murine leukemia virus/pathogenicity , NIH 3T3 Cells , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Signal Transduction , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Virion/genetics , Virion/metabolism , Virion/pathogenicity , Virus Replication
12.
Pediatr Transplant ; 25(5): e13930, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33326675

ABSTRACT

Paediatric heart transplantation recipients suffer an increased incidence of infectious, autoimmune and allergic problems. The relative roles of thymus excision and immunosuppressive treatments in contributing to these sequelae are not clear. We compared the immunological phenotypes of 25 heart transplant recipients (Tx), 10 children who underwent thymus excision during non-transplantation cardiac surgery (TE) and 25 age range-matched controls, in two age bands: 1-9 and 10-16 years. Significant differences from controls were seen mainly in the younger age band with Tx showing lower CD3 and CD4 cell counts whilst TE showed lower CD8 cell counts. Naïve T cell and recent thymic emigrant proportions and counts were significantly lower than controls in both groups in the lower age band. T cell recombination excision circle (TREC) levels were lower than controls in both groups in both age bands. There were no differences in regulatory T cells, but in those undergoing thymus excision in infancy, their proportions were higher in TE than Tx, a possible direct effect of immunosuppression. T cell receptor V beta spectratyping showed fewer peaks in both groups than in controls (predominantly in the older age band). Thymus excision in infancy was associated with lower CD8 cell counts and higher proportions of Tregs in TE compared to Tx. These data are consistent with thymus excision, particularly in infancy, being the most important influence on immunological phenotype after heart transplantation.


Subject(s)
Heart Transplantation , Immunophenotyping , T-Lymphocytes, Regulatory/immunology , Thymus Gland/surgery , Adolescent , Antibodies, Monoclonal , Child , Child, Preschool , Female , Humans , Immune Tolerance , Immunosuppression Therapy , Infant , Lymphocyte Count , Male
13.
Immunology ; 159(4): 365-372, 2020 04.
Article in English | MEDLINE | ID: mdl-31792954

ABSTRACT

Interferon-inducible transmembrane (IFITM) proteins are a family of small homologous proteins, localized in the plasma and endolysosomal membranes, which confer cellular resistance to many viruses. In addition, several distinct functions have been associated with different IFITM family members, including germ cell specification (IFITM1-IFITM3), osteoblast function and bone mineralization (IFITM5) and immune functions (IFITM1-3, IFITM6). IFITM1-3 are expressed by T cells and recent experiments have shown that the IFITM proteins are directly involved in adaptive immunity and that they regulate CD4+ T helper cell differentiation in a T-cell-intrinsic manner. Here we review the role of the IFITM proteins in T-cell differentiation and function.


Subject(s)
Adaptive Immunity , Antigens, Differentiation/immunology , Asthma/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Virus Diseases/immunology , Animals , Antigens, Differentiation/genetics , Asthma/genetics , Asthma/pathology , Cell Differentiation , Cell Membrane/immunology , Cell Membrane/metabolism , Endosomes/immunology , Endosomes/metabolism , Gene Expression Regulation , Humans , Inflammation , Mice , Protein Isoforms/genetics , Protein Isoforms/immunology , Signal Transduction , Th1 Cells/metabolism , Th2 Cells/metabolism , Virus Diseases/genetics , Virus Diseases/pathology , Virus Diseases/virology , Viruses/immunology , Viruses/pathogenicity
14.
Neurobiol Dis ; 140: 104845, 2020 07.
Article in English | MEDLINE | ID: mdl-32205255

ABSTRACT

We analyzed Trim2A/A mice, generated by CRISPR-Cas9, which have a recessive, null mutation of Trim2. Trim2A/A mice develop ataxia that is associated with a severe loss of cerebellar Purkinje cells and a peripheral neuropathy. Myelinated axons in the CNS, including those in the deep cerebellar nuclei, have focal enlargements that contain mitochondria and neurofilaments. In the PNS, there is a loss of myelinated axons, particularly in the most distal nerves. The pathologically affected neuronal populations - primary sensory and motor neurons as well as cerebellar Purkinje cells - express TRIM2, suggesting that loss of TRIM2 in these neurons results in cell autonomous effects on their axons. In contrast, these pathological findings were not found in a second strain of Trim2 mutant mice (Trim2C/C), which has a partial deletion in the RING domain that is needed for ubiquitin ligase activity. Both the Trim2Aand the Trim2C alleles encode mutant TRIM2 proteins with reduced ubiquitination activity. In sum, Trim2A/A mice are a genetically authentic animal model of a recessive axonal neuropathy of humans, apparently for a function that does not depend on the ubiquitin ligase activity.


Subject(s)
Axons/pathology , Charcot-Marie-Tooth Disease/genetics , Mutation , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Intermediate Filaments/metabolism , Mice , Motor Neurons/pathology
15.
Eur J Immunol ; 49(1): 66-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30365177

ABSTRACT

The interferon-inducible transmembrane (Ifitm/Fragilis) genes encode homologous proteins that are induced by IFNs. Here, we show that IFITM proteins regulate murine CD4+ Th cell differentiation. Ifitm2 and Ifitm3 are expressed in wild-type (WT) CD4+ T cells. On activation, Ifitm3 was downregulated and Ifitm2 was upregulated. Resting Ifitm-family-deficient CD4+ T cells had higher expression of Th1-associated genes than WT and purified naive Ifitm-family-deficient CD4+ T cells differentiated more efficiently to Th1, whereas Th2 differentiation was inhibited. Ifitm-family-deficient mice, but not Ifitm3-deficient mice, were less susceptible than WT to induction of allergic airways disease, with a weaker Th2 response and less severe disease and lower Il4 but higher Ifng expression and IL-27 secretion. Thus, the Ifitm family is important in adaptive immunity, influencing Th1/Th2 polarization, and Th2 immunopathology.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Membrane Proteins/metabolism , Respiratory System/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-27/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1-Th2 Balance/genetics
16.
J Virol ; 93(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31341050

ABSTRACT

Endogenous retroviruses (ERV) are found throughout vertebrate genomes, and failure to silence their activation can have deleterious consequences on the host. Mutation and subsequent disruption of ERV loci is therefore an indispensable component of the cell-intrinsic defenses that maintain the integrity of the host genome. Abundant in vitro and in silico evidence have revealed that APOBEC3 cytidine-deaminases, including human APOBEC3G (hA3G), can potently restrict retrotransposition; yet, in vivo data demonstrating such activity is lacking, since no replication-competent human ERV have been identified. In mice deficient for Toll-like receptor 7 (TLR7), transcribed ERV loci can recombine and generate infectious ERV. In this study, we show that ectopic expression of hA3G can prevent the emergence of replication-competent, infectious ERV in Tlr7-/- mice. Mice encode one copy of Apobec3 in their genome. ERV reactivation in Tlr7-/- mice was comparable in the presence or absence of Apobec3 In contrast, expression of a human APOBEC3G transgene abrogated emergence of infectious ERV in the Tlr7-/- background. No ERV RNA was detected in the plasma of hA3G+Apobec3-/-Tlr7-/- mice, and infectious ERV virions could not be amplified through coculture with permissive cells. These data reveal that hA3G can potently restrict active ERV in vivo and suggest that expansion of the APOBEC3 locus in primates may have helped to provide for the continued restraint of ERV in the human genome.IMPORTANCE Although APOBEC3 proteins are known to be important antiviral restriction factors in both mice and humans, their roles in the restriction of endogenous retroviruses (ERV) have been limited to in vitro studies. Here, we report that human APOBEC3G expressed as a transgene in mice prevents the emergence of infectious ERV from endogenous loci. This study reveals that APOBEC3G can powerfully restrict active retrotransposons in vivo and demonstrates how transgenic mice can be used to investigate host mechanisms that inhibit retrotransposons and reinforce genomic integrity.


Subject(s)
APOBEC-3G Deaminase/metabolism , Endogenous Retroviruses/physiology , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Virus Replication , Animals , Gene Dosage , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Mice , Mice, Knockout , Open Reading Frames , Retroviridae Infections/immunology , Toll-Like Receptors/metabolism
17.
Eur J Immunol ; 48(4): 716-719, 2018 04.
Article in English | MEDLINE | ID: mdl-29318612

ABSTRACT

Nude mouse human thymus transplant model: Fresh or cryopreserved and thawed human thymus slices were transplanted subcutaneously into recipient nude mice. Nude mice subsequently produced mouse CD3+ CD4+ T-cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/transplantation , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cryopreservation , Humans , Lymphocyte Count , Mice , Mice, Nude , Transplantation, Heterologous
18.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593034

ABSTRACT

The apolipoprotein B editing complex 3 (APOBEC3) proteins are potent retroviral restriction factors that are under strong positive selection, both in terms of gene copy number and sequence diversity. A common feature of all the members of the APOBEC3 family is the presence of one or two cytidine deamination domains, essential for cytidine deamination of retroviral reverse transcripts as well as packaging into virions. Several studies have indicated that human and mouse APOBEC3 proteins restrict retrovirus infection via cytidine deaminase (CD)-dependent and -independent means. To understand the relative contribution of CD-independent restriction in vivo, we created strains of transgenic mice on an APOBEC3 knockout background that express a deaminase-dead mouse APOBEC3 due to point mutations in both CD domains (E73Q/E253Q). Here, we show that the CD-dead APOBEC3 can restrict murine retroviruses in vivo Moreover, unlike the wild-type protein, the mutant APOBEC3 is not packaged into virions but acts only as a cell-intrinsic restriction factor that blocks reverse transcription by incoming viruses. Finally, we show that wild-type and CD-dead mouse APOBEC3 can bind to murine leukemia virus (MLV) reverse transcriptase. Our findings suggest that the mouse APOBEC3 cytidine deaminase activity is not required for retrovirus restriction.IMPORTANCE APOBEC3 proteins are important host cellular restriction factors essential for restricting retrovirus infection by causing mutations in the virus genome and by blocking reverse transcription. While both methods of restriction function in vitro, little is known about their role during in vivo infection. By developing transgenic mice with mutations in the cytidine deamination domains needed for enzymatic activity and interaction with viral RNA, we show that APOBEC3 proteins can still restrict in vivo infection by interacting with reverse transcriptase and blocking its activity. These studies demonstrate that APOBEC3 proteins have evolved multiple means for blocking retrovirus infection and that all of these means function in vivo.


Subject(s)
Cytidine Deaminase/genetics , Leukemia Virus, Murine/genetics , Retroviridae Infections/prevention & control , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcription/genetics , Animals , Cell Line , Cytidine Deaminase/metabolism , Deamination/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Viral/genetics , RNA-Directed DNA Polymerase/genetics
19.
N Engl J Med ; 372(5): 407-17, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25629739

ABSTRACT

BACKGROUND: The effects of less-tight versus tight control of hypertension on pregnancy complications are unclear. METHODS: We performed an open, international, multicenter trial involving women at 14 weeks 0 days to 33 weeks 6 days of gestation who had nonproteinuric preexisting or gestational hypertension, office diastolic blood pressure of 90 to 105 mm Hg (or 85 to 105 mm Hg if the woman was taking antihypertensive medications), and a live fetus. Women were randomly assigned to less-tight control (target diastolic blood pressure, 100 mm Hg) or tight control (target diastolic blood pressure, 85 mm Hg). The composite primary outcome was pregnancy loss or high-level neonatal care for more than 48 hours during the first 28 postnatal days. The secondary outcome was serious maternal complications occurring up to 6 weeks post partum or until hospital discharge, whichever was later. RESULTS: Included in the analysis were 987 women; 74.6% had preexisting hypertension. The primary-outcome rates were similar among 493 women assigned to less-tight control and 488 women assigned to tight control (31.4% and 30.7%, respectively; adjusted odds ratio, 1.02; 95% confidence interval [CI], 0.77 to 1.35), as were the rates of serious maternal complications (3.7% and 2.0%, respectively; adjusted odds ratio, 1.74; 95% CI, 0.79 to 3.84), despite a mean diastolic blood pressure that was higher in the less-tight-control group by 4.6 mm Hg (95% CI, 3.7 to 5.4). Severe hypertension (≥160/110 mm Hg) developed in 40.6% of the women in the less-tight-control group and 27.5% of the women in the tight-control group (P<0.001). CONCLUSIONS: We found no significant between-group differences in the risk of pregnancy loss, high-level neonatal care, or overall maternal complications, although less-tight control was associated with a significantly higher frequency of severe maternal hypertension. (Funded by the Canadian Institutes of Health Research; CHIPS Current Controlled Trials number, ISRCTN71416914; ClinicalTrials.gov number, NCT01192412.).


Subject(s)
Antihypertensive Agents/administration & dosage , Hypertension, Pregnancy-Induced/drug therapy , Pregnancy Complications/etiology , Pregnancy Outcome , Abortion, Spontaneous/etiology , Adult , Blood Pressure/drug effects , Female , Humans , Infant, Newborn , Intensive Care, Neonatal/statistics & numerical data , Perinatal Death/etiology , Pregnancy , Pregnancy Complications/epidemiology , Puerperal Disorders/etiology
SELECTION OF CITATIONS
SEARCH DETAIL