Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.433
Filter
1.
J Med Virol ; 96(10): e29899, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39370775

ABSTRACT

BRAAVE (NCT03631732), a Phase 3b, multicenter, open-label US study, demonstrated the efficacy of switching to bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF) among Black individuals with suppressed HIV through 48 weeks. Here, 72-week resistance, adherence, and virologic outcomes are presented. Enrollment criteria permitted nonnucleoside reverse transcriptase inhibitor (NNRTI)-resistance (R), protease inhibitor (PI)-R, and certain nucleos(t)ide reverse transcriptase inhibitor (NRTI)-R (M184V/I allowed; ≥3 thymidine analog mutations [TAMs] excluded); but excluded primary integrase strand transfer inhibitor (INSTI)-R. Pre-existing resistance was determined using historical genotypes and retrospective baseline proviral DNA genotyping. Adherence, virologic outcomes, and viral blips were assessed. Of 489 participants receiving B/F/TAF with ≥1 post-switch HIV-1 RNA measurement: pre-existing NRTI-R (15% of participants), M184V/I (11%), ≥1 TAMs (8%), NNRTI-R (22%), and PI-R (13%) were observed; pre-existing INSTI-R substitutions (2%) were detected post-randomization; mean viral blip frequency was 0.9% across all timepoints (unassociated with virologic failure); 24% of participants had <95% adherence (98% of whom had HIV-1 RNA <50 copies/mL at last visit); none had treatment-emergent study-drug resistance. Overall, 99% of participants, including all with baseline NRTI-R/INSTI-R, had HIV-1 RNA <50 copies/mL at the last visit, demonstrating that B/F/TAF maintained virologic suppression through 72 weeks regardless of pre-existing resistance, viral blips, and suboptimal adherence.


Subject(s)
Anti-HIV Agents , Drug Resistance, Viral , Emtricitabine , HIV Infections , HIV-1 , Heterocyclic Compounds, 4 or More Rings , Medication Adherence , Piperazines , Pyridones , Tenofovir , Humans , HIV Infections/drug therapy , HIV Infections/virology , Tenofovir/therapeutic use , Tenofovir/analogs & derivatives , Male , Female , Emtricitabine/therapeutic use , Drug Resistance, Viral/genetics , Anti-HIV Agents/therapeutic use , Adult , United States , Pyridones/therapeutic use , Middle Aged , Piperazines/therapeutic use , Heterocyclic Compounds, 4 or More Rings/therapeutic use , HIV-1/drug effects , HIV-1/genetics , Medication Adherence/statistics & numerical data , Black or African American , Drug Combinations , Heterocyclic Compounds, 3-Ring/therapeutic use , Adenine/analogs & derivatives , Adenine/therapeutic use , Adenine/pharmacology , Amides/therapeutic use , Treatment Outcome , Alanine/therapeutic use , Viral Load/drug effects
2.
Cells ; 13(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39272980

ABSTRACT

Direct neuronal reprogramming is a promising approach to replace neurons lost due to disease via the conversion of endogenous glia reacting to brain injury into neurons. However, it is essential to demonstrate that the newly generated neurons originate from glial cells and/or show that they are not pre-existing endogenous neurons. Here, we use controls for both requirements while comparing two viral vector systems (Mo-MLVs and AAVs) for the expression of the same neurogenic factor, the phosphorylation-resistant form of Neurogenin2. Our results show that Mo-MLVs targeting proliferating glial cells after traumatic brain injury reliably convert astrocytes into neurons, as assessed by genetic fate mapping of astrocytes. Conversely, expressing the same neurogenic factor in a flexed AAV system results in artefactual labelling of endogenous neurons fatemapped by birthdating in development that are negative for the genetic fate mapping marker induced in astrocytes. These results are further corroborated by chronic live in vivo imaging. Taken together, the phosphorylation-resistant form of Neurogenin2 is more efficient in reprogramming reactive glia into neurons than its wildtype counterpart in vivo using retroviral vectors (Mo-MLVs) targeting proliferating glia. Conversely, AAV-mediated expression generates artefacts and is not sufficient to achieve fate conversion.


Subject(s)
Astrocytes , Cellular Reprogramming , Cerebral Cortex , Dependovirus , Genetic Vectors , Neurons , Animals , Astrocytes/metabolism , Neurons/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dependovirus/genetics , Cellular Reprogramming/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice, Inbred C57BL , Male , Retroviridae/genetics
3.
J Genet Genomics ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39265822

ABSTRACT

Human endogenous retroviruses (HERVs) comprise approximately 8% of the human genome, co-opted into the dynamic regulatory network of cellular potency in early embryonic development. In recent studies, resurgent HERVs' transcriptional activity has been frequently observed in many types of human cancers, suggesting their potential functions in the occurrence and progression of malignancy. However, a dedicated web resource for querying the relationship between activation of HERVs and cancer development is lacking. Here, we have constructed a database to explore the sequence information, expression profiles, survival prognosis, and genetic interactions of HERVs in diverse cancer types. Our database currently contains RNA sequencing data of 580 HERVs across 16246 samples, including that of 6478 tumoral and 634 normal tissues, 932 cancer cell lines, as well as 151 early embryonic and 8051 human adult tissues. The primary goal is to provide an easily accessible and user-friendly database for professionals in the fields of bioinformatics, pathology, pharmacology, and related areas, enabling them to efficiently screen the activity of HERVs of interest in normal and cancerous tissues and evaluate the clinical relevance. The ERVcancer database is available at http://kyuanlab.com/ervcancer/.

4.
Biomolecules ; 14(9)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39334897

ABSTRACT

Introducing genetic material into hard-to-transfect mammalian cell lines and primary cells is often best achieved through retroviral infection. An ideal retroviral vector should offer a compact, selectable, and screenable marker while maximizing transgene delivery capacity. However, a previously published retroviral vector featuring an EGFP/Puromycin fusion protein failed to meet these criteria in our experiments. We encountered issues such as low infection efficiency, weak EGFP fluorescence, and selection against infected cells. To address these shortcomings, we developed a novel retroviral vector based on the Moloney murine leukemia virus. This vector includes a compact bifunctional EGFP and Puromycin resistance cassette connected by a 2A peptide. Our extensively tested vector demonstrated superior EGFP expression, efficient Puromycin selection, and no growth penalty in infected cells compared with the earlier design. These benefits were consistent across multiple mammalian cell types, underscoring the versatility of our vector. In summary, our enhanced retroviral vector offers a robust solution for efficient infection, reliable detection, and effective selection in mammalian cells. Its improved performance and compact design make it an ideal choice for a wide range of applications involving precise genetic manipulation and characterization in cell-based studies.


Subject(s)
Genetic Vectors , Green Fluorescent Proteins , Moloney murine leukemia virus , Retroviridae , Genetic Vectors/genetics , Humans , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Moloney murine leukemia virus/genetics , Retroviridae/genetics , Animals , Puromycin/pharmacology , HEK293 Cells , Transgenes , Mice
5.
Microorganisms ; 12(9)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39338563

ABSTRACT

Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that, over millions of years, became integrated into the human genome. While normally inactive, environmental stimuli such as infections have contributed to the transcriptional reactivation of HERV-promoting pathological conditions, including the development of autoimmunity, neurodegenerative disease and cancer. What infections trigger HERV activation? Mycobacterium avium subspecies paratuberculosis (MAP) is a pluripotent driver of human disease. Aside from granulomatous diseases, Crohn's disease, sarcoidosis and Blau syndrome, MAP is associated with autoimmune disease: type one diabetes (T1D), multiple sclerosis (MS), rheumatoid arthritis (RA) and autoimmune thyroiditis. MAP is also associated with Alzheimer's disease (AD) and Parkinson's disease (PD). Autoimmune diabetes, MS and RA are the diseases with the strongest MAP/HERV association. There are several other diseases associated with HERV activation, including diseases whose epidemiology and/or pathology would prompt speculation for a causal role of MAP. These include non-solar uveal melanoma, colon cancer, glioblastoma and amyotrophic lateral sclerosis (ALS). This article further points to MAP infection as a contributor to autoimmunity, neurodegenerative disease and cancer via the un-silencing of HERV. We examine the link between the ever-increasing number of MAP-associated diseases and the MAP/HERV intersection with these diverse medical conditions, and propose treatment opportunities based upon this association.

6.
Proc Natl Acad Sci U S A ; 121(40): e2407990121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39320912

ABSTRACT

We explored how a simple retrovirus, Mason-Pfizer monkey virus (M-PMV) to facilitate its replication process, utilizes DHX15, a cellular RNA helicase, typically engaged in RNA processing. Through advanced genetic engineering techniques, we showed that M-PMV recruits DHX15 by mimicking cellular mechanisms, relocating it from the nucleus to the cytoplasm to aid in viral assembly. This interaction is essential for the correct packaging of the viral genome and critical for its infectivity. Our findings offer unique insights into the mechanisms of viral manipulation of host cellular processes, highlighting a sophisticated strategy that viruses employ to leverage cellular machinery for their replication. This study adds valuable knowledge to the understanding of viral-host interactions but also suggests a common evolutionary history between cellular processes and viral mechanisms. This finding opens a unique perspective on the export mechanism of intron-retaining mRNAs in the packaging of viral genetic information and potentially develop ways to stop it.


Subject(s)
Mason-Pfizer monkey virus , RNA, Viral , Virus Assembly , Animals , Humans , Cell Nucleus/metabolism , Cell Nucleus/virology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Genome, Viral , HEK293 Cells , Mason-Pfizer monkey virus/genetics , Mason-Pfizer monkey virus/metabolism , Mason-Pfizer monkey virus/physiology , RNA Helicases/metabolism , RNA Helicases/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , Virus Assembly/genetics , Virus Assembly/physiology , Virus Replication/genetics , Virus Replication/physiology
7.
J Virol ; : e0091524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39287391

ABSTRACT

Syncytins are envelope genes of retroviral origin that play a critical role in the formation of a syncytial structure at the fetomaternal interface via their fusogenic activity. The mouse placenta is unique among placental mammals since the fetomaternal interface comprises two syncytiotrophoblast layers (ST-I and ST-II) instead of one observed in all other hemochorial placentae. Each layer specifically expresses a distinct mouse syncytin, namely syncytin-A (SynA) for ST-I and syncytin-B (SynB) for ST-II, which have been shown to be essential to placentogenesis and embryonic development. The cellular receptor for SynA has been identified as the membrane protein LY6E and is not the receptor for SynB. Here, by combining a cell-cell fusion assay with the screening of a human ORFeome-derived expression library, we identified the transmembrane multipass sodium-dependent phosphate transporter 1 PiT1/SLC20A1 as the receptor for SynB. Transfection of cells with the cloned receptor, but not the closely related PiT2/SLC20A2, leads to their fusion with cells expressing SynB, with no cross-reactive fusion activity with SynA. The interaction between the two partners was further demonstrated by immunoprecipitation. PiT1/PiT2 chimera and truncation experiments identified the PiT1 N-terminus as the major determinant for SynB-mediated fusion. RT-qPCR analysis of PiT1 expression on a panel of mouse adult and fetal tissues revealed a concomitant increase of PiT1 and SynB specifically in the developing placenta. Finally, electron microscopy analysis of the placenta of PiT1 null embryo before they die (E11.5) disclosed default of ST-II formation with lack of syncytialization, as previously observed in cognate SynB null placenta, and consistent with the present identification of PiT1 as the SynB partner.IMPORTANCESyncytins are envelope genes of endogenous retroviruses, coopted for a physiological function in placentation. They are fusogenic proteins that mediate cell-cell fusion by interacting with receptors present on the partner cells. Here, by devising an in vitro fusion assay that enables the screening of an ORFeome-derived expression library, we identified the long-sought receptor for syncytin-B (SynB), a mouse syncytin responsible for syncytiotrophoblast formation at the fetomaternal interface of the mouse placenta. This protein - PiT1/SLC20A1 - is a multipass transmembrane protein, also known as the receptor for a series of infectious retroviruses. Its profile of expression is consistent with a role in both ancestral endogenization of a SynB founder retrovirus and present-day mouse placenta formation, with evidence-in PiT1 knockout mice-of unfused cells at the level of the cognate placental syncytiotrophoblast layer.

8.
Virology ; 600: 110247, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39307098

ABSTRACT

We have shown the induction of CD11b+Ly6C+ monocytic myeloid-derived suppressor cells (M-MDSCs) during infection of B6 mice by LP-BM5 immunodeficiency-inducing retrovirus. We published that the molecular mechanisms of these M-MDSCs vary, and depend on the cell type targeted by the suppression -defined by use of biochemical inhibitors, mouse M-MDSCs knock-out strains and blocking antibodies. These M-MDSCs suppressed proliferation and function of T cells, via nitric oxide synthase/nitric oxide; and that of B cells, ∼50% via INOS/NO along with the negative checkpoint regulator VISTA, reactive nitrogen and oxygen species, and other soluble mediators. Here, LP-BM5 infected mice were treated weekly with 5-Fluorouracil (5-FU), resulting in depletion of peripheral blood and splenic M-MDSCs, reduced MDSC activity, and significantly decreased standard disease parameters of: splenomegaly, impaired B-and T-cell ex vivo polyclonal responses, and viral load. In addition, 5-FU treatment significantly increased percentages of CD4+ and CD8+ T cells.

9.
Heliyon ; 10(17): e36955, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39281539

ABSTRACT

Chimeric antigen receptor T (CAR-T)-cell therapy targeting B-cell maturation antigen (BCMA) is currently one of the promising treatment methods for relapsed/refractory multiple myeloma (MM). Herein, this study is a case report on a 41-year-old male patient with MM. Unfortunately, he still developed multidrug-resistant, refractory, and bone marrow suppression after receiving multiline high-intensity chemotherapy. After a detailed evaluation, the physician recommended autologous hematopoietic stem cell transplantation (ASCT) support, followed by sequential immunotherapy with autologous anti- BCMA CAR-T cells. The CAR-T product is a novel anti-BCMA CAR-T based on Retrovirus vectors (RV). It was worth noting that the patient achieved VGPR (very good partial remission) one month after infusion of anti-BCMA CAR-T cells. Recent tests have found that the M protein was no longer detectable and the patient has achieved CR (complete response). Although grade 3 cytokine release syndrome (CRS) appeared, the symptom was well controlled and immune effector cell-associated neurotoxicity syndrome (ICANS) did not occur. This was the first case report of RV prepared anti-BCMA CAR-T cells combined with ASCT for the treatment of MM patient in clinical practice, indicating that the RV-based anti-BCMA-CAR-T cells with ASCT have excellent therapeutic efficacy and high safety in triple-refractory MM patients.

10.
Regen Ther ; 26: 729-740, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39290630

ABSTRACT

Introduction: In human placental development, the trophectoderm (TE) appears in blastocysts on day 5 post-fertilization and develops after implantation into three types of trophoblast lineages: cytotrophoblast (CT), syncytiotrophoblast (ST), and extravillous trophoblast (EVT). CDX2/Cdx2 is expressed in the TE, and Cdx2 expression is upregulated by knockdown of Foxo1 in mouse ESCs. However, the significance of FOXO1 in trophoblast lineage differentiation during the early developmental period remains unclear. In this study, we examined the effect of FOXO1 inhibition on the differentiation of naive human induced pluripotent stem cells (iPSCs) into TE and trophoblast lineages. Methods: We induced TE differentiation from naive iPSCs in the presence or absence of a FOXO1 inhibitor, and the resulting cells were subjected to trophoblast differentiation procedures without the FOXO1 inhibitor. The cells obtained in these processes were assessed for morphology, gene expression, and hCG secretion using phase-contrast microscopy, reverse transcription polymerase chain reaction (RT-PCR), quantitative RT-PCR (RT-qPCR), RNA-seq, immunochromatography, and a chemiluminescent enzyme immunoassay. Results: In the induction of trophoblast differentiation from naive iPSCs, treatment with a FOXO1 inhibitor resulted in the enhanced expression of TE markers, CDX2 and HAND1, but conversely decreased the expression of ST markers, such as ERVW1 (Syncytin-1) and GCM1, and an EVT marker, HLA-G. The proportion of cells positive for an early TE marker TACSTD2 and negative for a late TE marker ENPEP was higher in FOXO1 inhibitor-treated cells than in non-treated cells. The expressions of ERVW1 (Syncytin-1), ERVFRD-1 (Syncytin-2), and other endogenous retrovirus (ERV)-associated genes that have been reported to be expressed in trophoblasts were suppressed in the cells obtained by differentiating the TE cells treated with FOXO1 inhibitor. Conclusions: Treatment with a FOXO1 inhibitor during TE induction from naive iPSCs promotes early TE differentiation but hinders the progression of differentiation into ST and EVT. The suppression of ERV-associated genes may be involved in this process.

11.
Viruses ; 16(8)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39205286

ABSTRACT

Endogenous retroviruses (ERVs) are the remnants of retroviral germline infections and are highly abundant in the genomes of vertebrates. At one time considered to be nothing more than inert 'junk' within genomes, ERVs have been tolerated within host genomes over vast timescales, and their study continues to reveal complex co-evolutionary histories within their respective host species. For example, multiple instances have been characterized of ERVs having been 'borrowed' for normal physiology, from single copies to ones involved in various regulatory networks such as innate immunity and during early development. Within the cell, the accessibility of ERVs is normally tightly controlled by epigenetic mechanisms such as DNA methylation or histone modifications. However, these silencing mechanisms of ERVs are reversible, and epigenetic alterations to the chromatin landscape can thus lead to their aberrant expression, as is observed in abnormal cellular environments such as in tumors. In this review, we focus on ERV transcriptional control and draw parallels and distinctions concerning the loss of regulation in disease, as well as their precise regulation in early development.


Subject(s)
Endogenous Retroviruses , Epigenesis, Genetic , Endogenous Retroviruses/genetics , Humans , Animals , DNA Methylation , Gene Expression Regulation, Viral , Transcription, Genetic , Viral Transcription/genetics , Retroviridae Infections/virology
12.
Comp Immunol Microbiol Infect Dis ; 112: 102228, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39151280

ABSTRACT

The objective of this study was to categorise diseases associated with FeLV infection in cats. A total of 154 cats were submitted to necropsy, histopathology exam and anti-FeLV immunohistochemistry (IHC), and 83 (50.9 %) were IHC FeLV-positive. The cats age means of 4.1 years, including 3.6 % kittens, 34.9 % junior, 37.4 % prime, 18.1 % mature, 2.4 % senior, 3.6 % unknown age. Neoplastic diseases were most prevalent with leukaemia and lymphoma being most predominant, followed by viral diseases, bacterial, trauma, degenerative, intoxications, parasitic, malformation and others. FeLV+ cats were 5.73 times more likely to be diagnosed with neoplasms than other diseases. The odds ratio (OR) of FeLV+ cats developing leukaemia (OR = 7.75) and lymphoma (OR = 6.75) was higher than other neoplasms. FeLV infection was more prevalent in the mixed breed, junior to prime, male, with neoplastic diseases, including leukaemia and lymphoma. Therefore, understanding the diseases associated with FeLV is of paramount importance in Brazil due to its high prevalence, and it may encourage the implementation of prophylactic measures to reduce its dissemination.


Subject(s)
Cat Diseases , Leukemia Virus, Feline , Leukemia, Feline , Cats , Animals , Leukemia Virus, Feline/isolation & purification , Brazil/epidemiology , Male , Cat Diseases/virology , Cat Diseases/epidemiology , Female , Prevalence , Leukemia, Feline/epidemiology , Leukemia, Feline/virology , Lymphoma/epidemiology , Lymphoma/veterinary , Lymphoma/virology , Retroviridae Infections/epidemiology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Immunohistochemistry , Neoplasms/epidemiology , Neoplasms/veterinary , Neoplasms/virology , Tumor Virus Infections/veterinary , Tumor Virus Infections/epidemiology , Tumor Virus Infections/virology
13.
Immunol Rev ; 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39152687

ABSTRACT

The human genome harbors hundreds of thousands of integrations of ancient retroviruses, amassed over millions of years of evolution. To reduce further amplification in the genome, the host prevents transcription of these now endogenous retroviruses (ERVs) through epigenetic repression and, with evolutionary time, ERVs are incapacitated by accumulating mutations and deletions. However, several members of recently endogenized ERV groups still retain the capacity to produce viral RNA, retroviral proteins, and higher order structures, including virions. The retention of viral characteristics, combined with the reversible nature of epigenetic repression, particularly as seen in cancer, allow for immunologically unanticipated ERV expression, perceived by the adaptive immune system as a genuine retroviral infection, to which it has to respond. Accordingly, antibodies reactive with ERV antigens have been detected in diverse disorders and, occasionally, in healthy individuals. Although they are part of self, the retroviral legacy of ERV antigens, and association with and, possibly, causation of disease states may set them apart from typical self-antigens. Consequently, the pathogenic or, indeed, host-protective capacity of antibodies targeting ERV antigens is likely to be context-dependent. Here, we review the immunogenicity of typical ERV proteins, with emphasis on the antibody response and its potential disease implications.

14.
Biochem Biophys Res Commun ; 738: 150521, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39153451

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections in vertebrate genomes and are inherited by offspring. ERVs can produce pathogenic viruses through gene mutations or recombination. ERVs in domestic cats (ERV-DCs) generate feline leukemia virus subgroup D (FeLV-D) through viral recombination. Herein, we characterized the locus ERV-DC8, on chromosome B1, as an infectious replication-competent provirus. ERV-DC8 infected several cell lines, including human cells. Transmission electron microscopy of ERV-DC8 identified the viral release as a Gammaretrovirus. ERV-DC8 was identified as the FeLV-D viral interference group, with feline copper transporter 1 as its viral receptor. Insertional polymorphism analysis showed high ERV-DC8 integration in domestic cats. This study highlights the role, pathogenicity, and evolutionary relationships between ERVs and their hosts.

15.
Hum Gene Ther ; 35(17-18): 604-616, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150015

ABSTRACT

The invention of next-generation CRISPR/Cas gene editing tools, like base and prime editing, for correction of gene variants causing disease, has created hope for in vivo use in patients leading to wider clinical translation. To realize this potential, delivery vehicles that can ferry gene editing tool kits safely and effectively into specific cell populations or tissues are in great demand. In this review, we describe the development of enveloped retrovirus-derived particles as carriers of "ready-to-work" ribonucleoprotein complexes consisting of Cas9-derived editor proteins and single guide RNAs. We present arguments for adapting viruses for cell-targeted protein delivery and describe the status after a decade-long development period, which has already shown effective editing in primary cells, including T cells and hematopoietic stem cells, and in tissues targeted in vivo, including mouse retina, liver, and brain. Emerging evidence has demonstrated that engineered virus-derived nanoparticles can accommodate both base and prime editors and seems to fertilize a sprouting hope that such particles can be further developed and produced in large scale for therapeutic applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Ribonucleoproteins , Gene Editing/methods , Humans , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Animals , Virion/metabolism , Virion/genetics , Retroviridae/genetics , Genetic Vectors/genetics , Genetic Therapy/methods , Gene Transfer Techniques , RNA, Guide, CRISPR-Cas Systems/genetics
16.
J Comp Pathol ; 214: 12-18, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191090

ABSTRACT

Feline leukemia virus (FeLV) is a highly debilitating cat pathogen due to its ability to cause many pathological changes. Therefore, identifying the virus directly in bone marrow can be a highly relevant diagnostic tool even in the absence of viraemia. The aim of this study was to compare the diagnostic efficiency of immunocytochemistry (ICC) of bone marrow aspirates with enzyme-linked immunosorbent assay (ELISA) and polymerase chain reaction (PCR). Blood samples were collected from 188 cats and separated into aliquots of whole blood for nested PCR using the U3 LTR region and the gag gene of FeLV-A as reference and serum for detection of the p27 antigen by ELISA. Bone marrow samples from these cats were placed on silanized slides for anti-FeLV ICC using gp70 as primary antibody. A total of 28.2% of the cats tested for FeLV were positive in at least one of the tests, with 26.6% positive by PCR, 18.1% by ICC and 11.2% by ELISA. Cohen's kappa agreement test revealed moderate agreement between ELISA and PCR results and substantial agreement between ICC and ELISA and between ICC and PCR. The results indicated that ICC of bone marrow is an efficient novel diagnostic test for FeLV infection.


Subject(s)
Bone Marrow , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Leukemia Virus, Feline , Cats , Animals , Enzyme-Linked Immunosorbent Assay/veterinary , Bone Marrow/virology , Leukemia, Feline/diagnosis , Retroviridae Infections/veterinary , Retroviridae Infections/diagnosis , Polymerase Chain Reaction/veterinary , Tumor Virus Infections/veterinary , Tumor Virus Infections/diagnosis , Cat Diseases/diagnosis , Cat Diseases/virology
17.
Prev Vet Med ; 231: 106315, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146687

ABSTRACT

The feline immunodeficiency virus (FIV) is a retrovirus of the Lentivirus genus, distributed worldwide, that causes persistent infection with a significant impact on the cats' health. Due to the importance of this infection in feline medicine, this pioneering study aimed to obtain an integrated estimate of the global seroprevalence of FIV in cats and to characterize the factors associated with this infection. Four electronic databases were screened for observational studies with FIV seroprevalence in cats published globally for this systematic review and meta-analysis. The initial search method returned 873 studies, of which 113 met all predefined criteria and were therefore included in this review. Meta-analysis with general data was performed, and a combined global seropositivity of 9.43 % (95 % CI: 8.24 % - 10.78 %) was found. Seropositivity was 14.34 % (95 % CI = 10.92 % - 18.61 %) in Asia, 11.90 % (95 % CI = 9.82 % - 14.34 %) in Oceania, 10.90 % (95 % CI = 5.71 % - 19.82 %) in Central America, 9.43 % (95 % CI = 6.95 % - 12.66 %) in South America, 9 % (95 % CI = 0 - 80 %) in Africa, 8.98 % (95 % CI = 7.31 % - 10.98 %) in Europe, and 5.93 % (95 % CI = 4.33 % - 8.07 %) in North America. Meta-analysis of factors associated with seropositivity demonstrated that FIV seroprevalence was higher in male (Prevalence ratio [PR] = 2.53, 95 % CI = 2.16 - 2.95), adult (PR = 2.83, 95 % CI = 2.24 - 3.56), unowned status (PR = 1.47, 95 % CI = 1.07 - 2.03), sick status (PR = 2.46, 95 % CI = 1.97 - 3.06), and cats with outdoor access (PR = 4.38, 95 % CI = 2.26 - 8.47). The results demonstrated that FIV is globally distributed and has a high seroprevalence in some geographical areas. Information compiled from this research is relevant to understanding the worldwide epidemiology of FIV. It presents the potential to contribute to the planning of strategies focused on controlling and reducing cases in cat populations.


Subject(s)
Feline Acquired Immunodeficiency Syndrome , Immunodeficiency Virus, Feline , Animals , Cats , Feline Acquired Immunodeficiency Syndrome/epidemiology , Feline Acquired Immunodeficiency Syndrome/virology , Global Health/statistics & numerical data , Immunodeficiency Virus, Feline/isolation & purification , Risk Factors , Seroepidemiologic Studies
18.
Virus Evol ; 10(1): veae061, 2024.
Article in English | MEDLINE | ID: mdl-39175839

ABSTRACT

The enigmatic origins and transmission events of the gibbon ape leukemia virus (GALV) and its close relative the koala retrovirus (KoRV) have been a source of enduring debate. Bats and rodents are each proposed as major reservoirs of interspecies transmission, with ongoing efforts to identify additional animal hosts of GALV-KoRV-related retroviruses. In this study, we identified nine rodent species as novel hosts of GALV-KoRV-related retroviruses. Included among these hosts are two African rodents, revealing the first appearance of this clade beyond the Australian and Southeast Asian region. One of these African rodents, Mastomys natalensis, carries an endogenous GALV-KoRV-related retrovirus that is fully intact and potentially still infectious. Our findings support the hypothesis that rodents are the major carriers of GALV-KoRV-related retroviruses.

19.
Mol Biol Evol ; 41(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39101626

ABSTRACT

Retroviruses are an ancient viral family that have globally coevolved with vertebrates and impacted their evolution. In Australia, a continent that has been geographically isolated for millions of years, little is known about retroviruses in wildlife, despite the devastating impacts of a retrovirus on endangered koala populations. We therefore sought to identify and characterize Australian retroviruses through reconstruction of endogenous retroviruses from marsupial genomes, in particular the Tasmanian devil due to its high cancer incidence. We screened 19 marsupial genomes and identified over 80,000 endogenous retrovirus fragments which we classified into eight retrovirus clades. The retroviruses were similar to either Betaretrovirus (5/8) or Gammaretrovirus (3/8) retroviruses, but formed distinct phylogenetic clades compared to extant retroviruses. One of the clades (MEBrv 3) lost an envelope but retained retrotranspositional activity, subsequently amplifying throughout all Dasyuridae genomes. Overall, we provide insights into Australian retrovirus evolution and identify a highly active endogenous retrovirus within Dasyuridae genomes.


Subject(s)
Endogenous Retroviruses , Genome , Marsupialia , Phylogeny , Animals , Endogenous Retroviruses/genetics , Marsupialia/virology , Australia , Evolution, Molecular
20.
Sci Rep ; 14(1): 20236, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215087

ABSTRACT

Systemic lupus erythematosus (SLE) is a multifactorial disease characterized by the convergence of genetic, immunological, and viral elements resulting in a complex interaction of both internal and external factors. The role of the Epstein-Barr virus (EBV) and human endogenous retroviruses (HERV-E) as triggers and maintenance elements in the pathogenesis of SLE has been widely recognized. Previous studies have independently evaluated the effects of EBV and HERV-E in this disease. In this work, for the first time, these viral factors are jointly investigated in SLE patients. This study aimed at assessing the differential expression of immune regulatory genes and the incidence of specific viral pathogens (EBV and HERV-E), alongside the detailed characterization of surface markers in T- and B-lymphocytes in patients with SLE and control participants. A comparative analysis between patients with SLE and control participants was performed, evaluating the expression of phenotypic markers and genes involved in the immune response (TNF-α, IL-2, IL-6, IL-10, IFNG, TLR3), as well as HERV-E gag and EBV viral genes (LMP1 and BZLF1).A significant association between SLE and EBV was found in this study. A notable increase in EBV LMP1 gene expression was observed in patients with SLE . Also, a significant overexpression of HERV-E was observed, in addition to a considerable increase in the distribution of the cell surface marker CD27 + on T- and B-lymphocytes, observed in individuals with SLE compared to the control group. This study provides evidence regarding the role that EBV virus plays in lymphocytes in the context of SLE, highlighting how both the virus and the host gene expression may influence disease pathogenesis by altering immune regulatory pathways mediated by TNF-α, IFN-γ, and IL-10, as well as parallel overexpression of HERV-E gag. The decrease in TLR3 could indicate a compromised antiviral response, which could facilitate viral reactivation and contribute to disease activity.


Subject(s)
Endogenous Retroviruses , Herpesvirus 4, Human , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/virology , Endogenous Retroviruses/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Adult , Female , Male , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/metabolism , Gene Expression Profiling , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Middle Aged , B-Lymphocytes/immunology , B-Lymphocytes/virology , Case-Control Studies , T-Lymphocytes/immunology , Cytokines/metabolism , Cytokines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL