Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.688
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38731951

RESUMO

Distal sensory polyneuropathy (DSP) and distal neuropathic pain (DNP) remain significant challenges for older people with HIV (PWH), necessitating enhanced clinical attention. HIV and certain antiretroviral therapies (ARTs) can compromise mitochondrial function and impact mitochondrial DNA (mtDNA) replication, which is linked to DSP in ART-treated PWH. This study investigated mtDNA, mitochondrial fission and fusion proteins, and mitochondrial electron transport chain protein changes in the dorsal root ganglions (DRGs) and sural nerves (SuNs) of 11 autopsied PWH. In antemortem standardized assessments, six had no or one sign of DSP, while five exhibited two or more DSP signs. Digital droplet polymerase chain reaction was used to measure mtDNA quantity and the common deletions in isolated DNA. We found lower mtDNA copy numbers in DSP+ donors. SuNs exhibited a higher proportion of mtDNA common deletion than DRGs in both groups. Mitochondrial electron transport chain (ETC) proteins were altered in the DRGs of DSP+ compared to DSP- donors, particularly Complex I. These findings suggest that reduced mtDNA quantity and increased common deletion abundance may contribute to DSP in PWH, indicating diminished mitochondrial activity in the sensory neurons. Accumulated ETC proteins in the DRG imply impaired mitochondrial transport to the sensory neuron's distal portion. Identifying molecules to safeguard mitochondrial integrity could aid in treating or preventing HIV-associated peripheral neuropathy.


Assuntos
DNA Mitocondrial , Infecções por HIV , Humanos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Masculino , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Infecções por HIV/genética , Projetos Piloto , Feminino , Pessoa de Meia-Idade , Idoso , Gânglios Espinais/metabolismo , Gânglios Espinais/virologia , Mitocôndrias/metabolismo , Mitocôndrias/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Nervos Periféricos/metabolismo , Nervos Periféricos/virologia , Nervos Periféricos/patologia , Adulto , Nervo Sural/metabolismo , Nervo Sural/patologia
2.
Nat Commun ; 15(1): 3813, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714682

RESUMO

Innate antiviral factors are essential for effective defense against viral pathogens. However, the identity of major restriction mechanisms remains elusive. Current approaches to discover antiviral factors usually focus on the initial steps of viral replication and are limited to a single round of infection. Here, we engineered libraries of >1500 replication-competent HIV-1 constructs each expressing a single gRNAs to target >500 cellular genes for virus-driven discovery of antiviral factors. Passaging in CD4+ T cells robustly enriched HIV-1 encoding sgRNAs against GRN, CIITA, EHMT2, CEACAM3, CC2D1B and RHOA by >50-fold. Using an HIV-1 library lacking the accessory nef gene, we identified IFI16 as a Nef target. Functional analyses in cell lines and primary CD4+ T cells support that the HIV-driven CRISPR screen identified restriction factors targeting virus entry, transcription, release and infectivity. Our HIV-guided CRISPR technique enables sensitive discovery of physiologically relevant cellular defense factors throughout the entire viral replication cycle.


Assuntos
Linfócitos T CD4-Positivos , HIV-1 , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Humanos , HIV-1/genética , HIV-1/fisiologia , Replicação Viral/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Células HEK293 , Sistemas CRISPR-Cas , Infecções por HIV/virologia , Infecções por HIV/genética , Infecções por HIV/imunologia , RNA Guia de Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Internalização do Vírus
3.
AIDS Res Ther ; 21(1): 27, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38698440

RESUMO

BACKGROUND: Human genetic contribution to HIV progression remains inadequately explained. The type 1 interferon (IFN) pathway is important for host control of HIV and variation in type 1 IFN genes may contribute to disease progression. This study assessed the impact of variations at the gene and pathway level of type 1 IFN on HIV-1 viral load (VL). METHODS: Two cohorts of antiretroviral (ART) naïve participants living with HIV (PLWH) with either early (START) or advanced infection (FIRST) were analysed separately. Type 1 IFN genes (n = 17) and receptor subunits (IFNAR1, IFNAR2) were examined for both cumulated type 1 IFN pathway analysis and individual gene analysis. SKAT-O was applied to detect associations between the genotype and HIV-1 study entry viral load (log10 transformed) as a proxy for set point VL; P-values were corrected using Bonferroni (P < 0.0025). RESULTS: The analyses among those with early infection included 2429 individuals from five continents. The median study entry HIV VL was 14,623 (IQR 3460-45100) copies/mL. Across 673 SNPs within 19 type 1 IFN genes, no significant association with study entry VL was detected. Conversely, examining individual genes in START showed a borderline significant association between IFNW1, and study entry VL (P = 0.0025). This significance remained after separate adjustments for age, CD4+ T-cell count, CD4+/CD8+ T-cell ratio and recent infection. When controlling for population structure using linear mixed effects models (LME), in addition to principal components used in the main model, this was no longer significant (p = 0.0244). In subgroup analyses stratified by geographical region, the association between IFNW1 and study entry VL was only observed among African participants, although, the association was not significant when controlling for population structure using LME. Of the 17 SNPs within the IFNW1 region, only rs79876898 (A > G) was associated with study entry VL (p = 0.0020, beta = 0.32; G associated with higher study entry VL than A) in single SNP association analyses. The findings were not reproduced in FIRST participants. CONCLUSION: Across 19 type 1 IFN genes, only IFNW1 was associated with HIV-1 study entry VL in a cohort of ART-naïve individuals in early stages of their infection, however, this was no longer significant in sensitivity analyses that controlled for population structures using LME.


Assuntos
Infecções por HIV , HIV-1 , Interferon Tipo I , Polimorfismo de Nucleotídeo Único , Carga Viral , Humanos , Infecções por HIV/virologia , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/genética , Interferon Tipo I/genética , Masculino , Feminino , Adulto , Genótipo , Pessoa de Meia-Idade , Receptor de Interferon alfa e beta/genética , Estudos de Coortes , Progressão da Doença , Contagem de Linfócito CD4
4.
Methods Mol Biol ; 2807: 209-227, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38743231

RESUMO

The post-transcriptional processing and chemical modification of HIV RNA are understudied aspects of HIV virology, primarily due to the limited ability to accurately map and quantify RNA modifications. Modification-specific antibodies or modification-sensitive endonucleases coupled with short-read RNA sequencing technologies have allowed for low-resolution or limited mapping of important regulatory modifications of HIV RNA such as N6-methyladenosine (m6A). However, a high-resolution map of where these sites occur on HIV transcripts is needed for detailed mechanistic understanding. This has recently become possible with new sequencing technologies. Here, we describe the direct RNA sequencing of HIV transcripts using an Oxford Nanopore Technologies sequencer and the use of this technique to map m6A at near single nucleotide resolution. This technology also provides the ability to identify splice variants with long RNA reads and thus, can provide high-resolution RNA modification maps that distinguish between overlapping splice variants. The protocols outlined here for m6A also provide a powerful paradigm for studying any other RNA modifications that can be detected on the nanopore platform.


Assuntos
Adenosina , Sequenciamento por Nanoporos , RNA Mensageiro , RNA Viral , Sequenciamento por Nanoporos/métodos , RNA Viral/genética , Metilação , Humanos , Adenosina/análogos & derivados , Adenosina/genética , RNA Mensageiro/genética , Análise de Sequência de RNA/métodos , HIV-1/genética , Processamento Pós-Transcricional do RNA , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Infecções por HIV/virologia , Infecções por HIV/genética , HIV/genética
5.
Methods Mol Biol ; 2807: 229-242, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38743232

RESUMO

The identification of RNA modifications at single nucleotide resolution has become an emerging area of interest within biology and specifically among virologists seeking to ascertain how this untapped area of RNA regulation may be altered or hijacked upon viral infection. Herein, we describe a straightforward biochemical approach modified from two original published Ψ mapping protocols, BID-seq and PRAISE, to specifically identify pseudouridine modifications on mRNA transcripts from an HIV-1 infected T cell line. This protocol could readily be adapted for other viral infected cell types and additionally for populations of purified virions from infected cells.


Assuntos
HIV-1 , Pseudouridina , RNA Mensageiro , RNA Viral , Pseudouridina/metabolismo , Pseudouridina/genética , HIV-1/genética , Humanos , RNA Viral/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Infecções por HIV/virologia , Infecções por HIV/genética , Processamento Pós-Transcricional do RNA , Linhagem Celular
6.
Sci Rep ; 14(1): 10852, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741006

RESUMO

Hematopoietic stem-cell (HSC) transplantation using a donor with a homozygous mutation in the HIV co-receptor CCR5 (CCR5Δ32/Δ32) holds great promise as a cure for HIV-1. Previously, there were three patients that had been reported to be completely cured from HIV infection by this approach. However, finding a naturally suitable Human Leukocyte Antigen (HLA)-matched homozygous CCR5Δ32 donor is very difficult. The prevalence of this allele is only 1% in the Caucasian population. Therefore, additional sources of CCR5Δ32/Δ32 HSCs are required. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) system is one method to mediate CCR5 knockout in HSCs that has been successfully employed as a gene editing tool in clinical trials. Additional anti-HIV-1 strategies are still required for broad-spectrum inhibition of HIV-1 replication. Here in this study, we combined an additional anti-HIV-1 therapy, which is C46, a cell membrane-anchored HIV-1 fusion inhibitor with the CRISPR/Cas9 mediated knockout CCR5. The combined HIV-1 therapeutic genes were investigated for the potential prevention of both CCR5 (R5)- and CXCR4 (X4)-tropic HIV-1 infections in the MT4CCR5 cell line. The combinatorial CRISPR/Cas9 therapies were superior compared to single method therapy for achieving the HIV-1 cure strategy and shows potential for future applications.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Inibidores da Fusão de HIV , Infecções por HIV , HIV-1 , Receptores CCR5 , Receptores CCR5/genética , Receptores CCR5/metabolismo , Edição de Genes/métodos , Humanos , HIV-1/genética , HIV-1/efeitos dos fármacos , Infecções por HIV/genética , Infecções por HIV/virologia , Infecções por HIV/terapia , Inibidores da Fusão de HIV/farmacologia , Linhagem Celular , Replicação Viral/efeitos dos fármacos , Proteínas Recombinantes de Fusão
7.
Am J Reprod Immunol ; 91(5): e13845, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38720636

RESUMO

PROBLEM: Bacterial vaginosis (BV) disproportionally impacts Black and Hispanic women, placing them at risk for HIV, sexually transmitted infections and preterm birth. It is unknown whether there are differences by genetic ancestry in BV risk or whether polymorphisms associated with BV risk differ by ancestry. METHODS: Women's Interagency HIV Study (WIHS) participants with longitudinal Nugent scores were dichotomized as having (n = 319, Nugent 7-10) or not having BV (n = 367, Nugent 0-3). Genetic ancestry was defined by clustering of principal components from ancestry informative markers and further stratified by BV status. 627 single nucleotide polymorphisms (SNPs) across 41 genes important in mucosal defense were identified in the WIHS GWAS. A logistic regression analysis was adjusted for nongenetic predictors of BV and self-reported race/ethnicity to assess associations between genetic ancestry and genotype. RESULTS: Self-reported race and genetic ancestry were associated with BV risk after adjustment for behavioral factors. Polymorphisms in mucosal defense genes including syndecans, cytokines and toll-like receptors (TLRs) were associated with BV in all ancestral groups. CONCLUSIONS: The common association of syndecan, cytokine and TLR genes and the importance of immune function and inflammatory pathways in BV, suggests these should be targeted for further research on BV pathogenesis and therapeutics.


Assuntos
Infecções por HIV , Polimorfismo de Nucleotídeo Único , Vaginose Bacteriana , Humanos , Feminino , Vaginose Bacteriana/genética , Adulto , Infecções por HIV/genética , Predisposição Genética para Doença , Citocinas/genética , Fatores de Risco , Estudo de Associação Genômica Ampla , Receptores Toll-Like/genética
9.
Viruses ; 16(4)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38675845

RESUMO

Schlafen (SLFN) is a family of proteins upregulated by type I interferons with a regulatory role in translation. Intriguingly, SLFN14 associates with the ribosome and can degrade rRNA, tRNA, and mRNA in vitro, but a role in translation is still unknown. Ribosomes are important regulatory hubs during translation elongation of mRNAs rich in rare codons. Therefore, we evaluated the potential role of SLFN14 in the expression of mRNAs enriched in rare codons, using HIV-1 genes as a model. We found that, in a variety of cell types, including primary immune cells, SLFN14 regulates the expression of HIV-1 and non-viral genes based on their codon adaptation index, a measurement of the synonymous codon usage bias; consequently, SLFN14 inhibits the replication of HIV-1. The potent inhibitory effect of SLFN14 on the expression of the rare codon-rich transcript HIV-1 Gag was minimized by codon optimization. Mechanistically, we found that the endoribonuclease activity of SLFN14 is required, and that ribosomal RNA degradation is involved. Therefore, we propose that SLFN14 impairs the expression of HIV-1 transcripts rich in rare codons, in a catalytic-dependent manner.


Assuntos
Uso do Códon , HIV-1 , Replicação Viral , Humanos , Códon/genética , Regulação Viral da Expressão Gênica , Células HEK293 , Infecções por HIV/virologia , Infecções por HIV/genética , HIV-1/genética , HIV-1/fisiologia , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linhagem Celular Tumoral
10.
Viruses ; 16(4)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38675842

RESUMO

TREX1 acts in the initial prevention of an autoimmune response, but it may contribute to the permissiveness of retrovirus infections. This study investigated the association between the levels of TREX1 gene expression with the polymorphisms TREX1 rs3135941 (T/C) and TREX1 rs3135945 (G/A), and the presence of antinuclear antibodies (ANA) in antiretroviral therapy (ART)-naïve individuals and after 1 year of treatment. Blood samples from 119 individuals with HIV-1 were subjected to genotyping of polymorphisms and quantification of TREX1 gene expression and HIV-1 viral load by qPCR. The concentration of IFN-α and the number of CD4+/CD8+ T lymphocytes were determined by ELISA and flow cytometry, respectively; ANA was investigated by immunofluorescence. A control group of 167 seronegative individuals was used for the comparison of genotypic frequencies. The frequency of the polymorphisms were not associated with HIV infection or with variations in the expression of TREX1 and IFN-α (p > 0.05). ART-naïve individuals exhibited higher TREX1 expression and lower IFN-α expression. After 1 year of ART, TREX1 levels were reduced, while IFN-α and CD4+ T lymphocytes were elevated (p < 0.05). Some individuals on ART presented ANA. These results suggest that ART-mediated restoration of immune competence is associated with a reduction in TREX1 expression, which may induce the development of ANA, regardless of the polymorphism investigated.


Assuntos
Exodesoxirribonucleases , Infecções por HIV , HIV-1 , Reconstituição Imune , Fosfoproteínas , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Anticorpos Antinucleares/sangue , Linfócitos T CD4-Positivos/imunologia , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Genótipo , Infecções por HIV/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/imunologia , Reconstituição Imune/genética , Reconstituição Imune/imunologia , Interferon-alfa/sangue , Interferon-alfa/metabolismo , Fosfoproteínas/genética , Polimorfismo de Nucleotídeo Único , Carga Viral , Antirretrovirais/efeitos adversos , Antirretrovirais/uso terapêutico
11.
Front Immunol ; 15: 1369311, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38601162

RESUMO

Background: Coronavirus disease (COVID-19), caused by SARS-CoV-2, has emerged as a infectious disease, coexisting with widespread seasonal and sporadic influenza epidemics globally. Individuals living with HIV, characterized by compromised immune systems, face an elevated risk of severe outcomes and increased mortality when affected by COVID-19. Despite this connection, the molecular intricacies linking COVID-19, influenza, and HIV remain unclear. Our research endeavors to elucidate the shared pathways and molecular markers in individuals with HIV concurrently infected with COVID-19 and influenza. Furthermore, we aim to identify potential medications that may prove beneficial in managing these three interconnected illnesses. Methods: Sequencing data for COVID-19 (GSE157103), influenza (GSE185576), and HIV (GSE195434) were retrieved from the GEO database. Commonly expressed differentially expressed genes (DEGs) were identified across the three datasets, followed by immune infiltration analysis and diagnostic ROC analysis on the DEGs. Functional enrichment analysis was performed using GO/KEGG and Gene Set Enrichment Analysis (GSEA). Hub genes were screened through a Protein-Protein Interaction networks (PPIs) analysis among DEGs. Analysis of miRNAs, transcription factors, drug chemicals, diseases, and RNA-binding proteins was conducted based on the identified hub genes. Finally, quantitative PCR (qPCR) expression verification was undertaken for selected hub genes. Results: The analysis of the three datasets revealed a total of 22 shared DEGs, with the majority exhibiting an area under the curve value exceeding 0.7. Functional enrichment analysis with GO/KEGG and GSEA primarily highlighted signaling pathways associated with ribosomes and tumors. The ten identified hub genes included IFI44L, IFI44, RSAD2, ISG15, IFIT3, OAS1, EIF2AK2, IFI27, OASL, and EPSTI1. Additionally, five crucial miRNAs (hsa-miR-8060, hsa-miR-6890-5p, hsa-miR-5003-3p, hsa-miR-6893-3p, and hsa-miR-6069), five essential transcription factors (CREB1, CEBPB, EGR1, EP300, and IRF1), and the top ten significant drug chemicals (estradiol, progesterone, tretinoin, calcitriol, fluorouracil, methotrexate, lipopolysaccharide, valproic acid, silicon dioxide, cyclosporine) were identified. Conclusion: This research provides valuable insights into shared molecular targets, signaling pathways, drug chemicals, and potential biomarkers for individuals facing the complex intersection of COVID-19, influenza, and HIV. These findings hold promise for enhancing the precision of diagnosis and treatment for individuals with HIV co-infected with COVID-19 and influenza.


Assuntos
COVID-19 , Infecções por HIV , Influenza Humana , MicroRNAs , Humanos , Influenza Humana/genética , COVID-19/genética , SARS-CoV-2 , Biologia Computacional , MicroRNAs/genética , Fatores de Transcrição , Regulação da Expressão Gênica , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética
12.
Proc Natl Acad Sci U S A ; 121(15): e2316662121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38557187

RESUMO

Drug resistance in HIV type 1 (HIV-1) is a pervasive problem that affects the lives of millions of people worldwide. Although records of drug-resistant mutations (DRMs) have been extensively tabulated within public repositories, our understanding of the evolutionary kinetics of DRMs and how they evolve together remains limited. Epistasis, the interaction between a DRM and other residues in HIV-1 protein sequences, is key to the temporal evolution of drug resistance. We use a Potts sequence-covariation statistical-energy model of HIV-1 protein fitness under drug selection pressure, which captures epistatic interactions between all positions, combined with kinetic Monte-Carlo simulations of sequence evolutionary trajectories, to explore the acquisition of DRMs as they arise in an ensemble of drug-naive patient protein sequences. We follow the time course of 52 DRMs in the enzymes protease, RT, and integrase, the primary targets of antiretroviral therapy. The rates at which DRMs emerge are highly correlated with their observed acquisition rates reported in the literature when drug pressure is applied. This result highlights the central role of epistasis in determining the kinetics governing DRM emergence. Whereas rapidly acquired DRMs begin to accumulate as soon as drug pressure is applied, slowly acquired DRMs are contingent on accessory mutations that appear only after prolonged drug pressure. We provide a foundation for using computational methods to determine the temporal evolution of drug resistance using Potts statistical potentials, which can be used to gain mechanistic insights into drug resistance pathways in HIV-1 and other infectious agents.


Assuntos
Fármacos Anti-HIV , Infecções por HIV , Soropositividade para HIV , HIV-1 , Humanos , HIV-1/genética , Farmacorresistência Viral/genética , Genótipo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Mutação , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico
13.
Cell Stem Cell ; 31(4): 499-518.e6, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38579682

RESUMO

Allogeneic hematopoietic stem and progenitor cell transplant (HSCT) of CCR5 null (CCR5Δ32) cells can be curative for HIV-1-infected patients. However, because allogeneic HSCT poses significant risk, CCR5Δ32 matched bone marrow donors are rare, and CCR5Δ32 transplant does not confer resistance to the CXCR4-tropic virus, it is not a viable option for most patients. We describe a targeted Cas9/AAV6-based genome editing strategy for autologous HSCT resulting in both CCR5- and CXCR4-tropic HIV-1 resistance. Edited human hematopoietic stem and progenitor cells (HSPCs) maintain multi-lineage repopulation capacity in vivo, and edited primary human T cells potently inhibit infection by both CCR5-tropic and CXCR4-tropic HIV-1. Modification rates facilitated complete loss of CCR5-tropic replication and up to a 2,000-fold decrease in CXCR4-tropic replication without CXCR4 locus disruption. This multi-factor editing strategy in HSPCs could provide a broad approach for autologous HSCT as a functional cure for both CCR5-tropic and CXCR4-tropic HIV-1 infections.


Assuntos
Edição de Genes , Infecções por HIV , HIV-1 , Humanos , Edição de Genes/métodos , Células-Tronco Hematopoéticas , Infecções por HIV/genética , Infecções por HIV/terapia , HIV-1/genética , Receptores CCR5/genética , Receptores CXCR4/genética
14.
Eur Rev Med Pharmacol Sci ; 28(6): 2430-2463, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38567606

RESUMO

Human Immunodeficiency Virus (HIV) has continuously been the greatest epidemic for humanity over a period spanning almost five decades. With no specific cure or treatment available to date despite extensive research, the C-C Chemokine Receptor 5, Delta 32 (CCR5 Δ32) allele genetic point mutation plays an imperative role in the prevention of acquired immunodeficiency syndrome (AIDS). This comprehensive study aims to review the induction of the homozygous recessive deletion genotype using the Clustered Regularly Interspaced Short Palindromic Repeats, Cas 9 Enzyme (CRISPR-Cas9), and hematopoietic stem cell transplantation under positive selection pressure for active immunity in seropositive patients' populations as the phenotype. A methodology is proposed to trigger a significant increase in the expression of Delta 32 beneficial mutant alleles within controlled modern healthcare facilities utilizing totipotent stem cells through somatic gene therapy. It acts upon two dysfunctional CCR5 genes, translating mutant G protein-coupled co-receptors, whose primary function is similar to that of C-X-C Motif Chemokine receptor 4 (CXCR4), by blocking the entry of viral RNA into the CD4+ T helper lymphocytes, halting infection and seizing viral life cycle. This modification is endemic in Northern Europe, where it naturally pertains to the Caucasian descent population samples in the form of polymorphism, p (X=0.01), where X is the probability of frequency of complete immunity against HIV-1 in population samples. The epigenetics of the single nucleotide polymorphism (SNP) are analyzed as they play a significant role in immunity distribution. Furthermore, a comparative analysis within the ethical boundaries of CRISPR-Cas9 is conducted to discuss the practical aspects and challenges of the presented methodologies and treatment alternatives. Additionally, the study assembles all available data and summarizes preexisting research while providing a promising solution to this ethical dilemma. Finally, a methodology is devised to answer the question of whether the variant-specific epidemic of AIDS caused by HIV-1 can be cured via artificially inducing immunity by CRISPR-Cas9.


Assuntos
Síndrome da Imunodeficiência Adquirida , Infecções por HIV , HIV-1 , Humanos , HIV-1/genética , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/terapia , Infecções por HIV/genética , Infecções por HIV/terapia , Sistemas CRISPR-Cas/genética , Receptores CCR5/genética , Receptores CCR5/metabolismo , Mutação , Terapia Genética , Polimorfismo de Nucleotídeo Único , Frequência do Gene
15.
Viruses ; 16(4)2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38675828

RESUMO

The innate immune response to viruses is formed in part by interferon (IFN)-induced restriction factors, including ISG15, p21, and SAMHD1. IFN production can be blocked by the ISG15-specific protease USP18. HIV-1 has evolved to circumvent host immune surveillance. This mechanism might involve USP18. In our recent studies, we demonstrate that HIV-1 infection induces USP18, which dramatically enhances HIV-1 replication by abrogating the antiviral function of p21. USP18 downregulates p21 by accumulating misfolded dominant negative p53, which inactivates wild-type p53 transactivation, leading to the upregulation of key enzymes involved in de novo dNTP biosynthesis pathways and inactivated SAMHD1. Despite the USP18-mediated increase in HIV-1 DNA in infected cells, it is intriguing to note that the cGAS-STING-mediated sensing of the viral DNA is abrogated. Indeed, the expression of USP18 or knockout of ISG15 inhibits the sensing of HIV-1. We demonstrate that STING is ISGylated at residues K224, K236, K289, K347, K338, and K370. The inhibition of STING K289-linked ISGylation suppresses its oligomerization and IFN induction. We propose that human USP18 is a novel factor that potentially contributes in multiple ways to HIV-1 replication.


Assuntos
HIV-1 , Ubiquitina Tiolesterase , Ubiquitinas , Replicação Viral , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Humanos , HIV-1/fisiologia , HIV-1/genética , Ubiquitinas/metabolismo , Ubiquitinas/genética , Citocinas/metabolismo , Citocinas/genética , Imunidade Inata , Infecções por HIV/virologia , Infecções por HIV/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Interações Hospedeiro-Patógeno , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética
16.
Viruses ; 16(4)2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38675839

RESUMO

Human and viral microRNAs (miRNAs) are involved in the regulation of gene transcription, and the establishment of their profiles in acute (AHI) and chronic (CHI) HIV infections may shed light on the pathogenetic events related to different phases of HIV disease. Next-generation sequencing (NGS) of miRNA libraries was performed, and the reads were used to analyze miRNA differential expression in the plasma with AHI and CHI. Functional analysis was then undertaken to investigate the biological processes characterizing the two phases of HIV infection. Except for hsa-miR-122-5p, which was found in 3.39% AHI vs. 0.18% CHI, the most represented human miRNAs were similarly represented in AHI and CHI. However, when considering the overall detected miRNAs in AHI and CHI, 15 displayed differential expression (FDR p < 0.05). Functional analysis identified 163 target mRNAs involved in promoting angiogenesis activation in AHI versus CHI through the action of hsa-miR10b-5p, hsa-miR1290, hsa-miR1-3p, and hsa-miR296-5p. The viral miRNAs detected, all belonging to herpesviruses, accounted for only 0.014% of total reads. The present data suggest that AHI patients exhibit strong innate immune activation through the upregulation of hsa-miR-122-5p and early activation of angiogenesis. More specific investigations are needed to study the role of viral miRNAs in HIV pathogenesis.


Assuntos
Infecções por HIV , Sequenciamento de Nucleotídeos em Larga Escala , MicroRNAs , RNA Viral , Humanos , MicroRNAs/genética , Infecções por HIV/virologia , Infecções por HIV/genética , RNA Viral/genética , Perfilação da Expressão Gênica , Masculino , Adulto , Feminino , Doença Aguda , Doença Crônica , Pessoa de Meia-Idade , HIV-1/genética , Imunidade Inata , Regulação da Expressão Gênica
17.
Viruses ; 16(4)2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38675925

RESUMO

The interferon-induced host cell protein Shiftless (SFL) inhibits -1 programmed ribosomal frameshifting (-1PRF) required for the expression of HIV-1 Gal-Pol and the formation of infectious HIV-1 particles. However, the specific regions in SFL required for antiviral activity and the mechanism by which SFL inhibits -1PRF remain unclear. Employing alanine scanning mutagenesis, we found that basic amino acids in the predicted zinc ribbon motif of SFL are essential for the suppression of Gag-Pol expression but dispensable for anti-HIV-1 activity. We have shown that SFL inhibits the expression of the murine leukemia virus (MLV) Gag-Pol polyprotein and the formation of infectious MLV particles, although Gag-Pol expression of MLV is independent of -1PRF but requires readthrough of a stop codon. These findings indicate that SFL might inhibit HIV-1 infection by more than one mechanism and that SFL might target programmed translational readthrough as well as -1PRF signals, both of which are regulated by mRNA secondary structure elements.


Assuntos
Proteínas de Fusão gag-pol , Infecções por HIV , HIV-1 , HIV-1/genética , HIV-1/fisiologia , Humanos , Infecções por HIV/virologia , Infecções por HIV/genética , Infecções por HIV/metabolismo , Proteínas de Fusão gag-pol/genética , Proteínas de Fusão gag-pol/metabolismo , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Mudança da Fase de Leitura do Gene Ribossômico , Replicação Viral , Células HEK293 , Regulação Viral da Expressão Gênica
18.
Cell Stem Cell ; 31(4): 433-434, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38579679

RESUMO

The chemokine receptors CCR5 and CXCR4 are "front doors" for HIV-1 infection in host cells, and their targeting represents a potential solution for a cure. Dudek et al.1 now propose a new gene editing strategy to simultaneously block CCR5- and CXCR4-mediated HIV-1 entry in autologous hematopoietic stem and progenitor cells (HSPCs).


Assuntos
Infecções por HIV , HIV-1 , Humanos , HIV-1/genética , Células-Tronco Hematopoéticas , Receptores CCR5/genética , Infecções por HIV/genética , Receptores CXCR4/genética , Edição de Genes
19.
PLoS Pathog ; 20(4): e1012172, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38662769

RESUMO

The implementation of antiretroviral therapy (ART) has effectively restricted the transmission of Human Immunodeficiency Virus (HIV) and improved overall clinical outcomes. However, a complete cure for HIV remains out of reach, as the virus persists in a stable pool of infected cell reservoir that is resistant to therapy and thus a main barrier towards complete elimination of viral infection. While the mechanisms by which host proteins govern viral gene expression and latency are well-studied, the emerging regulatory functions of non-coding RNAs (ncRNA) in the context of T cell activation, HIV gene expression and viral latency have not yet been thoroughly explored. Here, we report the identification of the Cytoskeleton Regulator (CYTOR) long non-coding RNA (lncRNA) as an activator of HIV gene expression that is upregulated following T cell stimulation. Functional studies show that CYTOR suppresses viral latency by directly binding to the HIV promoter and associating with the cellular positive transcription elongation factor (P-TEFb) to activate viral gene expression. CYTOR also plays a global role in regulating cellular gene expression, including those involved in controlling actin dynamics. Depletion of CYTOR expression reduces cytoplasmic actin polymerization in response to T cell activation. In addition, treating HIV-infected cells with pharmacological inhibitors of actin polymerization reduces HIV gene expression. We conclude that both direct and indirect effects of CYTOR regulate HIV gene expression.


Assuntos
Regulação Viral da Expressão Gênica , Infecções por HIV , HIV-1 , RNA Longo não Codificante , Latência Viral , Humanos , RNA Longo não Codificante/genética , Infecções por HIV/virologia , Infecções por HIV/genética , HIV-1/genética , HIV-1/fisiologia , Regiões Promotoras Genéticas , Ativação Linfocitária , Células Jurkat
20.
J Neuroinflammation ; 21(1): 107, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38659061

RESUMO

Neuroinflammation and synaptodendritic damage represent the pathological hallmarks of HIV-1 associated cognitive disorders (HAND). The post-synaptic protein neurogranin (Nrgn) is significantly reduced in the frontal cortex of postmortem brains from people with HIV (PWH) and it is associated with inflammatory factors released by infected microglia/macrophages. However, the mechanism involved in synaptic loss have yet to be elucidated. In this study, we characterized a newly identified long non-coding RNA (lncRNA) transcript (RP11-677M14.2), which is antisense to the NRGN locus and is highly expressed in the frontal cortex of HIV-1 individuals. Further analysis indicates an inverse correlation between the expression of RP11-677M14.2 RNA and Nrgn mRNA. Additionally, the Nrgn-lncRNA axis is dysregulated in neurons exposed to HIV-1 infected microglia conditioned medium enriched with IL-1ß. Moreover, in vitro overexpression of this lncRNA impacts Nrgn expression at both mRNA and protein levels. Finally, we modeled the Nrgn-lncRNA dysregulation within an HIV-1-induced inflammatory environment using brain organoids, thereby corroborating our in vivo and in vitro findings. Together, our study implicates a plausible role for lncRNA RP11-677M14.2 in modulating Nrgn expression that might serve as the mechanistic link between Nrgn loss and cognitive dysfunction in HAND, thus shedding new light on the mechanisms underlying synaptodendritic damage.


Assuntos
HIV-1 , Neurogranina , Doenças Neuroinflamatórias , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/metabolismo , RNA Longo não Codificante/genética , Neurogranina/metabolismo , Neurogranina/genética , Doenças Neuroinflamatórias/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/genética , Infecções por HIV/patologia , Microglia/metabolismo , Masculino , Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA