Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pediatr Res ; 93(6): 1626-1633, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36434204

RESUMEN

BACKGROUND: One potential mechanism for protection from SARS-CoV-2 in children is through passive immunity via breast milk from a mother infected with the novel coronavirus. The primary objectives of this study were to establish the presence of SARS-CoV-2-specific IgA and IgG and to characterize the antigenic regions of SARS-CoV-2 proteins that were reactive with antibodies in breast milk. METHODS: Between March 2020 and September 2020, 21 women with confirmed SARS-CoV-2 infection were enrolled in Mommy's Milk. Participants donated serial breast milk samples around their time of illness. Breast milk samples were used to probe a multi-coronavirus protein microarray containing full-length and variable-length overlapping fragments of SARS-CoV-2 proteins. Samples were also tested against S and N proteins by electrochemiluminescence assay. RESULTS: The breast milk samples contained IgA reactive with a variety of SARS-CoV-2 antigens. The most IgA-reactive SARS-CoV-2 proteins were N (42.9% of women responded to ≥1 N fragment) and S proteins (23.9% responded to ≥1 fragment of S1 or S2). IgG responses were similar. A striking observation was the dissimilarity between mothers in antibody recognition, giving distinct antibody reactivity and kinetic profiles. CONCLUSIONS: Individual COVID-19 cases had diverse and unique milk IgA profiles following the onset of symptoms. IMPACT: In this observational longitudinal case series of 21 women with confirmed SARS-CoV-2 infection, IgA binding to SARS-CoV-2 proteins detected by orthologous proteome microarray and electrochemiluminescence assays was observed in >75% of women, but there was heterogeneity in which antigens and how many were reactive between women. Immunological profiles of protein regions recognized by each woman were distinct. Diverse repertoires of mucosal breast milk antibody to SARS-CoV-2 reflect heterogeneous passive transfer of maternal antibody to exposed breastfeeding infants.


Asunto(s)
COVID-19 , Leche Humana , Niño , Lactante , Humanos , Femenino , SARS-CoV-2 , Anticuerpos Antivirales , Inmunoglobulina A , Inmunoglobulina G
2.
J Virol ; 91(23)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28931690

RESUMEN

Previous studies in our laboratory showed that the RNA debranching enzyme (DBR1) is not required for early steps in HIV cDNA formation but is necessary for synthesis of intermediate and late cDNA products. To further characterize this effect, we evaluated the topology of the 5' end of the HIV-1 RNA genome during early infection with and without inhibition of DBR1 synthesis. Cells were transfected with DBR1 short hairpin RNA (shRNA) followed 48 h later by infection with an HIV-1-derived vector containing an RNase H-deficient reverse transcriptase (RT). RNA was isolated at several times postinfection and treated with various RNA-modifying enzymes prior to rapid amplification of 5' cDNA ends (5' RACE) for HIV-1 RNA and quantitative reverse transcriptase PCR (qRT-PCR). In infected cells, DBR1 knockdown inhibited detection of free HIV-1 RNA 5' ends at all time points. The difference in detection of free HIV-1 RNA 5' ends in infected DBR1 knockdown versus control cells was eliminated by in vitro incubation of infected cell RNAs with yeast or human DBR1 enzyme prior to 5' RACE and qRT-PCR. This was dependent on the 2'-5' phosphatase activity of DBR1, since it did not occur when we used the catalytically inactive DBR1(N85A) mutant. Finally, HIV-1 RNA from infected DBR1 knockdown cells was resistant to RNase R that degrades linear RNAs but not RNAs in circular or lariat-like conformations. These results provide evidence for formation of a lariat-like structure involving the 5' end of HIV-1 RNA during an early step in infection and the involvement of DBR1 in resolving it.IMPORTANCE Our findings support a new view of the early steps in HIV genome replication. We show that the HIV genomic RNA is rapidly decapped and forms a lariat-like structure after entering a cell. The lariat-like structure is subsequently resolved by the cellular enzyme DBR1, leaving a 5' phosphate. This pathway is similar to the formation and resolution of pre-mRNA intron lariats and therefore suggests that similar mechanisms may be used by HIV. Our work therefore opens a new area of investigation in HIV replication and may ultimately uncover new targets for inhibiting HIV replication and for preventing the development of AIDS.


Asunto(s)
Genoma Viral , VIH-1/genética , Caperuzas de ARN/química , ARN Nucleotidiltransferasas/genética , ARN Viral/química , Transcripción Reversa , Células HEK293 , VIH-1/química , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Nucleotidiltransferasas/deficiencia , ARN Nucleotidiltransferasas/metabolismo , ARN Nucleotidiltransferasas/farmacología , Precursores del ARN/química , Empalme del ARN , ARN Interferente Pequeño , ARN Viral/metabolismo , Saccharomyces cerevisiae/genética , Replicación Viral
3.
Bull World Health Organ ; 95(7): 517-525I, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28670016

RESUMEN

OBJECTIVE: To analyse the proportions of protein identity between Zika virus and dengue, Japanese encephalitis, yellow fever, West Nile and chikungunya viruses as well as polymorphism between different Zika virus strains. METHODS: We used published protein sequences for the Zika virus and obtained protein sequences for the other viruses from the National Center for Biotechnology Information (NCBI) protein database or the NCBI virus variation resource. We used BLASTP to find regions of identity between viruses. We quantified the identity between the Zika virus and each of the other viruses, as well as within-Zika virus polymorphism for all amino acid k-mers across the proteome, with k ranging from 6 to 100. We assessed accessibility of protein fragments by calculating the solvent accessible surface area for the envelope and nonstructural-1 (NS1) proteins. FINDINGS: In total, we identified 294 Zika virus protein fragments with both low proportion of identity with other viruses and low levels of polymorphisms among Zika virus strains. The list includes protein fragments from all Zika virus proteins, except NS3. NS4A has the highest number (190 k-mers) of protein fragments on the list. CONCLUSION: We provide a candidate list of protein fragments that could be used when developing a sensitive and specific serological test to detect previous Zika virus infections.


Asunto(s)
Virus Chikungunya/ultraestructura , Bases de Datos de Proteínas , Flavivirus/ultraestructura , Animales , Virus del Dengue/ultraestructura , Virus de la Encefalitis Japonesa (Especie)/ultraestructura , Humanos , Insectos Vectores , Polimorfismo Genético , Virus del Nilo Occidental/ultraestructura , Virus de la Fiebre Amarilla/ultraestructura , Virus Zika/ultraestructura
4.
PLoS Pathog ; 10(8): e1004345, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25167059

RESUMEN

A rare subset of HIV-infected individuals, designated viremic non-progressors (VNP), remain asymptomatic and maintain normal levels of CD4+ T-cells despite persistently high viremia. To identify mechanisms potentially responsible for the VNP phenotype, we compared VNPs (average >9 years of HIV infection) to HIV-infected individuals who have similar CD4+ T-cell counts and viral load, but who are likely to progress if left untreated ("putative progressors", PP), thus avoiding the confounding effect of differences related to substantial CD4+ T cell depletion. We found that VNPs, compared to PPs, had preserved levels of CD4+ stem cell memory cells (TSCM (p<0.0001), which was associated with decreased HIV infection of these cells in VNPs (r = -0.649, p = 0.019). In addition, VNPs had decreased HIV infection in CD4+ central memory (TCM) cells (p = 0.035), and the total number of TCM cells was associated with increased proliferation of memory CD4+ T cells (r = 0.733, p = 0.01). Our results suggest that, in HIV-infected VNPs, decreased infection of CD4+ TCM and TSCM, cells are involved in preservation of CD4+ T cell homeostasis and lack of disease progression despite high viremia.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , Memoria Inmunológica/inmunología , Subgrupos de Linfocitos T/inmunología , Viremia/inmunología , Linfocitos T CD4-Positivos/virología , Separación Celular , ADN Viral/análisis , Progresión de la Enfermedad , Infecciones por VIH/virología , Humanos , Inmunofenotipificación , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/inmunología , Células Madre/virología , Subgrupos de Linfocitos T/virología , Carga Viral , Viremia/virología
5.
J Virol ; 88(12): 7054-69, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24672043

RESUMEN

UNLABELLED: Previous studies showed that short hairpin RNA (shRNA) knockdown of the RNA lariat debranching enzyme (DBR1) led to a decrease in the production of HIV-1 cDNA. To further characterize this effect, DBR1 shRNA was introduced into GHOST-R5X4 cells, followed by infection at a multiplicity near unity with HIV-1 or an HIV-1-derived vector. DNA and RNA were isolated from whole cells and from cytoplasmic and nuclear fractions at different times postinfection. Inhibition of DBR1 had little or no effect on the formation of minus-strand strong-stop cDNA but caused a significant reduction in the formation of intermediate and full-length cDNA. Moreover, minus-strand strong-stop DNA rapidly accumulated in the cytoplasm in the first 2 h of infection but shifted to the nuclear fraction by 6 h postinfection. Regardless of DBR1 inhibition, greater than 95% of intermediate-length and full-length HIV-1 cDNA was found in the nuclear fraction at all time points. Thus, under these experimental conditions, HIV-1 cDNA synthesis was initiated in the cytoplasm and completed in the nucleus or perinuclear region of the infected cell. When nuclear import of the HIV-1 reverse transcription complex was blocked by expressing a truncated form of the mRNA cleavage and polyadenylation factor CPSF6, the completion of HIV-1 vector cDNA synthesis was detected in the cytoplasm, where it was not inhibited by DBR1 knockdown. Refinement of the cell fractionation procedure indicated that the completion of reverse transcription occurred both within nuclei and in the perinuclear region. Taken together the results indicate that in infections at a multiplicity near 1, HIV-1 reverse transcription is completed in the nucleus or perinuclear region of the infected cell, where it is dependent on DBR1. When nuclear transport is inhibited, reverse transcription is completed in the cytoplasm in a DBR1-independent manner. Thus, there are at least two mechanisms of HIV-1 reverse transcription that require different factors and occur in different intracellular locations. IMPORTANCE: This study shows that HIV-1 reverse transcription starts in the cytoplasm but is completed in or on the surface of the nucleus. Moreover, we show that nuclear reverse transcription is dependent on the activity of the human RNA lariat debranchng enzyme (DBR1), while cytoplasmic reverse transcription is not. These findings may provide new avenues for inhibiting HIV-1 replication and therefore may lead to new medicines for treating HIV-1-infected individuals.


Asunto(s)
ADN Complementario/genética , Infecciones por VIH/enzimología , Infecciones por VIH/virología , VIH-1/genética , ARN Nucleotidiltransferasas/genética , Núcleo Celular/virología , Citoplasma/virología , ADN Complementario/metabolismo , Técnicas de Silenciamiento del Gen , Infecciones por VIH/genética , VIH-1/fisiología , Humanos , ARN Nucleotidiltransferasas/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral
6.
Retrovirology ; 11: 13, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24495362

RESUMEN

BACKGROUND: A small minority of HIV-1-infected individuals show low levels of immune activation and do not develop immunodeficiency despite high viral loads. Since the accessory viral Nef protein modulates T cell activation and plays a key role in the pathogenesis of AIDS, we investigated whether specific properties of Nef may be associated with this highly unusual clinical outcome of HIV-1 infection. FINDINGS: Comprehensive functional analyses of sequential HIV-1 strains from three viremic long-term non-progressors (VNP) showed that they encode full-length Nef proteins that are capable of modulating CD4, CD28, CD8ß, MHC-I and CD74 cell surface expression. Similar to Nef proteins from HIV-1-infected individuals with progressive infection (P-Nefs) and unlike Nefs from simian immunodeficiency viruses (SIVs) that do not cause chronic immune activation and disease in their natural simian hosts, VNP-Nefs were generally unable to down-modulate TCR-CD3 cell surface expression to block T cell activation and apoptosis. On average, VNP-Nefs suppressed NF-AT activation less effectively than P-Nefs and were slightly less active in enhancing NF-κB activity. Finally, we found that VNP-Nefs increased virion infectivity and enhanced HIV-1 replication and cytopathicity in primary human cells and in ex vivo infected lymphoid tissues. CONCLUSIONS: Our results show that nef alleles from VNPs and progressors of HIV-1 infection show only modest differences in established functions. Thus, the lack of chronic immune activation and disease progression in HIV-1-infected VNPs is apparently not associated with unusual functional properties of the accessory viral Nef protein.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/virología , Sobrevivientes de VIH a Largo Plazo , VIH-1/fisiología , Viremia/inmunología , Viremia/virología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Antígenos CD/análisis , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Antígenos de Histocompatibilidad Clase I/análisis , Humanos
7.
Virol J ; 8: 284, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21651801

RESUMEN

BACKGROUND: Retroviral vectors are widely used tools for gene delivery and gene therapy. They are useful for gene expression studies and genetic manipulation in vitro and in vivo. Many retroviral vectors are derived from the mouse gammaretrovirus, murine leukemia virus (MLV). These vectors have been widely used in gene therapy clinical trials. XMRV, initially found in prostate cancer tissue, was the first human gammaretrovirus described. FINDINGS: We developed a new retroviral vector based on XMRV called pXC. It was developed for gene transfer to human cells and is produced by transient cotransfection of LNCaP cells with pXC and XMRV-packaging plasmids. CONCLUSIONS: We demonstrated that pXC mediates expression of inserted transgenes in cell lines. This new vector will be a useful tool for gene transfer in human and non-human cell lines, including gene therapy studies.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/genética , Línea Celular , Terapia Genética/métodos , Humanos , Ensamble de Virus
8.
mBio ; 12(3): e0122921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34182775

RESUMEN

We sought to discover links between antibody responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and patient clinical variables, cytokine profiles, and antibodies to endemic coronaviruses. Serum samples from 30 patients of younger (26 to 39 years) and older (69 to 83 years) age groups and with varying clinical severities ranging from outpatient to mechanically ventilated were collected and used to probe a novel multi-coronavirus protein microarray. This microarray contained variable-length overlapping fragments of SARS-CoV-2 spike (S), envelope (E), membrane (M), nucleocapsid (N), and open reading frame (ORF) proteins created through in vitro transcription and translation (IVTT). The array also contained SARS-CoV, Middle East respiratory syndrome coronavirus (MERS-CoV), human coronavirus OC43 (HCoV-OC43), and HCoV-NL63 proteins. IgG antibody responses to specific epitopes within the S1 protein region spanning amino acids (aa) 500 to 650 and within the N protein region spanning aa 201 to 300 were found to be significantly higher in older patients and further significantly elevated in those older patients who were ventilated. Additionally, there was a noticeable overlap between antigenic regions and known mutation locations in selected emerging SARS-CoV-2 variants of current clinical consequence (B.1.1.7, B1.351, P.1, CAL20.C, and B.1.526). Moreover, the older age group displayed more consistent correlations of antibody reactivity with systemic cytokine and chemokine responses than the younger adult group. A subset of patients, however, had little or no response to SARS-CoV-2 antigens and disproportionately severe clinical outcomes. Further characterization of these slow-low-responding individuals with cytokine analysis revealed significantly higher interleukin-10 (IL-10), IL-15, and interferon gamma-induced protein 10 (IP-10) levels and lower epidermal growth factor (EGF) and soluble CD40 ligand (sCD40L) levels than those of seroreactive patients in the cohort. IMPORTANCE As numerous viral variants continue to emerge in the coronavirus disease 2019 (COVID-19) pandemic, determining antibody reactivity to SARS-CoV-2 epitopes becomes essential in discerning changes in the immune response to infection over time. This study enabled us to identify specific areas of antigenicity within the SARS-CoV-2 proteome, allowing us to detect correlations of epitopes with clinical metadata and immunological signals to gain holistic insight into SARS-CoV-2 infection. This work also emphasized the risk of mutation accumulation in viral variants and the potential for evasion of the adaptive immune responses in the event of reinfection. We additionally highlighted the correlation of antigenicity between structural proteins of SARS-CoV-2 and endemic HCoVs, raising the possibility of cross-protection between homologous lineages. Finally, we identified a subset of patients with minimal antibody reactivity to SARS-CoV-2 infection, prompting discussion of the potential consequences of this alternative immune response.


Asunto(s)
Anticuerpos Antivirales/sangre , Coronavirus Humano NL63/inmunología , Coronavirus Humano OC43/inmunología , Citocinas/sangre , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , SARS-CoV-2/inmunología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Proteínas de la Envoltura de Coronavirus/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Femenino , Humanos , Inmunoglobulina G/inmunología , Masculino , Fosfoproteínas/inmunología , Análisis por Matrices de Proteínas , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/inmunología
9.
Microbiol Spectr ; 9(2): e0141621, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34704808

RESUMEN

The rapid worldwide spread of SARS-CoV-2 has accelerated research and development for controlling the COVID-19 pandemic. A multi-coronavirus protein microarray was created containing full-length proteins, overlapping protein fragments of various lengths, and peptide libraries from SARS-CoV-2 and four other human coronaviruses. Sera from confirmed COVID-19 patients as well as unexposed individuals were applied to multicoronavirus arrays to identify specific antibody reactivity. High-level IgG, IgM, and IgA reactivity to structural proteins S, M, and N of SARS-CoV-2, as well as accessory proteins such as ORF3a and ORF7a, were observed that were specific to COVID-19 patients. Antibody reactivity against overlapping 100-, 50-, and 30-amino acid fragments of SARS-CoV-2 proteins was used to identify antigenic regions. Numerous proteins of SARS-CoV, Middle East respiratory syndrome coronavirus (MERS-CoV), and the endemic human coronaviruses HCoV-NL63 and HCoV-OC43 were also more reactive with IgG, IgM, and IgA in COVID-19 patient sera than in unexposed control sera, providing further evidence of immunologic cross-reactivity between these viruses. Whereas unexposed individuals had minimal reactivity against SARS-CoV-2 proteins that poorly correlated with reactivity against HCoV-NL63 and HCoV-OC43 S2 and N proteins, COVID-19 patient sera had higher correlation between SARS-CoV-2 and HCoV responses, suggesting that de novo antibodies against SARS-CoV-2 cross-react with HCoV epitopes. Array responses were compared with validated spike protein-specific IgG enzyme-linked immunosorbent assays (ELISAs), showing agreement between orthologous methods. SARS-CoV-2 microneutralization titers were low in the COVID-19 patient sera but correlated with array responses against S and N proteins. The multi-coronavirus protein microarray is a useful tool for mapping antibody reactivity in COVID-19 patients. IMPORTANCE With novel mutant SARS-CoV-2 variants of concern on the rise, knowledge of immune specificities against SARS-CoV-2 proteins is increasingly important for understanding the impact of structural changes in antibody-reactive protein epitopes on naturally acquired and vaccine-induced immunity, as well as broader topics of cross-reactivity and viral evolution. A multi-coronavirus protein microarray used to map the binding of COVID-19 patient antibodies to SARS-CoV-2 proteins and protein fragments as well as to the proteins of four other coronaviruses that infect humans has shown specific regions of SARS-CoV-2 proteins that are highly reactive with patient antibodies and revealed cross-reactivity of these antibodies with other human coronaviruses. These data and the multi-coronavirus protein microarray tool will help guide further studies of the antibody response to COVID-19 and to vaccination against this worldwide pandemic.


Asunto(s)
Anticuerpos Antivirales/inmunología , Coronavirus Humano NL63/inmunología , Coronavirus Humano OC43/inmunología , Epítopos/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , SARS-CoV-2/inmunología , Anticuerpos Antivirales/sangre , Sitios de Unión de Anticuerpos/inmunología , COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Reacciones Cruzadas/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Fosfoproteínas/inmunología , Análisis por Matrices de Proteínas , Glicoproteína de la Espiga del Coronavirus/inmunología , Proteínas Virales/inmunología , Proteínas Viroporinas/inmunología
10.
J Clin Invest ; 116(7): 1963-73, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16794734

RESUMEN

To develop an animal model of Kaposi sarcoma-associated herpesvirus (KSHV) infection uniquely suited to evaluate longitudinal patterns of viral gene expression, cell tropism, and immune responses, we injected NOD/SCID mice intravenously with purified virus and measured latent and lytic viral transcripts in distal organs over the subsequent 4 months. We observed sequential escalation of first latent and then lytic KSHV gene expression coupled with electron micrographic evidence of virion production within the murine spleen. Using novel technology that integrates flow cytometry with immunofluorescence microscopy, we found that the virus establishes infection in murine B cells, macrophages, NK cells, and, to a lesser extent, dendritic cells. To investigate the potential for human KSHV-specific immune responses within this immunocompromised host, we implanted NOD/SCID mice with functional human hematopoietic tissue grafts (NOD/SCID-hu mice) and observed that a subset of animals produced human KSHV-specific antibodies. Furthermore, treatment of these chimeric mice with ganciclovir at the time of inoculation led to prolonged but reversible suppression of KSHV DNA and RNA levels, suggesting that KSHV can establish latent infection in vivo despite ongoing suppression of lytic replication.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Humano 8/metabolismo , Leucocitos/inmunología , Animales , Antígenos CD/metabolismo , Antígenos Virales/inmunología , Antivirales/farmacología , Linaje de la Célula , Ganciclovir/farmacología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/ultraestructura , Humanos , Ratones , Ratones SCID , Proteínas Nucleares/inmunología , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/virología , Bazo/citología , Bazo/virología , Trasplantes , Virión/metabolismo , Virión/ultraestructura
11.
Lab Chip ; 19(9): 1524-1533, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30806409

RESUMEN

There is a growing need to screen multiple infections simultaneously rather than diagnosis of one pathogen at a time in order to improve the quality of healthcare while saving initial screening time and reduce costs. This is the first demonstration of a five-step protein array assay for the multiplexed detection of HIV, HPV and HSV antibodies on an integrated microfluidic system. HIV, HPV and HSV reactive antibodies from both serum and saliva were rapidly detected by acoustic streaming-based mixing and pumping to enable an integrated, rapid and simple-to-use multiplexed assay device. We validated this device with 37 serum and saliva samples to verify reactivity of patient antibodies with HIV, HPV and HSV antigens. Our technology can be adapted with different protein microarrays to detect a variety of other infections, thus demonstrating a powerful platform to detect multiple putative protein biomarkers for rapid detection of infectious diseases. This integrated microfluidic protein array platform is the basis of a potent strategy to delay progression of primary infection, reduce the risk of co-infections and prevent onward transmission of infections by point-of-care detection of multiple pathogens in both serum and oral fluids.


Asunto(s)
Acústica/instrumentación , Inmunoensayo/instrumentación , Dispositivos Laboratorio en un Chip , Saliva/virología , Virosis/sangre , Virosis/diagnóstico , Humanos , Virosis/inmunología
12.
Retrovirology ; 5: 42, 2008 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-18510766

RESUMEN

AIDS-associated, CCR5-tropic (R5) HIV-1 clones, isolated from a patient that never developed CXCR4-tropic HIV-1, replicate to a greater extent and cause greater cytopathic effects than R5 HIV-1 clones isolated before the onset of AIDS. Previously, we showed that HIV-1 Env substantially contributed to the enhanced replication of an AIDS clone. In order to determine if Nef makes a similar contribution, we cloned and phenotypically analyzed nef genes from a series of patient ACH142 derived R5 HIV-1 clones. The AIDS-associated Nef contains a series of residues found in Nef proteins from progressors 1. In contrast to other reports 123, this AIDS-associated Nef downmodulated MHC-I to a greater extent and CD4 less than pre-AIDS Nef proteins. Additionally, all Nef proteins enhanced infectivity similarly in a single round of replication. Combined with our previous study, these data show that evolution of the HIV-1 env gene, but not the nef gene, within patient ACH142 significantly contributed to the enhanced replication and cytopathic effects of the AIDS-associated R5 HIV-1 clone.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Secuencia de Aminoácidos , Antígenos CD4/biosíntesis , Clonación Molecular , Efecto Citopatogénico Viral , Regulación hacia Abajo , VIH-1/genética , VIH-1/crecimiento & desarrollo , VIH-1/patogenicidad , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
13.
Mol Cell Biol ; 24(21): 9286-94, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15485898

RESUMEN

Eukaryotic cells have evolved a complex mechanism for sensing DNA damage during genome replication. Activation of this pathway prevents entry into mitosis to allow for either DNA repair or, in the event of irreparable damage, commitment to apoptosis. Under conditions of replication stress, the damage signal is initiated by the ataxia-telangiectasia-mutated and Rad3-related kinase ATR. We recently demonstrated that the human immunodeficiency virus type 1 (HIV-1) gene product viral protein R (Vpr) arrests infected cells in the G(2) phase via the activation of ATR. In the present study, we show that the activation of ATR by Vpr is analogous to activation by certain genotoxic agents, both mechanistically and in its downstream consequences. Specifically, we show a requirement for Rad17 and Hus1 to induce G(2) arrest as well as Vpr-induced phosphorylation of histone 2A variant X (H2AX) and formation of nuclear foci containing H2AX and breast cancer susceptibility protein 1. These results demonstrate that G(2) arrest mediated by the HIV-1 gene product Vpr utilizes the cellular signaling pathway whose physiological function is to recognize replication stress. These findings should contribute to a greater understanding of how HIV-1 manipulates the CD4(+)-lymphocyte cell cycle and apoptosis induction in the progressive CD4(+)-lymphocyte depletion characteristic of HIV-1 pathogenesis.


Asunto(s)
Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fase G2 , Productos del Gen vpr/metabolismo , VIH-1/fisiología , Histonas/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/genética , Antígenos CD4/metabolismo , Proteínas de Ciclo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Productos del Gen vpr/genética , Histonas/genética , Humanos , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Proteínas de Schizosaccharomyces pombe , Timo/metabolismo , Timo/patología , Proteínas Supresoras de Tumor , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
14.
Bio Protoc ; 7(20): e2584, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34595266

RESUMEN

This technique allows for efficient, highly purified cytoplasmic and nuclear-associated compartment fractionation utilizing NP-40 detergent in mammalian cells. The nuclear membrane is not disturbed during the fractionation thus leaving all nuclear and perinuclear associated components in the nuclear fraction. This protocol has been modified from Sambrook and Russell (2001) in order to downscale the amount of cells needed. To determine the efficiency of fractionation, we recommend using qPCR to compare the subcellular compartments that have been purified with equivalent amount of control whole cell extracts.

15.
Retrovirology ; 2: 63, 2005 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-16232320

RESUMEN

BACKGROUND: HIV-1 and all retroviruses are related to retroelements of simpler organisms such as the yeast Ty elements. Recent work has suggested that the yeast retroelement Ty1 replicates via an unexpected RNA lariat intermediate in cDNA synthesis. The putative genomic RNA lariat intermediate is formed by a 2'-5' phosphodiester bond, like that found in pre-mRNA intron lariats and it facilitates the minus-strand template switch during cDNA synthesis. We hypothesized that HIV-1 might also form a genomic RNA lariat and therefore that siRNA-mediated inhibition of expression of the human RNA lariat de-branching enzyme (DBR1) expression would specifically inhibit HIV-1 replication. RESULTS: We designed three short interfering RNA (siRNA) molecules targeting DBR1, which were capable of reducing DBR1 mRNA expression by 80% and did not significantly affect cell viability. We assessed HIV-1 replication in the presence of DBR1 siRNA and found that DBR1 knockdown led to decreases in viral cDNA and protein production. These effects could be reversed by cotransfection of a DBR1 cDNA indicating that the inhibition of HIV-1 replication was a specific effect of DBR1 underexpression. CONCLUSION: These data suggest that DBR1 function may be needed to debranch a putative HIV-1 genomic RNA lariat prior to completion of reverse transcription.


Asunto(s)
VIH-1/fisiología , ARN Nucleotidiltransferasas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Replicación Viral , Línea Celular , VIH-1/genética , Humanos , ARN Nucleotidiltransferasas/genética , Empalme del ARN , ARN Mensajero/análisis , Transcripción Genética , Transfección
16.
AIDS Res Hum Retroviruses ; 30(1): 37-44, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24134221

RESUMEN

The role of HIV-1-specific antibody responses in HIV disease progression is complex and would benefit from analysis techniques that examine clusterings of responses. Protein microarray platforms facilitate the simultaneous evaluation of numerous protein-specific antibody responses, though excessive data are cumbersome in analyses. Principal components analysis (PCA) reduces data dimensionality by generating fewer composite variables that maximally account for variance in a dataset. To identify clusters of antibody responses involved in disease control, we investigated the association of HIV-1-specific antibody responses by protein microarray, and assessed their association with disease progression using PCA in a nested cohort design. Associations observed among collections of antibody responses paralleled protein-specific responses. At baseline, greater antibody responses to the transmembrane glycoprotein (TM) and reverse transcriptase (RT) were associated with higher viral loads, while responses to the surface glycoprotein (SU), capsid (CA), matrix (MA), and integrase (IN) proteins were associated with lower viral loads. Over 12 months greater antibody responses were associated with smaller decreases in CD4 count (CA, MA, IN), and reduced likelihood of disease progression (CA, IN). PCA and protein microarray analyses highlighted a collection of HIV-specific antibody responses that together were associated with reduced disease progression, and may not have been identified by examining individual antibody responses. This technique may be useful to explore multifaceted host-disease interactions, such as HIV coinfections.


Asunto(s)
Progresión de la Enfermedad , Anticuerpos Anti-VIH/sangre , Inmunoglobulina G/sangre , Análisis de Componente Principal , Análisis por Matrices de Proteínas , Adulto , Recuento de Linfocito CD4 , Cápside/inmunología , Estudios Transversales , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Integrasa de VIH/inmunología , Transcriptasa Inversa del VIH/inmunología , Seropositividad para VIH , VIH-1/inmunología , Humanos , Kenia , Masculino , Glicoproteínas de Membrana/inmunología , Datos de Secuencia Molecular , Carga Viral/inmunología , Proteínas de la Matriz Viral/inmunología
18.
Proteomics Clin Appl ; 5(11-12): 613-23, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21956928

RESUMEN

INTRODUCTION: Humoral immune responses play a pivotal role in naturally acquired immunity to malaria. Understanding which humoral responses are impaired among individuals at higher risk for malaria may improve our understanding of malaria immune control and contribute to vaccine development. METHODS: We compared humoral responses with 483 Plasmodium falciparum antigens between adults in, Kisumu (high, year-long malaria transmission leading to partial immunity), and adults in Kisii (low, seasonal malaria transmission). Then within each site, we compared malaria-specific humoral responses between those at higher risk for malaria (CD4(+) ≤500) and those at lower risk for malaria (CD4(+) >500). A protein microarray chip containing 483 P. falciparum antigens and 71 HIV antigens was used. Benjamini-Hochberg adjustments were made to control for multiple comparisons. RESULTS: Fifty-seven antigens including CSP, MSP1, LSA1 and AMA1 were identified as significantly more reactive in Kisumu than in Kisii. Ten of these antigens had been identified as protective in an earlier study. CD4(+) T-cell count did not significantly impact humoral responses. CONCLUSION: Protein microarrays are a useful method to screen multiple humoral responses simultaneously. This study provides useful clues for potential vaccine candidates. Modest decreases in CD4 counts may not significantly impact malaria-specific humoral immunity.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/parasitología , Inmunidad Humoral , Plasmodium falciparum/inmunología , Adulto , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Recuento de Linfocito CD4 , Enfermedades Endémicas/prevención & control , Femenino , VIH-1 , Humanos , Kenia/epidemiología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Masculino , Plasmodium falciparum/patogenicidad , Especificidad de la Especie
19.
PLoS One ; 5(3): e9737, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20305815

RESUMEN

Defensins are antimicrobial peptides expressed by plants and animals. In mammals there are three subfamilies of defensins, distinguished by structural features: alpha, beta and theta. Alpha and beta-defensins are linear peptides with broad anti-microbial activity that are expressed by many mammals including humans. In contrast, theta-defensins are cyclic anti-microbial peptides made by several non-human primates but not humans. All three defensin types have anti-HIV-1 activity, but their mechanisms of action differ. We studied the anti-HIV-1 activity of one defensin from each group, HNP-1 (alpha), HBD-2 (beta) and RTD-1 (theta). We examined how each defensin affected HIV-1 infection and demonstrated that the cyclic defensin RTD-1 inhibited HIV-1 entry, while acyclic HNP-1 and HBD-2 inhibited HIV-1 replication even when added 12 hours post-infection and blocked viral replication after HIV-1 cDNA formation. We further found that all three defensins downmodulated CXCR4. Moreover, RTD-1 inactivated X4 HIV-1, while HNP-1 and HBD-2 inactivated both X4 and R5 HIV-1. The data presented here show that acyclic and cyclic defensins block HIV-1 replication by shared and diverse mechanisms. Moreover, we found that HNP-1 and RTD-1 directly inhibited firefly luciferase enzymatic activity, which may affect the interpretation of previously published data.


Asunto(s)
Fármacos Anti-VIH/farmacología , Defensinas/fisiología , VIH-1/metabolismo , Replicación Viral , Animales , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Humanos , Leucocitos Mononucleares/metabolismo , Luciferasas/metabolismo , Neutrófilos/metabolismo , Péptidos Cíclicos/metabolismo , Receptores CXCR4/metabolismo , alfa-Defensinas/metabolismo , beta-Defensinas/metabolismo
20.
J Virol ; 81(16): 8838-42, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17537849

RESUMEN

Long-term asymptomatic human immunodeficiency virus (HIV)-infected individuals (LTA) usually have low viral load and low immune activation. To discern whether viral load or immune activation is dominant in determining progression to AIDS, we studied three exceptional LTA with high viral loads. HIV type 1 isolates from these LTA were as pathogenic as viruses from progressors in organ culture. Despite high viral loads, these LTA had low levels of proliferating and activated T cells compared to progressors, like other LTA. In contrast to those in progressors, HIV-specific CD4(+) T-cell responses in these LTA were maintained. Thus, low immune activation despite a high viral load preserved HIV-specific T-cell responses and resulted in a long-term asymptomatic phenotype.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/diagnóstico , Síndrome de Inmunodeficiencia Adquirida/inmunología , Linfocitos T CD4-Positivos/inmunología , VIH-1/inmunología , VIH-1/aislamiento & purificación , Humanos , Activación de Linfocitos , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA