Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.757
Filtrar
1.
Methods Mol Biol ; 2807: 261-270, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743234

RESUMEN

The development of 3D-organoid models has revolutionized the way diseases are studied. Recently, our brain organoid model has been shown to recapitulate in in vitro the human brain cytoarchitecture originally encountered in HIV-1 neuropathogenesis, allowing downstream applications. Infected monocytes, macrophages, and microglia are critically important immune cells for infection and dissemination of HIV-1 throughout brain during acute and chronic phase of the disease. Once in the brain parenchyma, long-lived infected monocytes/macrophages along with resident microglia contribute to the establishment of CNS latency in people with HIV (PWH). Hence, it is important to better understand how HIV-1 enters and establishes infection and latency in CNS to further develop cure strategies. Here we detailed an accessible protocol to incorporate monocytes (infected and/or labeled) as a model of transmigration of peripheral monocytes into brain organoids that can be applied to characterize HIV-1 neuroinvasion and virus dissemination.


Asunto(s)
Encéfalo , Infecciones por VIH , VIH-1 , Monocitos , Organoides , Organoides/virología , Organoides/patología , Humanos , VIH-1/fisiología , VIH-1/patogenicidad , Monocitos/virología , Monocitos/inmunología , Infecciones por VIH/virología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Encéfalo/virología , Encéfalo/patología , Encéfalo/inmunología , Microglía/virología , Microglía/inmunología , Microglía/patología , Macrófagos/virología , Macrófagos/inmunología , Latencia del Virus
2.
Circ Res ; 134(11): e150-e175, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38781298

RESUMEN

HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.


Asunto(s)
Infecciones por VIH , Hipertensión , Humanos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología , Infecciones por VIH/complicaciones , Hipertensión/tratamiento farmacológico , Hipertensión/epidemiología , Factores de Riesgo , VIH-1/patogenicidad , Animales
3.
Viruses ; 15(12)2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38140631

RESUMEN

Human interferon-induced transmembrane (IFITM) proteins inhibit the fusion of a broad spectrum of enveloped viruses, both when expressed in target cells and when present in infected cells. Upon expression in infected cells, IFITMs incorporate into progeny virions and reduce their infectivity by a poorly understood mechanism. Since only a few envelope glycoproteins (Envs) are present on HIV-1 particles, and Env clustering has been proposed to be essential for optimal infectivity, we asked if IFITM protein incorporation modulates HIV-1 Env clustering. The incorporation of two members of the IFITM family, IFITM1 and IFITM3, into HIV-1 pseudoviruses correlated with a marked reduction of infectivity. Super-resolution imaging of Env distribution on single HIV-1 pseudoviruses did not reveal significant effects of IFITMs on Env clustering. However, IFITM3 reduced the Env processing and incorporation into virions relative to the control and IFITM1-containing viruses. These results show that, in addition to interfering with the Env function, IFITM3 restricts HIV-1 Env cleavage and incorporation into virions. The lack of notable effect of IFITMs on Env clustering supports alternative restriction mechanisms, such as modification of the properties of the viral membrane.


Asunto(s)
Antígenos de Diferenciación , VIH-1 , Proteínas de la Membrana , Internalización del Virus , Humanos , Genes env , Glicoproteínas/metabolismo , VIH-1/patogenicidad , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Antígenos de Diferenciación/metabolismo
4.
J Cell Biol ; 222(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36988579

RESUMEN

Macrophages are essential for HIV-1 pathogenesis and represent major viral reservoirs. Therefore, it is critical to understand macrophage infection, especially in tissue macrophages, which are widely infected in vivo, but poorly permissive to cell-free infection. Although cell-to-cell transmission of HIV-1 is a determinant mode of macrophage infection in vivo, how HIV-1 transfers toward macrophages remains elusive. Here, we demonstrate that fusion of infected CD4+ T lymphocytes with human macrophages leads to their efficient and productive infection. Importantly, several tissue macrophage populations undergo this heterotypic cell fusion, including synovial, placental, lung alveolar, and tonsil macrophages. We also find that this mode of infection is modulated by the macrophage polarization state. This fusion process engages a specific short-lived adhesion structure and is controlled by the CD81 tetraspanin, which activates RhoA/ROCK-dependent actomyosin contractility in macrophages. Our study provides important insights into the mechanisms underlying infection of tissue-resident macrophages, and establishment of persistent cellular reservoirs in patients.


Asunto(s)
Linfocitos T CD4-Positivos , Fusión Celular , Infecciones por VIH , Macrófagos , Humanos , Linfocitos T CD4-Positivos/metabolismo , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Macrófagos/metabolismo , Macrófagos/virología , Actomiosina/metabolismo
5.
J Virol ; 97(4): e0020023, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36971578

RESUMEN

Tetherin prevents viral cross-species transmission by inhibiting the release of multiple enveloped viruses from infected cells. With the evolution of simian immunodeficiency virus of chimpanzees (SIVcpz), a pandemic human immunodeficiency virus type 1 (HIV-1) precursor, its Vpu protein can antagonize human tetherin (hTetherin). Macaca leonina (northern pig-tailed macaque [NPM]) is susceptible to HIV-1, but host-specific restriction factors limit virus replication in vivo. In this study, we isolated the virus from NPMs infected with strain stHIV-1sv (with a macaque-adapted HIV-1 env gene from simian-human immunodeficiency virus SHIV-KB9, a vif gene replaced by SIVmac239, and other genes originating from HIV-1NL4.3) and found that a single acidic amino acid substitution (G53D) in Vpu could increase its ability to degrade the tetherin of macaques (mTetherin) mainly through the proteasome pathway, resulting in an enhanced release and resistance to interferon inhibition of the mutant stHIV-1sv strain, with no influence on the other functions of Vpu. IMPORTANCE HIV-1 has obvious host specificity, which has greatly hindered the construction of animal models and severely restricted the development of HIV-1 vaccines and drugs. To overcome this barrier, we attempted to isolate the virus from NPMs infected with stHIV-1sv, search for a strain with an adaptive mutation in NPMs, and develop a more appropriate nonhuman primate model of HIV-1. This is the first report identifying HIV-1 adaptations in NPMs. It suggests that while tetherin may limit HIV-1 cross-species transmission, the Vpu protein in HIV-1 can overcome this species barrier through adaptive mutation, increasing viral replication in the new host. This finding will be beneficial to building an appropriate animal model for HIV-1 infection and promoting the development of HIV-1 vaccines and drugs.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea , VIH-1 , Macaca , Proteínas Virales , Liberación del Virus , VIH-1/genética , VIH-1/patogenicidad , Proteínas Virales/genética , Proteínas Virales/metabolismo , Mutación , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Ubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Liberación del Virus/genética , Sustitución de Aminoácidos/genética , Infecciones por VIH/virología , Modelos Animales de Enfermedad , Replicación Viral/genética
6.
J Biol Chem ; 298(4): 101805, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35259395

RESUMEN

HIV-1 encodes accessory proteins that neutralize antiviral restriction factors to ensure its successful replication. One accessory protein, the HIV-1 viral infectivity factor (Vif), is known to promote ubiquitination and proteasomal degradation of the antiviral restriction factor apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like 3G (APOBEC3G), a cytosine deaminase that leads to hypermutations in the viral DNA and subsequent aberrant viral replication. We have previously demonstrated that the HIV-1 viral transcription mediator Tat activates the host progrowth PI-3-AKT pathway, which in turn promotes HIV-1 replication. Because the HIV-1 Vif protein contains the putative AKT phosphorylation motif RMRINT, here we investigated whether AKT directly phosphorylates HIV-1 Vif to regulate its function. Coimmunoprecipitation experiments showed that AKT and Vif interact with each other, supporting this hypothesis. Using in vitro kinase assays, we further showed that AKT phosphorylates Vif at threonine 20, which promotes its stability, as Vif becomes destabilized after this residue is mutated to alanine. Moreover, expression of dominant-negative kinase-deficient AKT as well as treatment with a chemical inhibitor of AKT increased K48-ubiquitination and proteasomal degradation of HIV-1 Vif. In contrast, constitutively active AKT (Myr-AKT) reduced K48-ubiquitination of Vif to promote its stability. Finally, inhibition of AKT function restored APOBEC3G levels, which subsequently reduced HIV-1 infectivity. Thus, our results establish a novel mechanism of HIV-1 Vif stabilization through AKT-mediated phosphorylation at threonine 20, which reduces APOBEC3G levels and potentiates HIV-1 infectivity.


Asunto(s)
Desaminasa APOBEC-3G , Infecciones por VIH , VIH-1 , Productos del Gen vif del Virus de la Inmunodeficiencia Humana , Desaminasa APOBEC-3G/genética , Desaminasa APOBEC-3G/metabolismo , Infecciones por VIH/fisiopatología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Fosforilación , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Treonina/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
7.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216446

RESUMEN

Human hematopoietic stem/progenitor cell (HSPC)-based gene therapy is a promising direction for curing HIV-1-infected individuals. The zinc finger protein (2LTRZFP) designed to target the 2-LTR-circle junction of HIV-1 cDNA was previously reported as an intracellular antiviral molecular scaffold that prevents HIV integration. Here, we elucidate the efficacy and safety of using 2LTRZFP in human CD34+ HSPCs. We transduced 2LTRZFP which has the mCherry tag (2LTRZFPmCherry) into human CD34+ HSPCs using a lentiviral vector. The 2LTRZFPmCherry-transduced HSPCs were subsequently differentiated into macrophages. The expression levels of pro-apoptotic proteins of the 2LTRZFPmCherry-transduced HSPCs showed no significant difference from those of the non-transduced control. Furthermore, the 2LTRZFPmCherry-transduced HSPCs were successfully differentiated into mature macrophages, which had normal phagocytic function. The cytokine secretion assay demonstrated that 2LTRZFPmCherry-transduced CD34+ derived macrophages promoted the polarization towards classically activated (M1) subtypes. More importantly, the 2LTRZFPmCherry transduced cells significantly exhibited resistance to HIV-1 integration in vitro. Our findings demonstrate that the 2LTRZFPmCherry-transduced macrophages were found to be functionally and phenotypically normal, with no adverse effects of the anti-HIV-1 scaffold. Our data suggest that the anti-HIV-1 integrase scaffold is a promising antiviral molecule that could be applied to human CD34+ HSPC-based gene therapy for AIDS patients.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Células Madre Hematopoyéticas/metabolismo , Macrófagos/metabolismo , Células Madre/metabolismo , Dedos de Zinc/fisiología , Antígenos CD34/metabolismo , Terapia Genética/métodos , Humanos
8.
Viruses ; 14(1)2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35062355

RESUMEN

So far, only two retroviruses, human immunodeficiency virus (HIV) (type 1 and 2) and human T-cell lymphotropic virus type 1 (HTLV-1), have been recognized as pathogenic for humans. Both viruses mainly infect CD4+ T lymphocytes. HIV replication induces the apoptosis of CD4 lymphocytes, leading to the development of acquired immunodeficiency syndrome (AIDS). After a long clinical latency period, HTLV-1 can transform lymphocytes, with subsequent uncontrolled proliferation and the manifestation of a disease called adult T-cell leukemia (ATLL). Certain infected patients develop neurological autoimmune disorder called HTLV-1-associated myelopathy, also known as tropical spastic paraparesis (HAM/TSP). Both viruses are transmitted between individuals via blood transfusion, tissue/organ transplantation, breastfeeding, and sexual intercourse. Within the host, these viruses can spread utilizing either cell-free or cell-to-cell modes of transmission. In this review, we discuss the mechanisms and importance of each mode of transmission for the biology of HIV-1 and HTLV-1.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/transmisión , VIH-1/patogenicidad , Infecciones por HTLV-I/transmisión , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Leucemia-Linfoma de Células T del Adulto/virología , Animales , Linfocitos T CD4-Positivos/inmunología , Infecciones por HTLV-I/complicaciones , Humanos , Ratones
9.
PLoS One ; 17(1): e0262454, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35025927

RESUMEN

BACKGROUND: People living with HIV (PLHIV) co-infected with tuberculosis (TB) have a distinct clinical presentation and poorer treatment outcomes compared to HIV-seronegative TB patients. Excluding low CD4 count, innate immune factors associated with TB are not fully elucidated. We, therefore, characterised and compared the expression of IL-6, TNF-α, IFN-γ, and IL-10 in whole blood of treatment naïve TB patients stimulated with heat-killed Mycobacterium tuberculosis stratified by HIV status and the level of CD4 count. RESULTS: We recruited 39 HIV seropositive and 31 HIV seronegative TB patients. Median (IQR) age was 35(28-42) years and 31(25-36) years respectively, and a majority had pulmonary tuberculosis i.e. 38(95%) and 30(97%), respectively. The two groups were significantly different in the distribution of CD4 count, 563 [465-702.5 cells/mm3] vs 345 [157-483 cell/mm3] in HIV negative vs HIV positive respectively p = <0.001. Post stimulation, the expression of IL-6 in HIV negative TB patients was significantly higher than in the HIV positive 16,757366 [8,827-23,686 pg/ml] vs. 9,508 [5,514-15,008 pg/ml], respectively; p = 0.0360. TNF-α and IFN-γ were highly expressed in HIV negative TB patients compared to the HIV positive though not statistically significant. We only observed higher expression of IL-6 in HIV negative patients in comparison to the HIV positive when stratified by level of CD4 counts as < 500 and ≥ 500 cell/mm3 for both cohorts. 21,953 [8,990-24,206 pg/ml] vs 9,505 [5,400-15,313 pg/ml], p value = 0.0585 in patients with CD4 count < 500 cell/mm3 and 13,168 [7,087-22,584 pg/ml] vs 10,413 [7,397-14,806 pg/ml], p value = 0.3744 for patients with CD4 count of ≥ 500 cell/mm3 respectively. We found a positive pairwise correlation between TNF-α -alpha and IL-6 in both HIV positive and HIV negative patients, r = 0.61 (95% CI 0.36-0.72; p < 0.0001) and r = 0.48 (95% CI 0.15-0.68; p = 0.005) respectively. The IFNγ/IL-10 ratio was higher in HIV negative when compared to HIV positive individuals, 0.052 [0.0-0.28] vs 0.007 [0-0.32] respectively; p = 0.05759. IL-6 independently reduced the probability of TB/HIV, Adjusted odds ratio 0.99, p value 0.007. CONCLUSIONS: This study suggests that HIV seronegative TB patients have a higher pro-inflammatory response to MTB than HIV seropositive TB patients. Further, it also shows that the level of CD4 influences immunomodulation. The findings suggest that the difference in cytokine expression may be responsible for the distinct patterns of TB presentation between HIV positive and HIV negative patient.


Asunto(s)
Infecciones por VIH/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Adulto , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/metabolismo , Coinfección/complicaciones , Estudios Transversales , Femenino , Infecciones por VIH/complicaciones , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Interferón gamma/sangre , Interferón gamma/metabolismo , Interleucina-10/sangre , Interleucina-10/metabolismo , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/complicaciones , Tuberculosis Pulmonar/complicaciones , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Zambia/epidemiología
10.
PLoS One ; 17(1): e0262477, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35085286

RESUMEN

A transframe region within HIV-1 Gag-Pol (referred to as p6* or p6pol), directly linked to the protease (PR) N-terminus, plays a pivotal role in modulating PR activation. To identify specific p6* residues involved in PR activation, we created a series of p6* mutants by making substitutions for conserved p6* residues. Our results indicate that some p6* mutants were defective in terms of virus infectivity, despite displaying a wild-type virus particle processing pattern. Mutations at p6* F8 reduced virus infectivity associated with insufficient virus processing, due in part to impaired PR maturation and RT packaging. Our data strongly suggest that conserved Phe (F) residues at position 8 of p6* are involved in the PR maturation process.


Asunto(s)
Sustitución de Aminoácidos/genética , VIH-1/genética , VIH-1/patogenicidad , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Proteínas de Fusión gag-pol/genética , Células HEK293 , Proteasa del VIH/genética , Seropositividad para VIH/genética , Células HeLa , Humanos , Mutación/genética , Virión/genética , Replicación Viral/genética
11.
Medicine (Baltimore) ; 100(51): e28328, 2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-34941134

RESUMEN

ABSTRACT: Pulmonary Kaposi sarcoma (pKS) caused by Human herpesvirus 8 (HHV-8) is a devastating form of KS in patients with advanced acquired immunodeficiency syndrome (AIDS) and is associated with increased morbidity and mortality. Blood T cells play a central role in the response of HIV-1 and HHV-8. However, little information is available on T cells in the alveolar space of HIV-1-associated pKS patients.Therefore, we examined CD8+ and CD4+ T cells in the alveolar space in comparison with the blood of patients with pKS. We recruited 26 HIV-1 positive patients with KS, including 15 patients with pKS. Bronchoalveolar lavage (BAL) cells and blood mononuclear cells were analyzed for T cell memory phenotypes, surface markers associated with exhaustion, and intracellular cytokine staining (ICS) using flow cytometry. HIV-1 and HHV-8 viral loads were measured in plasma by quantitative PCR.BAL T cells showed reduced inflammatory capacities and significantly diminished polyfunctionality compared to blood T cells from patients with pKS. This was not accompanied by increased expression of exhaustion markers, such as TIM-3 and PD-1.More importantly, we found a negative correlation between the production of MIP1-ß and TNF-α in T cells in BAL and blood, indicating compartmentalised immune responses to pKS and accentuated chronic HIV-1/HHV-8 pathogenesis via T cells in the lungs of people with pKS.


Asunto(s)
Infecciones Oportunistas Relacionadas con el SIDA/virología , Líquido del Lavado Bronquioalveolar/virología , Seropositividad para VIH/complicaciones , Herpesvirus Humano 8/inmunología , Neoplasias Pulmonares/virología , Sarcoma de Kaposi/virología , Linfocitos T Reguladores/inmunología , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , VIH-1/patogenicidad , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/aislamiento & purificación , Humanos , Reacción en Cadena de la Polimerasa
12.
Viruses ; 13(12)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34960728

RESUMEN

Staufen, the RNA-binding family of proteins, affects various steps in the Human Immuno-Deficiency Virus (HIV-1) replication cycle. While our previous study established Staufen-2-HIV-1 Rev interaction and its role in augmenting nucleocytoplasmic export of RRE-containing viral RNA, viral incorporation of Staufen-2 and its effect on viral propagation were unknown. Here, we report that Staufen-2 interacts with HIV-1 Gag and is incorporated into virions and that encapsidated Staufen-2 boosted viral infectivity. Further, Staufen-2 gets co-packaged into virions, possibly by interacting with host factors Staufen-1 or antiviral protein APOBEC3G, which resulted in different outcomes on the infectivity of Staufen-2-encapsidated virions. These observations suggest that encapsidated host factors influence viral population dynamics and infectivity. With the explicit identification of the incorporation of Staufen proteins into HIV-1 and other retroviruses, such as Simian Immunodeficiency Virus (SIV), we propose that packaging of RNA binding proteins, such as Staufen, in budding virions of retroviruses is probably a general phenomenon that can drive or impact the viral population dynamics, infectivity, and evolution.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1/fisiología , VIH-1/patogenicidad , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Empaquetamiento del Genoma Viral , Línea Celular , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Proteínas del Tejido Nervioso/genética , Unión Proteica , Proteínas de Unión al ARN/genética , Virulencia
13.
Int J Mol Sci ; 22(21)2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34769038

RESUMEN

Tetraspanins are transmembrane glycoproteins that have been shown increasing interest as host factors in infectious diseases. In particular, they were implicated in the pathogenesis of both non-enveloped (human papillomavirus (HPV)) and enveloped (human immunodeficiency virus (HIV), Zika, influenza A virus, (IAV), and coronavirus) viruses through multiple stages of infection, from the initial cell membrane attachment to the syncytium formation and viral particle release. However, the mechanisms by which different tetraspanins mediate their effects vary. This review aimed to compare and contrast the role of tetraspanins in the life cycles of HPV, HIV, Zika, IAV, and coronavirus viruses, which cause the most significant health and economic burdens to society. In doing so, a better understanding of the relative contribution of tetraspanins in virus infection will allow for a more targeted approach in the treatment of these diseases.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Tetraspaninas/fisiología , Virosis/metabolismo , Regulación Viral de la Expresión Génica , VIH-1/patogenicidad , Humanos , Virus de la Influenza A/patogenicidad , Papillomaviridae/patogenicidad , SARS-CoV-2/patogenicidad , Virosis/genética , Virosis/virología , Internalización del Virus , Virus Zika/patogenicidad
14.
Viruses ; 13(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34835027

RESUMEN

Paradigm shifts throughout the history of microbiology have typically been ignored, or met with skepticism and resistance, by the scientific community. This has been especially true in the field of virology, where the discovery of a "contagium vivum fluidum", or infectious fluid remaining after excluding bacteria by filtration, was initially ignored because it did not coincide with the established view of microorganisms. Subsequent studies on such infectious agents, eventually termed "viruses", were met with skepticism. However, after an abundance of proof accumulated, viruses were eventually acknowledged as defined microbiological entities. Next, the proposed role of viruses in oncogenesis in animals was disputed, as was the unique mechanism of genome replication by reverse transcription of RNA by the retroviruses. This same pattern of skepticism holds true for the prediction of the existence of retroviral "antisense" transcripts and genes. From the time of their discovery, it was thought that retroviruses encoded proteins on only one strand of proviral DNA. However, in 1988, it was predicted that human immunodeficiency virus type 1 (HIV-1), and other retroviruses, express an antisense protein encoded on the DNA strand opposite that encoding the known viral proteins. Confirmation came quickly with the characterization of the antisense protein, HBZ, of the human T-cell leukemia virus type 1 (HTLV-1), and the finding that both the protein and its antisense mRNA transcript play key roles in viral replication and pathogenesis. However, acceptance of the existence, and potential importance, of a corresponding antisense transcript and protein (ASP) in HIV-1 infection and pathogenesis has lagged, despite gradually accumulating theoretical and experimental evidence. The most striking theoretical evidence is the finding that asp is highly conserved in group M viruses and correlates exclusively with subtypes, or clades, responsible for the AIDS pandemic. This review outlines the history of the major shifts in thought pertaining to the nature and characteristics of viruses, and in particular retroviruses, and details the development of the hypothesis that retroviral antisense transcripts and genes exist. We conclude that there is a need to accelerate studies on ASP, and its transcript(s), with the view that both may be important, and overlooked, targets in anti-HIV therapeutic and vaccine strategies.


Asunto(s)
ARN sin Sentido/genética , ARN Mensajero/genética , Proteínas de los Retroviridae/genética , Retroviridae/genética , Carcinogénesis/genética , Genoma Viral , VIH-1/genética , VIH-1/patogenicidad , VIH-1/fisiología , Historia del Siglo XX , Historia del Siglo XXI , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Sistemas de Lectura Abierta , Retroviridae/patogenicidad , Retroviridae/fisiología , Transcripción Genética , Proteínas del Envoltorio Viral/genética , Virología/historia , Replicación Viral
15.
Sci Rep ; 11(1): 22901, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824340

RESUMEN

Macrophages serve as viral reservoirs due to their resistance to apoptosis and HIV-cytopathic effects. We have previously shown that inhibitor of apoptosis proteins (IAPs) confer resistance to HIV-Vpr-induced apoptosis in normal macrophages. Herein, we show that second mitochondrial activator of caspases (SMAC) mimetics (SM) induce apoptosis of monocyte-derived macrophages (MDMs) infected in vitro with a R5-tropic laboratory strain expressing heat stable antigen, chronically infected U1 cells, and ex-vivo derived MDMs from HIV-infected individuals. To understand the mechanism governing SM-induced cell death, we show that SM-induced cell death of primary HIV-infected macrophages was independent of the acquisition of M1 phenotype following HIV infection of macrophages. Instead, SM-induced cell death was found to be mediated by IAPs as downregulation of IAPs by siRNAs induced cell death of HIV-infected macrophages. Moreover, HIV infection caused receptor interacting protein kinase-1 (RIPK1) degradation which in concert with IAP1/2 downregulation following SM treatment may result in apoptosis of macrophages. Altogether, our results show that SM selectively induce apoptosis in primary human macrophages infected in vitro with HIV possibly through RIPK1. Moreover, modulation of the IAP pathways may be a potential strategy for selective killing of HIV-infected macrophages in vivo.


Asunto(s)
Fármacos Anti-VIH/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , VIH-1/patogenicidad , Macrófagos/efectos de los fármacos , Imitación Molecular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Efecto Citopatogénico Viral , Infecciones por VIH/enzimología , Infecciones por VIH/patología , Infecciones por VIH/virología , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Macrófagos/enzimología , Macrófagos/patología , Macrófagos/virología , Fenotipo , Células U937 , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
Cell Rep Med ; 2(10): 100412, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34755132

RESUMEN

In humans, pre-existing anti-HIV-1 neutralizing antibodies (nAbs) have not been associated with decreased HIV-1 acquisition. Here, we evaluate antibody-dependent cellular cytotoxicity (ADCC) present in pre-transmission infant and maternal plasma and breast milk (BM) against the contemporaneous maternal HIV-1 variants. HIV-1-exposed uninfected compared with HIV-1-exposed infected infants have higher ADCC and a combination of ADCC and nAb responses against their corresponding mother's strains. ADCC does not correlate with nAbs, suggesting they are independent activities. The infected infants with high ADCC compared with low ADCC, but not those with higher ADCC plus nAbs, have lower morbidity up to 1 year after birth. A higher IgA to IgG ratio, observed in BM supernatants and in a higher proportion of the infected compared with the uninfected infants, associates with lower ADCC. Against the exposure strains, ADCC, more than nAbs, associates with both lower mother-to-child transmission and decreased post-infection infant morbidity.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Citotoxicidad Celular Dependiente de Anticuerpos , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/inmunología , VIH-1/inmunología , Adulto , Citotoxicidad Inmunológica , Femenino , Infecciones por VIH/genética , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Sueros Inmunes/química , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Lactante , Transmisión Vertical de Enfermedad Infecciosa , Leche Humana/química , Leche Humana/inmunología , Embarazo
17.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34686593

RESUMEN

Many viruses directly engage and require the dynein-dynactin motor-adaptor complex in order to transport along microtubules (MTs) to the nucleus and initiate infection. HIV type 1 (HIV-1) exploits dynein, the dynein adaptor BICD2, and core dynactin subunits but unlike several other viruses, does not require dynactin-1 (DCTN1). The underlying reason for HIV-1's variant dynein engagement strategy and independence from DCTN1 remains unknown. Here, we reveal that DCTN1 actually inhibits early HIV-1 infection by interfering with the ability of viral cores to interact with critical host cofactors. Specifically, DCTN1 competes for binding to HIV-1 particles with cytoplasmic linker protein 170 (CLIP170), one of several MT plus-end tracking proteins (+TIPs) that regulate the stability of viral cores after entry into the cell. Outside of its function as a dynactin subunit, DCTN1 also functions as a +TIP that we find sequesters CLIP170 from incoming particles. Deletion of the Zinc knuckle (Zn) domain in CLIP170 that mediates its interactions with several proteins, including DCTN1, increased CLIP170 binding to virus particles but failed to promote infection, further suggesting that DCTN1 blocks a critical proviral function of CLIP170 mediated by its Zn domain. Our findings suggest that the unique manner in which HIV-1 binds and exploits +TIPs to regulate particle stability leaves them vulnerable to the negative effects of DCTN1 on +TIP availability and function, which may in turn have driven HIV-1 to evolve away from DCTN1 in favor of BICD2-based engagement of dynein during early infection.


Asunto(s)
Complejo Dinactina/fisiología , Infecciones por VIH/fisiopatología , Infecciones por VIH/virología , VIH-1/fisiología , Interacciones Microbiota-Huesped/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas de Neoplasias/fisiología , Unión Competitiva , Línea Celular , Complejo Dinactina/antagonistas & inhibidores , Complejo Dinactina/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , VIH-1/patogenicidad , Células HeLa , Humanos , Células Jurkat , Microglía/virología , Proteínas Asociadas a Microtúbulos/química , Modelos Biológicos , Proteínas de Neoplasias/química , Dominios Proteicos , ARN Interferente Pequeño/genética
18.
Turk J Med Sci ; 51(6): 3017-3021, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34688245

RESUMEN

Background/aim: Human HIV-1 TAT interactive protein 2 (HTATIP2/TIP30) is a gene that is extensively expressed in human tissues as well as in tumor tissues. This study aimed to explore the potential role of HTATIP2/TIP30 in contact dermatitis (CD), which is one of the most common inflammatory cutaneous conditions. Materials and methods: This cross-sectional study involved adult patients with acute contact dermatitis who were admitted to the outpatient dermatology clinic of a tertiary hospital and healthy adult volunteers without any cutaneous or systemic diseases. The blood concentration of HTATIP2/TIP30 was measured using ELISA kits. Results: The research sample consisted of 31 patients with CD (18 males, 13 females) and 20 healthy control subjects (14 males, 6 females). The mean ages of the patients with CD and healthy volunteers were 37 and 30 years, respectively (p > 0.05). The mean value of serum HTATIP2/TIP30 levels in patients with CD was 1.65 ng ml­1, which is 0.60 ng ml­1 in the control group (p = 0.02) Conclusion: In this study, serum levels of HTATIP2/TIP30 were statistically significantly higher in patients with CD when compared to healthy controls. This outcome may indicate possible role of HTATIP2/TIP30 in the pathogenesis of CD.


Asunto(s)
Acetiltransferasas/sangre , Biomarcadores de Tumor/sangre , Dermatitis por Contacto/sangre , VIH-1 , Factores de Transcripción/sangre , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Estudios Transversales , Dermatitis por Contacto/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , VIH-1/metabolismo , VIH-1/patogenicidad , Humanos , Masculino , Persona de Mediana Edad , Proteína 1 Supresora de la Señalización de Citocinas
19.
Retrovirology ; 18(1): 26, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530855

RESUMEN

BACKGROUND: Extracellular Vesicles (EV) recently have been implicated in the pathogenesis of HIV-1 syndromes, including neuroinflammation and HIV-1 associated neurological disorder (HAND). Cocaine, an illicit stimulant drug used worldwide is known to exacerbate these HIV-1 associated neurological syndromes. However, the effects of cocaine on EV biogenesis and roles of EVs in enhancing HIV-1 pathogenesis are not yet well defined. RESULTS: Here, we investigated the effects of cocaine on EV biogenesis and release in HIV-1 infected immune cells and explored their roles in elicitation of neuroinflammation. We found that cocaine significantly augmented the release of EVs from uninfected and HIV-1 infected T-cells, DCs and macrophages. Further analysis of the molecular components of EVs revealed enhanced expression of adhesion molecules integrin ß1 and LFA-1 in those EVs derived from cocaine treated cells. Intriguingly, in EVs derived from HIV-1 infected cells, cocaine treatment significantly increased the levels of viral genes in EVs released from macrophages and DCs, but not in T-cells. Exploring the molecular mechanism to account for this, we found that DCs and macrophages showed enhanced expression of the cocaine receptor Sigma 1-Receptor compared to T-cells. In addition, we found that cocaine significantly altered the integrity of the RNA-induced silencing complex (RISC) in HIV-1 infected macrophages and DCs compared to untreated HIV-1 infected cells. Characterizing further the molecular mechanisms involved in how cocaine increased EV release, we found that cocaine decreased the expression of the interferon-inducible protein BST-2; this resulted in altered trafficking of intracellular virus containing vesicles and EV biogenesis and release. We also observed EVs released from cocaine treated HIV-1 infected macrophages and DCs enhanced HIV-1 trans-infection to T-cells compared to those from untreated and HIV-1 infected cells. These EVs triggered release of proinflammatory cytokines in human brain microvascular endothelial cells (HBMECs) and altered monolayer integrity. CONCLUSIONS: Taken together, our results provide a novel mechanism which helps to elucidate the enhanced prevalence of neurological disorders in cocaine using HIV-1 infected individuals and offers insights into developing novel therapeutic strategies against HAND in these hosts.


Asunto(s)
Cocaína/efectos adversos , Cocaína/inmunología , Células Dendríticas/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , VIH-1/inmunología , Macrófagos/efectos de los fármacos , Enfermedades Neuroinflamatorias/complicaciones , Encéfalo/citología , Cocaína/farmacología , Citocinas/inmunología , Células Dendríticas/virología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/virología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/virología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Inflamación , Macrófagos/inmunología , Macrófagos/virología , Biogénesis de Organelos
20.
Cells ; 10(9)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34571832

RESUMEN

HIV enters the CNS early after peripheral infection, establishing reservoirs in perivascular macrophages that contribute to development of HIV-associated neurocognitive disorders (HAND) in 15-40% of people with HIV (PWH) despite effective antiretroviral therapy (ART). Opioid use may contribute to dysregulated macrophage functions resulting in more severe neurocognitive symptoms in PWH taking opioids. Macroautophagy helps maintain quality control in long-lived cell types, such as macrophages, and has been shown to regulate, in part, some macrophage functions in the CNS that contribute to HAND. Using Western blotting and confocal immunofluorescence in primary human macrophages, we demonstrated that morphine and a commonly prescribed ART regimen induce bulk autophagy. Morphine and ART also inhibited completion of autophagy. HIV infection increased these inhibitory effects. We also examined two types of selective autophagy that degrade aggregated proteins (aggrephagy) and dysfunctional mitochondria (mitophagy). Morphine and ART inhibited selective autophagy mediated by p62 regardless of HIV infection, and morphine inhibited mitophagic flux in HIV-infected cells demonstrating potential mitotoxicity. These results indicate that inhibition of autophagy, both in bulk and selective, in CNS macrophages may mediate neurocognitive dysfunction in PWH using opioids. Increasing autophagic activity in the context of HIV may represent a novel therapeutic strategy for reducing HAND in these individuals.


Asunto(s)
Antirretrovirales/farmacología , Autofagia/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Morfina/farmacología , Trastornos Neurocognitivos/tratamiento farmacológico , Células Cultivadas , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , Mitofagia/efectos de los fármacos , Trastornos Neurocognitivos/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA