Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Med Microbiol Immunol ; 211(2-3): 79-103, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35235048

RESUMEN

An ongoing pandemic of newly emerged SARS-CoV-2 has puzzled many scientists and health care policymakers around the globe. The appearance of the virus was accompanied by several distinct antigenic changes, specifically spike protein which is a key element for host cell entry of virus and major target of currently developing vaccines. Some of these mutations enable the virus to attach to receptors more firmly and easily. Moreover, a growing number of trials are demonstrating higher transmissibility and, in some of them, potentially more serious forms of illness related to novel variants. Some of these lineages, especially the Beta variant of concern, were reported to diminish the neutralizing activity of monoclonal and polyclonal antibodies present in both convalescent and vaccine sera. This could imply that these independently emerged variants could make antiviral strategies prone to serious threats. The rapid changes in the mutational profile of new clades, especially escape mutations, suggest the convergent evolution of the virus due to immune pressure. Nevertheless, great international efforts have been dedicated to producing efficacious vaccines with cutting-edge technologies. Despite the partial decrease in vaccines efficacy against worrisome clades, most current vaccines are still effective at preventing mild to severe forms of disease and hospital admission or death due to coronavirus disease 2019 (COVID-19). Here, we summarize existing evidence about newly emerged variants of SARS-CoV-2 and, notably, how well vaccines work against targeting new variants and modifications of highly flexible mRNA vaccines that might be required in the future.


Asunto(s)
COVID-19 , Vacunas , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Eficacia de las Vacunas
2.
Viruses ; 14(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35336957

RESUMEN

Despite the success of combinational antiretroviral therapy (cART), the high pervasiveness of human immunodeficiency virus-1 (HIV)-associated neurocognitive disorders (HAND) poses a significant challenge for society. Methamphetamine (meth) and related amphetamine compounds, which are potent psychostimulants, are among the most commonly used illicit drugs. Intriguingly, HIV-infected individuals who are meth users have a comparatively higher rate of neuropsychological impairment and exhibit a higher viral load in the brain than infected individuals who do not abuse meth. Effectively, all cell types secrete nano-sized lipid membrane vesicles, referred to as extracellular vesicles (EVs) that can function as intercellular communication to modulate the physiology and pathology of the cells. This study shows that meth treatments on chronically HIV-infected promonocytic U1 cells induce the release of EVs that promote cellular clustering and syncytia formation, a phenomenon that facilitates HIV pathogenesis. Our analysis also revealed that meth exposure increased intercellular adhesion molecule-1 (ICAM-1) and HIV-Nef protein expression in both large (10 K) and small (100 K) EVs. Further, when meth EVs are applied to uninfected naïve monocyte-derived macrophages (MDMs), we saw a significant increase in cell clustering and syncytia formation. Furthermore, treatment of MDMs with antibodies against ICAM-1 and its receptor, lymphocyte function-associated antigen 1 (LFA1), substantially blocked syncytia formation, and consequently reduced the number of multinucleated cells. In summary, our findings reveal that meth exacerbates HIV pathogenesis in the brain through release of proadhesive EVs, promoting syncytia formation and thereby aiding in the progression of HIV infection in uninfected cells.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH , VIH-1 , Metanfetamina , Vesículas Extracelulares/metabolismo , Células Gigantes , VIH-1/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Metanfetamina/farmacología
3.
Sci Rep ; 12(1): 2019, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35132117

RESUMEN

HIV-1 remains an incurable infection that is associated with substantial economic and epidemiologic impacts. HIV-associated neurocognitive disorders (HAND) are commonly linked with HIV-1 infection; despite the development of combination antiretroviral therapy (cART), HAND is still reported to affect at least 50% of HIV-1 infected individuals. It is believed that the over-amplification of inflammatory pathways, along with release of toxic viral proteins from infected cells, are primarily responsible for the neurological damage that is observed in HAND; however, the underlying mechanisms are not well-defined. Therefore, there is an unmet need to develop more physiologically relevant and reliable platforms for studying these pathologies. In recent years, neurospheres derived from induced pluripotent stem cells (iPSCs) have been utilized to model the effects of different neurotropic viruses. Here, we report the generation of neurospheres from iPSC-derived neural progenitor cells (NPCs) and we show that these cultures are permissive to retroviral (e.g. HIV-1, HTLV-1) replication. In addition, we also examine the potential effects of stem cell derived extracellular vesicles (EVs) on HIV-1 damaged cells as there is abundant literature supporting the reparative and regenerative properties of stem cell EVs in the context of various CNS pathologies. Consistent with the literature, our data suggests that stem cell EVs may modulate neuroprotective and anti-inflammatory properties in damaged cells. Collectively, this study demonstrates the feasibility of NPC-derived neurospheres for modeling HIV-1 infection and, subsequently, highlights the potential of stem cell EVs for rescuing cellular damage induced by HIV-1 infection.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1 , Células Madre Pluripotentes Inducidas/virología , Células-Madre Neurales/virología , Células Cultivadas , Vesículas Extracelulares/fisiología , Infecciones por VIH/complicaciones , VIH-1/fisiología , Humanos , Trastornos Neurocognitivos/etiología , Neuroprotección , Replicación Viral
4.
J Neurovirol ; 27(5): 667-690, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34581996

RESUMEN

The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a highly transmissible disease. SARS-CoV-2 is estimated to have infected over 153 million people and to have caused over 3.2 million global deaths since its emergence in December 2019. SARS-CoV-2 is the seventh coronavirus known to infect humans, and like other coronaviruses, SARS-CoV-2 infection is characterized by a variety of symptoms including general flu-like symptoms such as a fever, sore throat, fatigue, and shortness of breath. Severe cases often display signs of pneumonia, lymphopenia, acute kidney injury, cardiac injury, cytokine storms, lung damage, acute respiratory distress syndrome (ARDS), multiple organ failure, sepsis, and death. There is evidence that around 30% of COVID-19 cases have central nervous system (CNS) or peripheral nervous system (PNS) symptoms along with or in the absence of the previously mentioned symptoms. In cases of CNS/PNS impairments, patients display dizziness, ataxia, seizure, nerve pain, and loss of taste and/or smell. This review highlights the neurological implications of SARS-CoV-2 and provides a comprehensive summary of the research done on SARS-CoV-2 pathology, diagnosis, therapeutics, and vaccines up to May 5.


Asunto(s)
COVID-19/complicaciones , Enfermedades del Sistema Nervioso Central/virología , Enfermedades del Sistema Nervioso Central/diagnóstico , Enfermedades del Sistema Nervioso Central/terapia , Humanos , SARS-CoV-2
5.
Cells ; 10(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916140

RESUMEN

Here, we have attempted to address the timing of EV and virion release from virally infected cells. Uninfected (CEM), HIV-1-infected (J1.1), and human T cell leukemia virus-1 (HTLV-1)-infected (HUT102) cells were synchronized in G0. Viral latency was reversed by increasing gene expression with the addition of serum-rich media and inducers. Supernatants and cell pellets were collected post-induction at different timepoints and assayed for extracellular vesicle (EV) and autophagy markers; and for viral proteins and RNAs. Tetraspanins and autophagy-related proteins were found to be differentially secreted in HIV-1- and HTLV-1-infected cells when compared with uninfected controls. HIV-1 proteins were present at 6 h and their production increased up to 24 h. HTLV-1 proteins peaked at 6 h and plateaued. HIV-1 and HTLV-1 RNA production correlated with viral protein expression. Nanoparticle tracking analysis (NTA) showed increase of EV concentration over time in both uninfected and infected samples. Finally, the HIV-1 supernatant from the 6-h samples was found not to be infectious; however, the virus from the 24-h samples was successfully rescued and infectious. Overall, our data indicate that EV release may occur prior to viral release from infected cells, thereby implicating a potentially significant effect of EVs on uninfected recipient cells prior to subsequent viral infection and spread.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Infecciones por HTLV-I/metabolismo , Infecciones por HTLV-I/patología , Virión/metabolismo , Apoptosis , Biomarcadores/metabolismo , Línea Celular , Medios de Cultivo Condicionados , Citocinas/metabolismo , VIH-1/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Modelos Biológicos , Células Mieloides/metabolismo , ARN Viral/metabolismo , Linfocitos T/metabolismo
6.
Retrovirology ; 18(1): 6, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622348

RESUMEN

BACKGROUND: The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact. RESULTS: Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen. CONCLUSIONS: Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.


Asunto(s)
Células Endoteliales/virología , Vesículas Extracelulares/virología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Animales , Comunicación Celular , Citocinas/análisis , Citocinas/genética , Citocinas/inmunología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/fisiología , Femenino , Infecciones por HTLV-I/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Proteómica , Células THP-1 , Células U937
7.
Viruses ; 12(12)2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33322043

RESUMEN

Human T-cell lymphotropic virus type 1 (HTLV-1) infects 5-10 million people worldwide and is the causative agent of adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) as well as other inflammatory diseases. A major concern is that the most majority of individuals with HTLV-1 are asymptomatic carriers and that there is limited global attention by health care officials, setting up potential conditions for increased viral spread. HTLV-1 transmission occurs primarily through sexual intercourse, blood transfusion, intravenous drug usage, and breast feeding. Currently, there is no cure for HTLV-1 infection and only limited treatment options exist, such as class I interferons (IFN) and Zidovudine (AZT), with poor prognosis. Recently, small membrane-bound structures, known as extracellular vesicles (EVs), have received increased attention due to their potential to carry viral cargo (RNA and proteins) in multiple pathogenic infections (i.e., human immunodeficiency virus type I (HIV-1), Zika virus, and HTLV-1). In the case of HTLV-1, EVs isolated from the peripheral blood and cerebral spinal fluid (CSF) of HAM/TSP patients contained the viral transactivator protein Tax. Additionally, EVs derived from HTLV-1-infected cells (HTLV-1 EVs) promote functional effects such as cell aggregation which enhance viral spread. In this review, we present current knowledge surrounding EVs and their potential role as immune-modulating agents in cancer and other infectious diseases such as HTLV-1 and HIV-1. We discuss various features of EVs that make them prime targets for possible vehicles of future diagnostics and therapies.


Asunto(s)
Comunicación Celular , Vesículas Extracelulares/metabolismo , Infecciones por HTLV-I/virología , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/fisiología , Biomarcadores , Manejo de la Enfermedad , Infecciones por HTLV-I/complicaciones , Infecciones por HTLV-I/epidemiología , Infecciones por HTLV-I/metabolismo , Humanos , Metabolismo de los Lípidos , Estudios Seroepidemiológicos , Carga Viral
8.
Asian Pac J Cancer Prev ; 21(12): 3595-3603, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33369457

RESUMEN

OBJECTIVES: To assess knowledge about human papillomavirus (HPV), vaccination, and HPV-related oropharyngeal cancer (OPC) and to evaluate HPV vaccine acceptability among a sample of undergraduate dental students. METHODS: All third- and fourth-year dental students enrolled in any of the dental schools in Jeddah, Saudi Arabia, were invited to participate in this cross-sectional study. In total, 500 students completed a self-administered questionnaire on HPV, HPV vaccines, and HPV-related OPC knowledge. HPV, HPV vaccine and HPV-related OPC knowledge scores were calculated. Associations of participants characteristics with the knowledge scores and with willingness to take the HPV vaccine were assessed. RESULTS: Of the participants, 62% had heard of HPV infections, and 57% of HPV vaccines. Average knowledge scores were low: HPV knowledge score 3.8±4.5 out of 16, HPV vaccination knowledge score 0.9±1.6 out of 7, OPC knowledge score 0.9±1.2 out of 5. Clinical-year students had better knowledge and were more willing to take the HPV vaccine than were preclinical-year students, as were those vaccinated for hepatitis B versus those not vaccinated for hepatitis B. Students with higher HPV knowledge scores were more willing to take the HPV vaccine (66%) than were those with lower scores (43%) (p=0.018). Only 4% of males and 12% of females reported taking the HPV vaccine. Of those who refused it, 51% cited lack of knowledge as the primary reason. CONCLUSIONS: Knowledge about HPV, its vaccine, and HPV-related OPC is low among this sample of Saudi undergraduate dental students. Improving knowledge among them may increase their awareness, which could affect the care provided to patients.
.


Asunto(s)
Detección Precoz del Cáncer/psicología , Conocimientos, Actitudes y Práctica en Salud , Neoplasias Orofaríngeas/diagnóstico , Infecciones por Papillomavirus/complicaciones , Aceptación de la Atención de Salud , Estudiantes de Odontología/psicología , Vacunación/estadística & datos numéricos , Adulto , Alphapapillomavirus/aislamiento & purificación , Estudios Transversales , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasias Orofaríngeas/epidemiología , Neoplasias Orofaríngeas/psicología , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/administración & dosificación , Pronóstico , Estudiantes de Odontología/estadística & datos numéricos , Encuestas y Cuestionarios , Vacunación/psicología
9.
Front Microbiol ; 10: 2147, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31620104

RESUMEN

Human T-cell leukemia virus-1 (HTLV-1) is a neglected and incurable retrovirus estimated to infect 5 to 10 million worldwide. Specific indigenous Australian populations report infection rates of more than 40%, suggesting a potential evolution of the virus with global implications. HTLV-1 causes adult T-cell leukemia/lymphoma (ATLL), and a neurological disease named HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP). Even though HTLV-1 transmission primarily occurs from cell-to-cell, there is still a gap of knowledge regarding the mechanisms of viral spread and disease progression. We have recently shown that Extracellular Vesicles (EVs) ubiquitously produced by cells may be used by HTLV-1 to transport viral proteins and RNA, and elicit adverse effects on recipient uninfected cells. The viral proteins Tax and HBZ are involved in disease progression and impairment of autophagy in infected cells. Here, we show that activation of HTLV-1 via ionizing radiation (IR) causes a significant increase of intracellular Tax, but not EV-associated Tax. Also, lower density EVs from HTLV-1-infected cells, separated by an Iodixanol density gradient, are positive for gp61+++/Tax+++/HBZ+ proteins (HTLV-1 EVs). We found that HTLV-1 EVs are not infectious when tested in multiple cell lines. However, these EVs promote cell-to-cell contact of uninfected cells, a phenotype which was enhanced with IR, potentially promoting viral spread. We treated humanized NOG mice with HTLV-1 EVs prior to infection and observed an increase in viral RNA synthesis in mice compared to control (EVs from uninfected cells). Proviral DNA levels were also quantified in blood, lung, spleen, liver, and brain post-treatment with HTLV-1 EVs, and we observed a consistent increase in viral DNA levels across all tissues, especially the brain. Finally, we show direct implications of EVs in viral spread and disease progression and suggest a two-step model of infection including the release of EVs from donor cells and recruitment of recipient cells as well as an increase in recipient cell-to-cell contact promoting viral spread.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...