Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Scand J Immunol ; 95(2): e13131, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: covidwho-1583260

RESUMEN

The role of the immune system against coronavirus disease 2019 (COVID-19) is unknown in many aspects, and the protective or pathologic mechanisms of the immune response are poorly understood. Pro-inflammatory cytokine release and a consequent cytokine storm can lead to acute respiratory distress syndrome (ARDS) and result in multi-organ failure. There are many T cell subsets during anti-viral immunity. The Th17-associated response, as a pro-inflammatory pathway, and its consequent outcomes in many autoimmune disorders play a fundamental role in progression of systemic hyper-inflammation during COVID-19. Therapeutic strategies based on immunomodulation therapy could be helpful for targeting hyper-inflammatory immune responses in COVID-19, especially Th17-related inflammation and hyper-cytokinemia. Cell-based immunotherapeutic approaches including mesenchymal stem cells (MSCs), tolerogenic dendritic cells (tolDCs) and regulatory T cells (Tregs) seem to be promising strategies as orchestrators of the immune response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, we highlight Th17-related immunopathology of SARS-CoV-2 infection and discuss cell-based immunomodulatory strategies and their mechanisms for regulation of the hyper-inflammation during COVID-19.


Asunto(s)
COVID-19/patología , COVID-19/terapia , Síndrome de Liberación de Citoquinas/patología , Inmunomodulación/inmunología , Células Th17/inmunología , Traslado Adoptivo/métodos , COVID-19/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citocinas/sangre , Células Dendríticas/trasplante , Humanos , Trasplante de Células Madre Mesenquimatosas , SARS-CoV-2/inmunología , Linfocitos T Reguladores/trasplante
2.
Sci Immunol ; 6(60)2021 06 18.
Artículo en Inglés | MEDLINE | ID: covidwho-1276879

RESUMEN

The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Endosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , SARS-CoV-2/metabolismo , Transducción de Señal/genética , Traslado Adoptivo/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , COVID-19/virología , Células Cultivadas , Femenino , Proteína Forkhead Box O1/deficiencia , Proteína Forkhead Box O1/genética , Voluntarios Sanos , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/inmunología , Transfección , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética , Adulto Joven
3.
Sci Adv ; 6(47)2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-887414

RESUMEN

To affect the COVID-19 pandemic, lifesaving antiviral therapies must be identified. The number of clinical trials that can be performed is limited. We developed mathematical models to project multiple therapeutic approaches. Our models recapitulate off-treatment viral dynamics and predict a three-phase immune response. Simulated treatment with remdesivir, selinexor, neutralizing antibodies, or cellular immunotherapy demonstrates that rapid viral elimination is possible if in vivo potency is sufficiently high. Therapies dosed soon after peak viral load when symptoms develop may decrease shedding duration and immune response intensity but have little effect on viral area under the curve (AUC), which is driven by high early viral loads. Potent therapy dosed before viral peak during presymptomatic infection could lower AUC. Drug resistance may emerge with a moderately potent agent dosed before viral peak. Our results support early treatment for COVID-19 if shedding duration, not AUC, is most predictive of clinical severity.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Traslado Adoptivo/métodos , Alanina/análogos & derivados , Anticuerpos Antivirales/uso terapéutico , Antivirales/uso terapéutico , Anticuerpos ampliamente neutralizantes/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Hidrazinas/uso terapéutico , SARS-CoV-2/fisiología , Triazoles/uso terapéutico , Esparcimiento de Virus/efectos de los fármacos , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/uso terapéutico , Alanina/farmacocinética , Alanina/uso terapéutico , Antivirales/farmacocinética , COVID-19/inmunología , COVID-19/virología , Humanos , Células Asesinas Naturales/inmunología , Modelos Teóricos , Factores de Tiempo , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
4.
Front Immunol ; 11: 1512, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-642764

RESUMEN

Natural Killer (NK) cells are innate immune responders critical for viral clearance and immunomodulation. Despite their vital role in viral infection, the contribution of NK cells in fighting SARS-CoV-2 has not yet been directly investigated. Insights into pathophysiology and therapeutic opportunities can therefore be inferred from studies assessing NK cell phenotype and function during SARS, MERS, and COVID-19. These studies suggest a reduction in circulating NK cell numbers and/or an exhausted phenotype following infection and hint toward the dampening of NK cell responses by coronaviruses. Reduced circulating NK cell levels and exhaustion may be directly responsible for the progression and severity of COVID-19. Conversely, in light of data linking inflammation with coronavirus disease severity, it is necessary to examine NK cell potential in mediating immunopathology. A common feature of coronavirus infections is that significant morbidity and mortality is associated with lung injury and acute respiratory distress syndrome resulting from an exaggerated immune response, of which NK cells are an important component. In this review, we summarize the current understanding of how NK cells respond in both early and late coronavirus infections, and the implication for ongoing COVID-19 clinical trials. Using this immunological lens, we outline recommendations for therapeutic strategies against COVID-19 in clearing the virus while preventing the harm of immunopathological responses.


Asunto(s)
Traslado Adoptivo/métodos , Betacoronavirus/inmunología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Células Asesinas Naturales/inmunología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/inmunología , Corticoesteroides/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Ácido Ascórbico/uso terapéutico , Betacoronavirus/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/virología , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Susceptibilidad a Enfermedades/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Memoria Inmunológica , Interferón Tipo I/metabolismo , Interferón Tipo I/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Ratones , Pandemias , Neumonía Viral/mortalidad , Neumonía Viral/virología , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA