Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
STAR Protoc ; 5(1): 102939, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38451821

RESUMEN

M1- and M2-like macrophages infected with Mycobacterium tuberculosis (Mtb) have been found to differ in their capacity to elicit memory CD4+ T cell activation. Here, we present a protocol to quantify and isolate the subset of human memory CD4+ T cells activated in response to autologous monocyte-derived macrophages (MDMs) infected with virulent Mtb. We describe steps for CD14+ monocyte isolation, generating MDMs, culturing Mtb and infection of macrophages, and identifying activated CD4+ T cells by flow cytometry. For complete details on the use and execution of this protocol, please refer to Gail et al.1.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Linfocitos T , Macrófagos/microbiología , Linfocitos T CD4-Positivos
2.
iScience ; 26(9): 107706, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37694142

RESUMEN

Direct recognition of Mycobacterium tuberculosis (Mtb)-infected cells is required for protection by CD4+ T cells. While impaired T cell recognition of Mtb-infected macrophages was demonstrated in mice, data are lacking for humans. Using T cells and monocyte-derived macrophages (MDMs) from individuals with latent Mtb infection (LTBI), we quantified the frequency of memory CD4+ T cell activation in response to autologous MDMs infected with virulent Mtb. We observed robust T cell activation in response to Mtb infection of M1-like macrophages differentiated using GM-CSF, while M2-like macrophages differentiated using M-CSF were poorly recognized. However, non-infected GM-CSF and M-CSF MDMs loaded with exogenous antigens elicited similar CD4+ T cell activation. IL-10 was preferentially secreted by infected M-CSF MDMs, and neutralization improved T cell activation. These results suggest that preferential infection of macrophages with an M2-like phenotype limits T cell-mediated protection against Mtb. Vaccine development should focus on T cell recognition of Mtb-infected macrophages.

3.
iScience ; 25(8): 104716, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35813876

RESUMEN

The COVID-19 pandemic has caused over four million deaths and effective methods to control CoV-2 infection, in addition to vaccines, are needed. The CoV-2 binds to the ACE2 on human cells through the receptor-binding domain (RBD) of the trimeric spike protein. Our modeling studies show that a modified trimeric RBD (tRBD) can interact with three ACE2 receptors, unlike the native spike protein, which binds to only one ACE2. We found that tRBD binds to the ACE2 with 58-fold higher affinity than monomeric RBD (mRBD) and blocks spike-dependent pseudoviral infection over 4-fold more effectively compared to the mRBD. Although mRBD failed to block CoV-2 USA-WA1/2020 infection, tRBD efficiently blocked the true virus infection in plaque assays. We show that tRBD is a potent inhibitor of CoV-2 through both competitive binding to the ACE2 and steric hindrance, and has the potential to emerge as a first-line therapeutic method to control COVID-19.

4.
Nat Commun ; 12(1): 7200, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34893603

RESUMEN

Chimeric antigen receptor (CAR) T cells targeting the CD19 antigen are effective in treating adults and children with B-cell malignancies. Place-of-care manufacturing may improve performance and accessibility by obviating the need to cryopreserve and transport cells to centralized facilities. Here we develop an anti-CD19 CAR (CAR19) comprised of the 4-1BB co-stimulatory and TNFRSF19 transmembrane domains, showing anti-tumor efficacy in an in vivo xenograft lymphoma model. CAR19 T cells are manufactured under current good manufacturing practices (cGMP) at two disparate clinical sites, Moscow (Russia) and Cleveland (USA). The CAR19 T-cells is used to treat patients with relapsed/refractory pediatric B-cell Acute Lymphocytic Leukemia (ALL; n = 31) or adult B-cell Lymphoma (NHL; n = 23) in two independently conducted phase I clinical trials with safety as the primary outcome (NCT03467256 and NCT03434769, respectively). Probability of measurable residual disease-negative remission was also a primary outcome in the ALL study. Secondary outcomes include complete remission (CR) rates, overall survival and median duration of response. CR rates are 89% (ALL) and 73% (NHL). After a median follow-up of 17 months, one-year survival rate of ALL complete responders is 79.2% (95%CI 64.5‒97.2%) and median duration of response is 10.2 months. For NHL complete responders one-year survival is 92.9%, and median duration of response has not been reached. Place-of-care manufacturing produces consistent CAR-T cell products at multiple sites that are effective for the treatment of patients with B-cell malignancies.


Asunto(s)
Antígenos CD19/inmunología , Linfocitos B/inmunología , Linfoma de Células B/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Animales , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Supervivencia sin Progresión , Receptores de Antígenos de Linfocitos T , Receptores del Factor de Necrosis Tumoral/química , Federación de Rusia , Estados Unidos , Adulto Joven
5.
PLoS Pathog ; 17(7): e1009715, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34270613

RESUMEN

SARS-CoV and SARS-CoV-2 encode spike proteins that bind human ACE2 on the cell surface to enter target cells during infection. A small fraction of humans encode variants of ACE2, thus altering the biochemical properties at the protein interaction interface. These and other ACE2 coding mutants can reveal how the spike proteins of each virus may differentially engage the ACE2 protein surface during infection. We created an engineered HEK 293T cell line for facile stable transgenic modification, and expressed the major human ACE2 allele or 28 of its missense mutants, 24 of which are possible through single nucleotide changes from the human reference sequence. Infection with SARS-CoV or SARS-CoV-2 spike pseudotyped lentiviruses revealed that high ACE2 cell-surface expression could mask the effects of impaired binding during infection. Drastically reducing ACE2 cell surface expression revealed a range of infection efficiencies across the panel of mutants. Our infection results revealed a non-linear relationship between soluble SARS-CoV-2 RBD binding to ACE2 and pseudovirus infection, supporting a major role for binding avidity during entry. While ACE2 mutants D355N, R357A, and R357T abrogated entry by both SARS-CoV and SARS-CoV-2 spike proteins, the Y41A mutant inhibited SARS-CoV entry much more than SARS-CoV-2, suggesting differential utilization of the ACE2 side-chains within the largely overlapping interaction surfaces utilized by the two CoV spike proteins. These effects correlated well with cytopathic effects observed during SARS-CoV-2 replication in ACE2-mutant cells. The panel of ACE2 mutants also revealed altered ACE2 surface dependencies by the N501Y spike variant, including a near-complete utilization of the K353D ACE2 variant, despite decreased infection mediated by the parental SARS-CoV-2 spike. Our results clarify the relationship between ACE2 abundance, binding, and infection, for various SARS-like coronavirus spike proteins and their mutants, and inform our understanding for how changes to ACE2 sequence may correspond with different susceptibilities to infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/etiología , SARS-CoV-2/fisiología , Síndrome Respiratorio Agudo Grave/etiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Glicoproteína de la Espiga del Coronavirus/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/genética , COVID-19/virología , Células HEK293 , Humanos , Mutación Missense , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/virología
6.
Nat Commun ; 9(1): 3136, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087337

RESUMEN

Protective adaptive immunity to Zika virus (ZIKV) has been mainly attributed to cytotoxic CD8+ T cells and neutralizing antibodies, while the participation of CD4+ T cells in resistance has remained largely uncharacterized. Here, we show a neutralizing antibody response, dependent on CD4+ T cells and IFNγ signaling, which we detected during the first week of infection and is associated with reduced viral load in the brain, prevention of rapid disease onset and survival. We demonstrate participation of these components in the resistance to ZIKV during primary infection and in murine adoptive transfer models of heterologous ZIKV infection in a background of IFNR deficiency. The protective effect of adoptively transferred CD4+ T cells requires IFNγ signaling, CD8+ T cells and B lymphocytes in recipient mice. Together, this indicates the importance of CD4+ T cell responses in future vaccine design for ZIKV.


Asunto(s)
Inmunidad Adaptativa , Traslado Adoptivo , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Interferón gamma/metabolismo , Infección por el Virus Zika/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Peso Corporal , Chlorocebus aethiops , Femenino , Inmunoglobulina G , Masculino , Ratones , Células Vero , Virus Zika
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...