Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 3379, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099719

RESUMO

GATA3 is as a lineage-specific transcription factor that drives the differentiation of CD4+ T helper 2 (Th2) cells, but is also involved in a variety of processes such as immune regulation, proliferation and maintenance in other T cell and non-T cell lineages. Here we show a mechanism utilised by CD4+ T cells to increase mitochondrial mass in response to DNA damage through the actions of GATA3 and AMPK. Activated AMPK increases expression of PPARG coactivator 1 alpha (PPARGC1A or PGC1α protein) at the level of transcription and GATA3 at the level of translation, while DNA damage enhances expression of nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2). PGC1α, GATA3 and NRF2 complex together with the ATR to promote mitochondrial biogenesis. These findings extend the pleotropic interactions of GATA3 and highlight the potential for GATA3-targeted cell manipulation for intervention in CD4+ T cell viability and function after DNA damage.


Assuntos
Linfócitos T CD4-Positivos/citologia , Dano ao DNA , Fator de Transcrição GATA3/metabolismo , Mitocôndrias/metabolismo , Biogênese de Organelas , Proteínas Quinases Ativadas por AMP/metabolismo , Adulto , Linfócitos T CD4-Positivos/metabolismo , Sobrevivência Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Pessoa de Meia-Idade , Fator 2 Relacionado a NF-E2/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Cultura Primária de Células
2.
Front Aging ; 2: 681428, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35821991

RESUMO

Mitochondrial health and cellular metabolism can heavily influence the onset of senescence in T cells. CD8+ EMRA T cells exhibit mitochondrial dysfunction and alterations to oxidative phosphorylation, however, the metabolic properties of senescent CD8+ T cells from people living with type 2 diabetes (T2D) are not known. We show here that mitochondria from T2D CD8+ T cells had a higher oxidative capacity together with increased levels of mitochondrial reactive oxgen species (mtROS), compared to age-matched control cells. While fatty acid uptake was increased, fatty acid oxidation was impaired in T2D CD8+ EMRA T cells, which also showed an accumulation of lipid droplets and decreased AMPK activity. Increasing glucose and fatty acids in healthy CD8+ T cells resulted in increased p-p53 expression and a fragmented mitochondrial morphology, similar to that observed in T2D CD8+ EMRA T cells. The resulting mitochondrial changes are likely to have a profound effect on T cell function. Consequently, a better understanding of these metabolic abnormalities is crucial as metabolic manipulation of these cells may restore correct T cell function and help reduce the impact of T cell dysfunction in T2D.

3.
Front Immunol ; 11: 581758, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162995

RESUMO

We previously reported the Bruton's tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib improve outcomes in a mouse model of polymicrobial sepsis. Now we show that genetic deficiency of the BTK gene alone in Xid mice confers protection against cardiac, renal, and liver injury in polymicrobial sepsis and reduces hyperimmune stimulation ("cytokine storm") induced by an overwhelming bacterial infection. Protection is due in part to enhanced bacterial phagocytosis in vivo, changes in lipid metabolism and decreased activation of NF-κB and the NLRP3 inflammasome. The inactivation of BTK leads to reduced innate immune cell recruitment and a phenotypic switch from M1 to M2 macrophages, aiding in the resolution of sepsis. We have also found that BTK expression in humans is increased in the blood of septic non-survivors, while lower expression is associated with survival from sepsis. Importantly no further reduction in organ damage, cytokine production, or changes in plasma metabolites is seen in Xid mice treated with the BTK inhibitor ibrutinib, demonstrating that the protective effects of BTK inhibitors in polymicrobial sepsis are mediated solely by inhibition of BTK and not by off-target effects of this class of drugs.


Assuntos
Tirosina Quinase da Agamaglobulinemia/metabolismo , Insuficiência de Múltiplos Órgãos/metabolismo , Sepse/metabolismo , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Benzamidas/farmacologia , Modelos Animais de Doenças , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos CBA , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Fagocitose/efeitos dos fármacos , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Sepse/tratamento farmacológico , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/tratamento farmacológico
4.
Front Immunol ; 11: 1991, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903476

RESUMO

Evidence from the global outbreak of SARS-CoV-2 has clearly demonstrated that individuals with pre-existing comorbidities are at a much greater risk of dying from COVID-19. This is of great concern for individuals living with these conditions, and a major challenge for global healthcare systems and biomedical research. Not all comorbidities confer the same risk, however, many affect the function of the immune system, which in turn directly impacts the response to COVID-19. Furthermore, the myriad of drugs prescribed for these comorbidities can also influence the progression of COVID-19 and limit additional treatment options available for COVID-19. Here, we review immune dysfunction in response to SARS-CoV-2 infection and the impact of pre-existing comorbidities on the development of COVID-19. We explore how underlying disease etiologies and common therapies used to treat these conditions exacerbate COVID-19 progression. Moreover, we discuss the long-term challenges associated with the use of both novel and repurposed therapies for the treatment of COVID-19 in patients with pre-existing comorbidities.


Assuntos
Comorbidade , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/mortalidade , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/mortalidade , Betacoronavirus/efeitos dos fármacos , COVID-19 , Infecções por Coronavirus/patologia , Humanos , Pandemias , Pneumonia Viral/patologia , Risco , Fatores de Risco , SARS-CoV-2
5.
J Invest Dermatol ; 140(4): 806-815.e5, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31518559

RESUMO

The skin is our interface with the outside world, and consequently it is exposed to a wide range of microbes and allergens. Recent studies have indicated that allergen-specific skin-resident memory T (TRM) cells play a role in allergic contact dermatitis (ACD). However, the composition and dynamics of the epidermal T-cell subsets during ACD are not known. Here we show that exposure of the skin to the experimental contact allergen DNFB results in a displacement of the normally occurring dendritic epidermal T cells (DETC) concomitant with an accumulation of epidermal CD8+CD69+CD103+ TRM cells in mice. By studying knockout mice, we provide evidence that CD8+ T cells are required for the displacement of the DETC and that DETC are not required for recruitment of CD8+ TRM cells to the epidermis following allergen exposure. We demonstrate that the magnitude of the allergic reaction correlates with the number of CD8+ epidermal TRM cells, which again correlates with allergen dose and number of allergen exposures. Finally, in an attempt to elucidate why CD8+ epidermal TRM cells persist in the epidermis, we show that CD8+ epidermal TRM cells have a higher proliferative capability and are bioenergetically more stable, displaying a higher spare respiratory capacity than DETC.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Dermatite Alérgica de Contato/imunologia , Memória Imunológica , Animais , Linfócitos T CD8-Positivos/patologia , Células Dendríticas/patologia , Dermatite Alérgica de Contato/patologia , Modelos Animais de Doenças , Epiderme/patologia , Camundongos , Camundongos Knockout
6.
Aging Cell ; 19(2): e13067, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31788930

RESUMO

The susceptibility of human CD4+ and CD8+ T cells to senesce differs, with CD8+ T cells acquiring an immunosenescent phenotype faster than the CD4+ T cell compartment. We show here that it is the inherent difference in mitochondrial content that drives this phenotype, with senescent human CD4+ T cells displaying a higher mitochondrial mass. The loss of mitochondria in the senescent human CD8+ T cells has knock-on consequences for nutrient usage, metabolism and function. Senescent CD4+ T cells uptake more lipid and glucose than their CD8+ counterparts, leading to a greater metabolic versatility engaging either an oxidative or a glycolytic metabolism. The enhanced metabolic advantage of senescent CD4+ T cells allows for more proliferation and migration than observed in the senescent CD8+ subset. Mitochondrial dysfunction has been linked to both cellular senescence and aging; however, it is still unclear whether mitochondria play a causal role in senescence. Our data show that reducing mitochondrial function in human CD4+ T cells, through the addition of low-dose rotenone, causes the generation of a CD4+ T cell with a CD8+ -like phenotype. Therefore, we wish to propose that it is the inherent metabolic stability that governs the susceptibility to an immunosenescent phenotype.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Senescência Celular/imunologia , Imunossenescência/fisiologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Adulto , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Movimento Celular/imunologia , Proliferação de Células/fisiologia , Senescência Celular/fisiologia , Glucose/metabolismo , Glicólise/imunologia , Humanos , Antígenos Comuns de Leucócito/sangue , Antígenos Comuns de Leucócito/metabolismo , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Rotenona/farmacologia
7.
Ageing Res Rev ; 47: 24-30, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29902528

RESUMO

The impact of cellular senescence during ageing is well established, however senescence is now recognised to play a role in a variety of age related and metabolic diseases, such as cancer, autoimmune and cardiovascular diseases. It is therefore crucial to gain a better understanding of the mechanisms that control cellular senescence. In recent years our understanding of the intimate relationship between cell metabolism, cell signalling and cellular senescence has greatly improved. In this review we discuss the differing roles of glucose and protein metabolism in both senescent fibroblast and CD8+ T-cells, and explore the impact cellular metabolism has on the senescence-associated secretory phenotype (SASP) of these cell types.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Senescência Celular/fisiologia , Fibroblastos/metabolismo , Animais , Linfócitos T CD8-Positivos/patologia , Fibroblastos/patologia , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/fisiologia , Linfócitos T/metabolismo , Linfócitos T/patologia
8.
Aging Cell ; 17(1)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29024417

RESUMO

Cellular senescence is accompanied by a senescence-associated secretory phenotype (SASP). We show here that primary human senescent CD8+ T cells also display a SASP comprising chemokines, cytokines and extracellular matrix remodelling proteases that are unique to this subset and contribute to age-associated inflammation. We found the CD8+ CD45RA+ CD27- EMRA subset to be the most heterogeneous, with a population aligning with the naïve T cells and another with a closer association to the effector memory subset. However, despite the differing processes that give rise to these senescent CD8+ T cells once generated, they both adopt a unique secretory profile with no commonality to any other subset, aligning more closely with senescence than quiescence. Furthermore, we also show that the SASP observed in senescent CD8+ T cells is governed by p38 MAPK signalling.


Assuntos
Linfócitos T CD8-Positivos/citologia , Senescência Celular/genética , Citocinas/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Adulto , Citocinas/metabolismo , Dano ao DNA/genética , Voluntários Saudáveis , Humanos , Sistema de Sinalização das MAP Quinases/genética , Pessoa de Meia-Idade , Fenótipo , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...