Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Immunol ; 24(12): 2008-2020, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38012409

RESUMO

Our increased understanding of how key metabolic pathways are activated and regulated in malignant cells has identified metabolic vulnerabilities of cancers. Translating this insight to the clinics, however, has proved challenging. Roadblocks limiting efficacy of drugs targeting cancer metabolism may lie in the nature of the metabolic ecosystem of tumors. The exchange of metabolites and growth factors between cancer cells and nonmalignant tumor-resident cells is essential for tumor growth and evolution, as well as the development of an immunosuppressive microenvironment. In this Review, we will examine the metabolic interplay between tumor-resident cells and how targeted inhibition of specific metabolic enzymes in malignant cells could elicit pro-tumorigenic effects in non-transformed tumor-resident cells and inhibit the function of tumor-specific T cells. To improve the efficacy of metabolism-targeted anticancer strategies, a holistic approach that considers the effect of metabolic inhibitors on major tumor-resident cell populations is needed.


Assuntos
Ecossistema , Neoplasias , Humanos , Neoplasias/metabolismo , Carcinogênese , Linfócitos T/metabolismo , Microambiente Tumoral
2.
Nat Immunol ; 24(2): 349-358, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36717723

RESUMO

The biology driving individual patient responses to severe acute respiratory syndrome coronavirus 2 infection remains ill understood. Here, we developed a patient-centric framework leveraging detailed longitudinal phenotyping data and covering a year after disease onset, from 215 infected individuals with differing disease severities. Our analyses revealed distinct 'systemic recovery' profiles, with specific progression and resolution of the inflammatory, immune cell, metabolic and clinical responses. In particular, we found a strong inter-patient and intra-patient temporal covariation of innate immune cell numbers, kynurenine metabolites and lipid metabolites, which highlighted candidate immunologic and metabolic pathways influencing the restoration of homeostasis, the risk of death and that of long COVID. Based on these data, we identified a composite signature predictive of systemic recovery, using a joint model on cellular and molecular parameters measured soon after disease onset. New predictions can be generated using the online tool http://shiny.mrc-bsu.cam.ac.uk/apps/covid-19-systemic-recovery-prediction-app , designed to test our findings prospectively.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Síndrome de COVID-19 Pós-Aguda , Cinurenina , Assistência Centrada no Paciente
3.
Nat Immunol ; 20(10): 1311-1321, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31527833

RESUMO

Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.


Assuntos
Linfócitos B/imunologia , Complexo II de Transporte de Elétrons/genética , Inflamação/metabolismo , Linfocitose/imunologia , Mitocôndrias/metabolismo , Mutação/genética , Anti-Inflamatórios/farmacologia , Respiração Celular , Células Cultivadas , Fumaratos/metabolismo , Glicólise , Humanos , Inflamação/genética , Interleucina-6/antagonistas & inibidores , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Consumo de Oxigênio , Estudos Prospectivos , Transdução de Sinais , Sequenciamento do Exoma
4.
Immunity ; 48(3): 542-555.e6, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29523440

RESUMO

Glycolysis is linked to the rapid response of memory CD8+ T cells, but the molecular and subcellular structural elements enabling enhanced glucose metabolism in nascent activated memory CD8+ T cells are unknown. We found that rapid activation of protein kinase B (PKB or AKT) by mammalian target of rapamycin complex 2 (mTORC2) led to inhibition of glycogen synthase kinase 3ß (GSK3ß) at mitochondria-endoplasmic reticulum (ER) junctions. This enabled recruitment of hexokinase I (HK-I) to the voltage-dependent anion channel (VDAC) on mitochondria. Binding of HK-I to VDAC promoted respiration by facilitating metabolite flux into mitochondria. Glucose tracing pinpointed pyruvate oxidation in mitochondria, which was the metabolic requirement for rapid generation of interferon-γ (IFN-γ) in memory T cells. Subcellular organization of mTORC2-AKT-GSK3ß at mitochondria-ER contact sites, promoting HK-I recruitment to VDAC, thus underpins the metabolic reprogramming needed for memory CD8+ T cells to rapidly acquire effector function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Retículo Endoplasmático/metabolismo , Metabolismo Energético , Memória Imunológica , Mitocôndrias/metabolismo , Transdução de Sinais , Respiração Celular , Retículo Endoplasmático/ultraestrutura , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicólise , Membranas Intracelulares/metabolismo , Ativação Linfocitária , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Mitocôndrias/ultraestrutura , Modelos Biológicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/deficiência
5.
Nat Immunol ; 14(10): 1064-72, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23955661

RESUMO

Antigen-experienced memory T cells acquire effector function with innate-like kinetics; however, the metabolic requirements of these cells are unknown. Here we show that rapid interferon-γ (IFN-γ) production of effector memory (EM) CD8(+) T cells, activated through stimulation mediated by the T cell antigen receptor (TCR) and the costimulatory receptor CD28 or through cognate interactions, was linked to increased glycolytic flux. EM CD8(+) T cells exhibited more glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity at early time points, before proliferation commenced, than did naive cells activated under similar conditions. CD28 signaling via the serine-threonine kinase Akt and the metabolic-checkpoint kinase mTORC2 was needed to sustain TCR-mediated immediate-early glycolysis. Unlike glycolysis in proliferating cells, immediate-early glycolysis in memory CD8(+) T cells was rapamycin insensitive. Thus, CD8(+) memory T cells have an Akt-dependent 'imprinted' glycolytic potential that is required for efficient immediate-early IFN-γ recall responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Montagem e Desmontagem da Cromatina , Epitopos de Linfócito T/imunologia , Glicólise , Herpesvirus Humano 4/imunologia , Humanos , Interferon gama/biossíntese , Interferon gama/genética , Ativação Linfocitária , Alvo Mecanístico do Complexo 2 de Rapamicina , Metaboloma , Metabolômica , Complexos Multiproteicos/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
6.
Immunity ; 44(6): 1312-24, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27212436

RESUMO

How systemic metabolic alterations during acute infections impact immune cell function remains poorly understood. We found that acetate accumulates in the serum within hours of systemic bacterial infections and that these increased acetate concentrations are required for optimal memory CD8(+) T cell function in vitro and in vivo. Mechanistically, upon uptake by memory CD8(+) T cells, stress levels of acetate expanded the cellular acetyl-coenzyme A pool via ATP citrate lyase and promoted acetylation of the enzyme GAPDH. This context-dependent post-translational modification enhanced GAPDH activity, catalyzing glycolysis and thus boosting rapid memory CD8(+) T cell responses. Accordingly, in a murine Listeria monocytogenes model, transfer of acetate-augmented memory CD8(+) T cells exerted superior immune control compared to control cells. Our results demonstrate that increased systemic acetate concentrations are functionally integrated by CD8(+) T cells and translate into increased glycolytic and functional capacity. The immune system thus directly relates systemic metabolism with immune alertness.


Assuntos
Acetatos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Listeria monocytogenes/imunologia , Listeriose/imunologia , ATP Citrato (pro-S)-Liase/metabolismo , Acetil-CoA C-Acetiltransferase/metabolismo , Animais , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora) , Glicólise , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Estresse Fisiológico/imunologia
7.
Immunity ; 42(6): 1033-47, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26084023

RESUMO

Expansion and acquisition of Th1 cell effector function requires metabolic reprogramming; however, the signals instructing these adaptations remain poorly defined. Here we found that in activated human T cells, autocrine stimulation of the complement receptor CD46, and specifically its intracellular domain CYT-1, was required for induction of the amino acid (AA) transporter LAT1 and enhanced expression of the glucose transporter GLUT1. Furthermore, CD46 activation simultaneously drove expression of LAMTOR5, which mediated assembly of the AA-sensing Ragulator-Rag-mTORC1 complex and increased glycolysis and oxidative phosphorylation (OXPHOS), required for cytokine production. T cells from CD46-deficient patients, characterized by defective Th1 cell induction, failed to upregulate the molecular components of this metabolic program as well as glycolysis and OXPHOS, but IFN-γ production could be reinstated by retrovirus-mediated CD46-CYT-1 expression. These data establish a critical link between the complement system and immunometabolic adaptations driving human CD4(+) T cell effector function.


Assuntos
Proteínas do Sistema Complemento/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Proteína Cofatora de Membrana/metabolismo , Células Th1/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Reprogramação Celular/imunologia , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Glicólise , Proteínas de Homeodomínio/metabolismo , Humanos , Imunidade Celular/genética , Interferon gama/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteína Cofatora de Membrana/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Neuropeptídeos/metabolismo , Fosforilação Oxidativa , RNA Interferente Pequeno/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima
9.
J Autoimmun ; 124: 102714, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34403915

RESUMO

BACKGROUND: Viral infections may trigger autoimmunity in genetically predisposed individuals. Immunizations mimic viral infections immunologically, but only in rare instances vaccinations coincide with the onset of autoimmunity. Inadvertent vaccine injection into periarticular shoulder tissue can cause inflammatory tissue damage ('shoulder injury related to vaccine administration, SIRVA). Thus, this accident provides a model to study if vaccine-induced pathogen-specific immunity accompanied by a robust inflammatory insult may trigger autoimmunity in specific genetic backgrounds. METHODS: We studied 16 otherwise healthy adults with suspected SIRVA occurring following a single work-related influenza immunization campaign in 2017. We performed ultrasound, immunophenotypic analyses, HLA typing, and influenza- and self-reactivity functional immunoassays. Vaccine-related bone toxicity and T cell/osteoclast interactions were assessed in vitro. FINDINGS: Twelve of the 16 subjects had evidence of inflammatory tissue damage on imaging, including bone erosions in six. Tissue damage was associated with a robust peripheral blood T and B cell activation signature and extracellular matrix-reactive autoantibodies. All subjects with erosions were HLA-DRB1*04 positive and showed extracellular matrix-reactive HLA-DRB1*04 restricted T cell responses targeting heparan sulfate proteoglycan (HSPG). Antigen-specific T cells potently activated osteoclasts via RANK/RANK-L, and the osteoclast activation marker Trap5b was high in sera of patients with an erosive shoulder injury. In vitro, the vaccine component alpha-tocopheryl succinate recapitulated bone toxicity and stimulated osteoclasts. Auto-reactivity was transient, with no evidence of progression to rheumatoid arthritis or overt autoimmune disease. CONCLUSION: Vaccine misapplication, potentially a genetic predisposition, and vaccine components contribute to SIRVA. The association with autoimmunity risk allele HLA-DRB1*04 needs to be further investigated. Despite transient autoimmunity, SIRVA was not associated with progression to autoimmune disease during two years of follow-up.


Assuntos
Inflamação/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Cápsula Articular/imunologia , Orthomyxoviridae/fisiologia , Osteoclastos/imunologia , Linfócitos T/imunologia , Adulto , Autoimunidade , Doença Crônica , Matriz Extracelular/metabolismo , Feminino , Predisposição Genética para Doença , Cadeias HLA-DRB1/genética , Proteoglicanas de Heparan Sulfato/imunologia , Teste de Histocompatibilidade , Humanos , Masculino , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fosfatase Ácida Resistente a Tartarato/sangue , Vacinação/efeitos adversos , Adulto Jovem
10.
Eur J Immunol ; 48(10): 1632-1643, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30028501

RESUMO

The role of mitochondrial biogenesis during naïve to effector differentiation of CD8+ T cells remains ill explored. In this study, we describe a critical role for early mitochondrial biogenesis in supporting cytokine production of nascent activated human naïve CD8+ T cells. Specifically, we found that prior to the first round of cell division activated naïve CD8+ T cells rapidly increase mitochondrial mass, mitochondrial respiration, and mitochondrial reactive oxygen species (mROS) generation, which were all inter-linked and important for CD8+ T cell effector maturation. Inhibition of early mitochondrial biogenesis diminished mROS dependent IL-2 production - as well as subsequent IL-2 dependent TNF, IFN-γ, perforin, and granzyme B production. Together, these findings point to the importance of mitochondrial biogenesis during early effector maturation of CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Mitocôndrias/fisiologia , Biogênese de Organelas , Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Humanos , Interleucina-2/imunologia , Ativação Linfocitária , Espécies Reativas de Oxigênio/metabolismo
11.
J Immunol ; 196(1): 106-14, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26621861

RESUMO

Effector memory (EM) CD4(+) T cells recirculate between normoxic blood and hypoxic tissues to screen for cognate Ag. How mitochondria of these cells, shuttling between normoxia and hypoxia, maintain bioenergetic efficiency and stably uphold antiapoptotic features is unknown. In this study, we found that human EM CD4(+) T cells had greater spare respiratory capacity (SRC) than did naive counterparts, which was immediately accessed under hypoxia. Consequently, hypoxic EM cells maintained ATP levels, survived and migrated better than did hypoxic naive cells, and hypoxia did not impair their capacity to produce IFN-γ. EM CD4(+) T cells also had more abundant cytosolic GAPDH and increased glycolytic reserve. In contrast to SRC, glycolytic reserve was not tapped under hypoxic conditions, and, under hypoxia, glucose metabolism contributed similarly to ATP production in naive and EM cells. However, both under normoxic and hypoxic conditions, glucose was critical for EM CD4(+) T cell survival. Mechanistically, in the absence of glycolysis, mitochondrial membrane potential (ΔΨm) of EM cells declined and intrinsic apoptosis was triggered. Restoring pyruvate levels, the end product of glycolysis, preserved ΔΨm and prevented apoptosis. Furthermore, reconstitution of reactive oxygen species (ROS), whose production depends on ΔΨm, also rescued viability, whereas scavenging mitochondrial ROS exacerbated apoptosis. Rapid access of SRC in hypoxia, linked with built-in, oxygen-resistant glycolytic reserve that functionally insulates ΔΨm and mitochondrial ROS production from oxygen tension changes, provides an immune-metabolic basis supporting survival, migration, and function of EM CD4(+) T cells in normoxic and hypoxic conditions.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/metabolismo , Hipóxia Celular/imunologia , Glucose/metabolismo , Mitocôndrias/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Movimento Celular , Sobrevivência Celular/imunologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Glicólise , Humanos , Memória Imunológica/imunologia , Interferon gama/biossíntese , Potencial da Membrana Mitocondrial , Microfluídica , Oxigênio/metabolismo , Ácido Pirúvico/metabolismo , Espécies Reativas de Oxigênio/metabolismo
12.
Eur J Immunol ; 44(12): 3614-20, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25251877

RESUMO

ATP-binding cassette (ABC) transporters, including ABC-transporter B1 (ABCB1), extrude drugs, metabolites, and other compounds (such as mitotracker green (MTG)) from cells. Susceptibility of CD4(+) regulatory T (Treg) cells to the ABCB1-substrate cyclophosphamide (CPA) has been reported. Here, we characterized ABCB1 expression and function in human CD4(+) T-cell subsets. Naïve, central memory, and effector-memory CD4(+) T cells, but not Treg cells, effluxed MTG in an ABCB1-dependent manner. In line with this, ABCB1 mRNA and protein was expressed by nonregulatory CD4(+) T-cell subsets, but not Treg cells. In vitro, the ABCB1-substrate CPA was cytotoxic for Treg cells at a 100-fold lower dose than for nonregulatory counterparts, and, inversely, verapamil, an inhibitor of ABC transporters, increased CPA-toxicity in nonregulatory CD4(+) T cells but not Treg cells. Thus, Treg cells lack expression of ABCB1, rendering them selectively susceptible to CPA. Our findings provide mechanistic support for therapeutic strategies using CPA to boost anti-tumor immunity by selectively depleting Treg cells.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Ciclofosfamida/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/imunologia , Antineoplásicos Alquilantes/farmacocinética , Apoptose/imunologia , Ciclofosfamida/farmacocinética , Citotoxinas/farmacocinética , Citotoxinas/farmacologia , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Depleção Linfocítica/métodos , Masculino , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Linfócitos T Reguladores/citologia
13.
PLoS Pathog ; 9(3): e1003200, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23505367

RESUMO

Infection of the developing fetus with human cytomegalovirus (HCMV) is a major cause of central nervous system disease in infants and children; however, mechanism(s) of disease associated with this intrauterine infection remain poorly understood. Utilizing a mouse model of HCMV infection of the developing CNS, we have shown that peripheral inoculation of newborn mice with murine CMV (MCMV) results in CNS infection and developmental abnormalities that recapitulate key features of the human infection. In this model, animals exhibit decreased granule neuron precursor cell (GNPC) proliferation and altered morphogenesis of the cerebellar cortex. Deficits in cerebellar cortical development are symmetric and global even though infection of the CNS results in a non-necrotizing encephalitis characterized by widely scattered foci of virus-infected cells with mononuclear cell infiltrates. These findings suggested that inflammation induced by MCMV infection could underlie deficits in CNS development. We investigated the contribution of host inflammatory responses to abnormal cerebellar development by modulating inflammatory responses in infected mice with glucocorticoids. Treatment of infected animals with glucocorticoids decreased activation of CNS mononuclear cells and expression of inflammatory cytokines (TNF-α, IFN-ß and IFNγ) in the CNS while minimally impacting CNS virus replication. Glucocorticoid treatment also limited morphogenic abnormalities and normalized the expression of developmentally regulated genes within the cerebellum. Importantly, GNPC proliferation deficits were normalized in MCMV infected mice following glucocorticoid treatment. Our findings argue that host inflammatory responses to MCMV infection contribute to deficits in CNS development in MCMV infected mice and suggest that similar mechanisms of disease could be responsible for the abnormal CNS development in human infants infected in-utero with HCMV.


Assuntos
Doenças do Sistema Nervoso Central/tratamento farmacológico , Encefalite/tratamento farmacológico , Glucocorticoides/uso terapêutico , Infecções por Herpesviridae/tratamento farmacológico , Muromegalovirus , Animais , Animais Recém-Nascidos/virologia , Encéfalo/virologia , Proliferação de Células , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/virologia , Doenças do Sistema Nervoso Central/virologia , Cerebelo/efeitos dos fármacos , Cerebelo/embriologia , Cerebelo/virologia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Encefalite/virologia , Infecções por Herpesviridae/virologia , Interferon beta/biossíntese , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Prednisolona/uso terapêutico , Fator de Necrose Tumoral alfa/biossíntese
14.
Science ; 385(6704): eadk4898, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38781354

RESUMO

After infection of B cells, Epstein-Barr virus (EBV) engages host pathways that mediate cell proliferation and transformation, contributing to the propensity of the virus to drive immune dysregulation and lymphomagenesis. We found that the EBV protein EBNA2 initiates nicotinamide adenine dinucleotide (NAD) de novo biosynthesis by driving expression of the metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) in infected B cells. Virus-enforced NAD production sustained mitochondrial complex I activity, to match adenosine triphosphate (ATP) production with bioenergetic requirements of proliferation and transformation. In transplant patients, IDO1 expression in EBV-infected B cells, and a serum signature of increased IDO1 activity, preceded development of lymphoma. In humanized mice infected with EBV, IDO1 inhibition reduced both viremia and lymphomagenesis. Virus-orchestrated NAD biosynthesis is therefore a druggable metabolic vulnerability of EBV-driven B cell transformation, opening therapeutic possibilities for EBV-related diseases.


Assuntos
Trifosfato de Adenosina , Linfócitos B , Transformação Celular Viral , Infecções por Vírus Epstein-Barr , Antígenos Nucleares do Vírus Epstein-Barr , Herpesvirus Humano 4 , Indolamina-Pirrol 2,3,-Dioxigenase , NAD , Animais , Humanos , Camundongos , Trifosfato de Adenosina/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proliferação de Células , Complexo I de Transporte de Elétrons/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/fisiologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Linfoma/virologia , NAD/metabolismo , Proteínas Virais , Viremia
15.
Theranostics ; 11(9): 4011-4029, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754045

RESUMO

Rationale: Adenylosuccinate lyase (ADSL) is an essential enzyme for de novo purine biosynthesis. Here we sought to investigate the putative role of ADSL in colorectal carcinoma (CRC) carcinogenesis and response to antimetabolites. Methods: ADSL expression levels were assessed by immunohistochemistry or retrieved from The Cancer Genome Atlas (TCGA) dataset. The effects of ADSL silencing or overexpression were evaluated on CRC cell proliferation, cell migration and cell-cycle. In vivo tumor growth was assessed by the chicken chorioallantoic membrane (CAM). Transfected cell lines or patient-derived organoids (PDO) were treated with 5-fluorouracil (5-FU) and 6-mercaptopurine (6-MP) and drug response was correlated with ADSL expression levels. Metabolomic and transcriptomic profiling were performed to identify dysregulated pathways and ADSL downstream effectors. Mitochondrial respiration and glycolytic capacity were measured using Seahorse; mitochondrial membrane potential and the accumulation of ROS were measured by FACS using MitoTracker Red and MitoSOX staining, respectively. Activation of canonical pathways was assessed by immunohistochemistry and immunoblotting. Results: ADSL expression is significantly increased in CRC tumors compared to non-tumor tissue. ADSL-high CRCs show upregulation of genes involved in DNA synthesis, DNA repair and cell cycle. Accordingly, ADSL overexpression accelerated progression through the cell cycle and significantly increased proliferation and migration in CRC cell lines. Additionally, ADSL expression increased tumor growth in vivo and sensitized CRCs to 6-MP in vitro, ex vivo (PDOs) and in vivo (CAM model). ADSL exerts its oncogenic function by affecting mitochondrial function via alteration of the TCA cycle and impairment of mitochondrial respiration. The KEAP1-NRF2 and mTORC1-cMyc axis are independently activated upon ADSL overexpression and may favor the survival and proliferation of ROS-accumulating cells, favoring DNA damage and tumorigenesis. Conclusions: Our results suggest that ADSL is a novel oncogene in CRC, modulating mitochondrial function, metabolism and oxidative stress, thus promoting cell cycle progression, proliferation and migration. Our results also suggest that ADSL is a predictive biomarker of response to 6-mercaptopurine in the pre-clinical setting.


Assuntos
Adenilossuccinato Liase/genética , Neoplasias Colorretais/genética , Mitocôndrias/genética , Fator 2 Relacionado a NF-E2/genética , Oncogenes/genética , Proteínas Proto-Oncogênicas c-myc/genética , Serina-Treonina Quinases TOR/genética , Células CACO-2 , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Respiração Celular/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica/genética , Células HT29 , Humanos , Mitocôndrias/patologia
16.
Nat Commun ; 12(1): 1460, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674584

RESUMO

Mitochondria are important regulators of macrophage polarisation. Here, we show that arginase-2 (Arg2) is a microRNA-155 (miR-155) and interleukin-10 (IL-10) regulated protein localized at the mitochondria in inflammatory macrophages, and is critical for IL-10-induced modulation of mitochondrial dynamics and oxidative respiration. Mechanistically, the catalytic activity and presence of Arg2 at the mitochondria is crucial for oxidative phosphorylation. We further show that Arg2 mediates this process by increasing the activity of complex II (succinate dehydrogenase). Moreover, Arg2 is essential for IL-10-mediated downregulation of the inflammatory mediators succinate, hypoxia inducible factor 1α (HIF-1α) and IL-1ß in vitro. Accordingly, HIF-1α and IL-1ß are highly expressed in an LPS-induced in vivo model of acute inflammation using Arg2-/- mice. These findings shed light on a new arm of IL-10-mediated metabolic regulation, working to resolve the inflammatory status of the cell.


Assuntos
Arginase/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Animais , Arginase/genética , Regulação para Baixo , Feminino , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Mitocôndrias/enzimologia , Succinato Desidrogenase/metabolismo
17.
J Immunol ; 181(3): 2111-23, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641350

RESUMO

Human CMV infection of the neonatal CNS results in long-term neurologic sequelae. To define the pathogenesis of fetal human CMV CNS infections, we investigated mechanisms of virus clearance from the CNS of neonatal BALB/c mice infected with murine CMV (MCMV). Virus titers peaked in the CNS between postnatal days 10-14 and infectious virus was undetectable by postnatal day 21. Congruent with virus clearance was the recruitment of CD8(+) T cells into the CNS. Depletion of CD8(+) T cells resulted in death by postnatal day 15 in MCMV-infected animals and increased viral loads in the liver, spleen, and the CNS, suggesting an important role for these cells in the control of MCMV replication in the newborn brain. Examination of brain mononuclear cells revealed that CD8(+) T cell infiltrates expressed high levels of CD69, CD44, and CD49d. IE1(168)-specific CD8(+) T cells accumulated in the CNS and produced IFN-gamma and TNF-alpha but not IL-2 following peptide stimulation. Moreover, adoptive transfer of brain mononuclear cells resulted in decreased virus burden in immunodepleted MCMV-infected syngeneic mice. Depletion of the CD8(+) cell population following transfer eliminated control of virus replication. In summary, these results show that functionally mature virus-specific CD8(+) T cells are recruited to the CNS in mice infected with MCMV as neonates.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/virologia , Muromegalovirus/fisiologia , Replicação Viral , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/imunologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/imunologia , Separação Celular , Células Cultivadas , Doenças do Sistema Nervoso Central/metabolismo , Doenças do Sistema Nervoso Central/patologia , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Proteínas Imediatamente Precoces/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/citologia , Monócitos/imunologia , Fenótipo , Taxa de Sobrevida
18.
Cell Metab ; 32(3): 457-467.e5, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32738204

RESUMO

Serum acetate increases upon systemic infection. Acutely, assimilation of acetate expands the capacity of memory CD8+ T cells to produce IFN-γ. Whether acetate modulates memory CD8+ T cell metabolism and function during pathogen re-encounter remains unexplored. Here we show that at sites of infection, high acetate concentrations are being reached, yet memory CD8+ T cells shut down the acetate assimilating enzymes ACSS1 and ACSS2. Acetate, being thus largely excluded from incorporation into cellular metabolic pathways, now had different effects, namely (1) directly activating glutaminase, thereby augmenting glutaminolysis, cellular respiration, and survival, and (2) suppressing TCR-triggered calcium flux, and consequently cell activation and effector cell function. In vivo, high acetate abundance at sites of infection improved pathogen clearance while reducing immunopathology. This indicates that, during different stages of the immune response, the same metabolite-acetate-induces distinct immunometabolic programs within the same cell type.


Assuntos
Acetatos/metabolismo , Anti-Inflamatórios/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Acetatos/sangue , Acetatos/imunologia , Animais , Anti-Inflamatórios/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Sci Signal ; 12(599)2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530731

RESUMO

Transforming growth factor-ß (TGF-ß) is produced by tumors, and increased amounts of this cytokine in the tumor microenvironment and serum are associated with poor patient survival. TGF-ß-mediated suppression of antitumor T cell responses contributes to tumor growth and survival. However, TGF-ß also has tumor-suppressive activity; thus, dissecting cell type-specific molecular effects may inform therapeutic strategies targeting this cytokine. Here, using human peripheral and tumor-associated lymphocytes, we investigated how tumor-derived TGF-ß suppresses a key antitumor function of CD4+ T cells, interferon-γ (IFN-γ) production. Suppression required the expression and phosphorylation of Smad proteins in the TGF-ß signaling pathway, but not their nuclear translocation, and depended on oxygen availability, suggesting a metabolic basis for these effects. Smad proteins were detected in the mitochondria of CD4+ T cells, where they were phosphorylated upon treatment with TGF-ß. Phosphorylated Smad proteins were also detected in the mitochondria of isolated tumor-associated lymphocytes. TGF-ß substantially impaired the ATP-coupled respiration of CD4+ T cells and specifically inhibited mitochondrial complex V (ATP synthase) activity. Last, inhibition of ATP synthase alone was sufficient to impair IFN-γ production by CD4+ T cells. These results, which have implications for human antitumor immunity, suggest that TGF-ß targets T cell metabolism directly, thus diminishing T cell function through metabolic paralysis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interferon gama/imunologia , Mitocôndrias/imunologia , Neoplasias/imunologia , Consumo de Oxigênio/imunologia , Fator de Crescimento Transformador beta/imunologia , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Humanos , Interferon gama/metabolismo , Mitocôndrias/metabolismo , ATPases Mitocondriais Próton-Translocadoras/imunologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação/imunologia , Transdução de Sinais/imunologia , Proteínas Smad/imunologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral/imunologia
20.
Nat Rev Immunol ; 18(1): 19-34, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28944771

RESUMO

In healthy individuals, metabolically quiescent T cells survey lymph nodes and peripheral tissues in search of cognate antigens. During infection, T cells that encounter cognate antigens are activated and - in a context-specific manner - proliferate and/or differentiate to become effector T cells. This process is accompanied by important changes in cellular metabolism (known as metabolic reprogramming). The magnitude and spectrum of metabolic reprogramming as it occurs in T cells in the context of acute infection ensure host survival. By contrast, altered T cell metabolism, and hence function, is also observed in various disease states, in which T cells actively contribute to pathology. In this Review, we introduce the idea that the spectrum of immune cell metabolic states can provide a basis for categorizing human diseases. Specifically, we first summarize the metabolic and interlinked signalling requirements of T cells responding to acute infection. We then discuss how metabolic reprogramming of T cells is linked to disease.


Assuntos
Doenças Autoimunes/metabolismo , Infecções/metabolismo , Inflamação/metabolismo , Síndrome Metabólica/metabolismo , Neoplasias/metabolismo , Linfócitos T/metabolismo , Aminoácidos/metabolismo , Doenças Autoimunes/imunologia , Glucose/metabolismo , Humanos , Infecções/imunologia , Inflamação/imunologia , Metabolismo dos Lipídeos , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Síndrome Metabólica/imunologia , Neoplasias/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA