Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32822574

RESUMO

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Bactérias/imunologia , Movimento Celular/imunologia , Células Matadoras Naturais/imunologia , Trofoblastos/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Células HeLa , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placenta/imunologia , Placenta/microbiologia , Gravidez , Ratos , Células THP-1 , Trofoblastos/microbiologia
2.
Cell ; 171(5): 1125-1137.e11, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29107333

RESUMO

Human cytotoxic lymphocytes kill intracellular microbes. The cytotoxic granule granzyme proteases released by cytotoxic lymphocytes trigger oxidative bacterial death by disrupting electron transport, generating superoxide anion and inactivating bacterial oxidative defenses. However, they also cause non-oxidative cell death because anaerobic bacteria are also killed. Here, we use differential proteomics to identify granzyme B substrates in three unrelated bacteria: Escherichia coli, Listeria monocytogenes, and Mycobacteria tuberculosis. Granzyme B cleaves a highly conserved set of proteins in all three bacteria, which function in vital biosynthetic and metabolic pathways that are critical for bacterial survival under diverse environmental conditions. Key proteins required for protein synthesis, folding, and degradation are also substrates, including multiple aminoacyl tRNA synthetases, ribosomal proteins, protein chaperones, and the Clp system. Because killer cells use a multipronged strategy to target vital pathways, bacteria may not easily become resistant to killer cell attack.


Assuntos
Escherichia coli/citologia , Granzimas/metabolismo , Células Matadoras Naturais/enzimologia , Listeria monocytogenes/citologia , Mycobacterium tuberculosis/citologia , Linfócitos T Citotóxicos/enzimologia , Aminoacil-tRNA Sintetases/metabolismo , Animais , Escherichia coli/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Listeria monocytogenes/metabolismo , Redes e Vias Metabólicas , Camundongos , Mycobacterium tuberculosis/metabolismo , Biossíntese de Proteínas , Proteômica , Ribossomos/metabolismo , Linfócitos T Citotóxicos/imunologia
3.
Cell ; 165(1): 100-110, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26924577

RESUMO

The immunological synapse formed between a cytotoxic T lymphocyte (CTL) and an infected or transformed target cell is a physically active structure capable of exerting mechanical force. Here, we investigated whether synaptic forces promote the destruction of target cells. CTLs kill by secreting toxic proteases and the pore forming protein perforin into the synapse. Biophysical experiments revealed a striking correlation between the magnitude of force exertion across the synapse and the speed of perforin pore formation on the target cell, implying that force potentiates cytotoxicity by enhancing perforin activity. Consistent with this interpretation, we found that increasing target cell tension augmented pore formation by perforin and killing by CTLs. Our data also indicate that CTLs coordinate perforin release and force exertion in space and time. These results reveal an unappreciated physical dimension to lymphocyte function and demonstrate that cells use mechanical forces to control the activity of outgoing chemical signals.


Assuntos
Sinapses Imunológicas , Linfócitos T Citotóxicos/fisiologia , Animais , Fenômenos Biomecânicos , Degranulação Celular , Linhagem Celular Tumoral , Camundongos , Perforina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia
4.
Nat Immunol ; 19(5): 475-486, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29670239

RESUMO

CD4+ T lymphocytes are the principal target of human immunodeficiency virus (HIV), but infected macrophages also contribute to viral pathogenesis. The killing of infected cells by CD8+ cytotoxic T lymphocytes (CTLs) leads to control of viral replication. Here we found that the killing of macrophages by CTLs was impaired relative to the killing of CD4+ T cells by CTLs, and this resulted in inefficient suppression of HIV. The killing of macrophages depended on caspase-3 and granzyme B, whereas the rapid killing of CD4+ T cells was caspase independent and did not require granzyme B. Moreover, the impaired killing of macrophages was associated with prolonged effector cell-target cell contact time and higher expression of interferon-γ by CTLs, which induced macrophage production of pro-inflammatory chemokines that recruited monocytes and T cells. Similar results were obtained when macrophages presented other viral antigens, suggestive of a general mechanism for macrophage persistence as antigen-presenting cells that enhance inflammation and adaptive immunity. Inefficient killing of macrophages by CTLs might contribute to chronic inflammation, a hallmark of chronic disease caused by HIV.


Assuntos
Linfócitos T CD4-Positivos/virologia , Citotoxicidade Imunológica/imunologia , Infecções por HIV/imunologia , Macrófagos/virologia , Linfócitos T Citotóxicos/imunologia , Células Cultivadas , Humanos
5.
Cell ; 157(6): 1309-1323, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24906149

RESUMO

When killer lymphocytes recognize infected cells, perforin delivers cytotoxic proteases (granzymes) into the target cell to trigger apoptosis. What happens to intracellular bacteria during this process is unclear. Human, but not rodent, cytotoxic granules also contain granulysin, an antimicrobial peptide. Here, we show that granulysin delivers granzymes into bacteria to kill diverse bacterial strains. In Escherichia coli, granzymes cleave electron transport chain complex I and oxidative stress defense proteins, generating reactive oxygen species (ROS) that rapidly kill bacteria. ROS scavengers and bacterial antioxidant protein overexpression inhibit bacterial death. Bacteria overexpressing a GzmB-uncleavable mutant of the complex I subunit nuoF or strains that lack complex I still die, but more slowly, suggesting that granzymes disrupt multiple vital bacterial pathways. Mice expressing transgenic granulysin are better able to clear Listeria monocytogenes. Thus killer cells play an unexpected role in bacterial defense.


Assuntos
Antígenos de Diferenciação de Linfócitos T/metabolismo , Infecções Bacterianas/imunologia , Escherichia coli , Leucócitos Mononucleares/imunologia , Listeria monocytogenes , Staphylococcus aureus , Animais , Granzimas/metabolismo , Células HeLa , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Perforina/genética , Perforina/metabolismo , Espécies Reativas de Oxigênio/metabolismo
6.
Nature ; 589(7843): 597-602, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33361818

RESUMO

Isoprenoids are vital for all organisms, in which they maintain membrane stability and support core functions such as respiration1. IspH, an enzyme in the methyl erythritol phosphate pathway of isoprenoid synthesis, is essential for Gram-negative bacteria, mycobacteria and apicomplexans2,3. Its substrate, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), is not produced in metazoans, and in humans and other primates it activates cytotoxic Vγ9Vδ2 T cells at extremely low concentrations4-6. Here we describe a class of IspH inhibitors and refine their potency to nanomolar levels through structure-guided analogue design. After modification of these compounds into prodrugs for delivery into bacteria, we show that they kill clinical isolates of several multidrug-resistant bacteria-including those from the genera Acinetobacter, Pseudomonas, Klebsiella, Enterobacter, Vibrio, Shigella, Salmonella, Yersinia, Mycobacterium and Bacillus-yet are relatively non-toxic to mammalian cells. Proteomic analysis reveals that bacteria treated with these prodrugs resemble those after conditional IspH knockdown. Notably, these prodrugs also induce the expansion and activation of human Vγ9Vδ2 T cells in a humanized mouse model of bacterial infection. The prodrugs we describe here synergize the direct killing of bacteria with a simultaneous rapid immune response by cytotoxic γδ T cells, which may limit the increase of antibiotic-resistant bacterial populations.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/imunologia , Ativação Linfocitária/efeitos dos fármacos , Viabilidade Microbiana/efeitos dos fármacos , Oxirredutases/antagonistas & inibidores , Linfócitos T Citotóxicos/efeitos dos fármacos , Animais , Resistência Microbiana a Medicamentos , Resistência a Múltiplos Medicamentos , Inibidores Enzimáticos/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Feminino , Meia-Vida , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Oxirredutases/deficiência , Oxirredutases/genética , Oxirredutases/metabolismo , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Especificidade por Substrato , Suínos/sangue , Linfócitos T Citotóxicos/imunologia
7.
Nat Immunol ; 14(2): 179-85, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23263557

RESUMO

The innate immune system senses viral DNA that enters mammalian cells, or in aberrant situations self-DNA, and triggers type I interferon production. Here we present an integrative approach that combines quantitative proteomics, genomics and small molecule perturbations to identify genes involved in this pathway. We silenced 809 candidate genes, measured the response to dsDNA and connected resulting hits with the known signaling network. We identified ABCF1 as a critical protein that associates with dsDNA and the DNA-sensing components HMGB2 and IFI204. We also found that CDC37 regulates the stability of the signaling molecule TBK1 and that chemical inhibition of the CDC37-HSP90 interaction and several other pathway regulators potently modulates the innate immune response to DNA and retroviral infection.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , DNA Viral/imunologia , Células Dendríticas/imunologia , Fibroblastos/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imunidade Inata , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Chaperoninas/antagonistas & inibidores , Chaperoninas/genética , Chaperoninas/imunologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Citosol/virologia , DNA Viral/genética , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Regulação da Expressão Gênica/imunologia , Inativação Gênica , HIV-1/fisiologia , Proteína HMGB2/genética , Proteína HMGB2/imunologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Fosfoproteínas/genética , Fosfoproteínas/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteômica , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Bibliotecas de Moléculas Pequenas/farmacologia , Vesiculovirus/fisiologia
10.
Proc Natl Acad Sci U S A ; 110(1): E41-9, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23169651

RESUMO

Budding yeast cells suffering a single unrepaired double-strand break (DSB) trigger the Mec1 (ATR)-dependent DNA damage response that causes them to arrest before anaphase for 12-15 h. Here we find that hyperactivation of the cytoplasm-to-vacuole (CVT) autophagy pathway causes the permanent G2/M arrest of cells with a single DSB that is reflected in the nuclear exclusion of both Esp1 and Pds1. Transient relocalization of Pds1 is also seen in wild-type cells lacking vacuolar protease activity after induction of a DSB. Arrest persists even as the DNA damage-dependent phosphorylation of Rad53 diminishes. Permanent arrest can be overcome by blocking autophagy, by deleting the vacuolar protease Prb1, or by driving Esp1 into the nucleus with a SV40 nuclear localization signal. Autophagy in response to DNA damage can be induced in three different ways: by deleting the Golgi-associated retrograde protein complex (GARP), by adding rapamycin, or by overexpression of a dominant ATG13-8SA mutation.


Assuntos
Anáfase/fisiologia , Autofagia/fisiologia , Pontos de Checagem do Ciclo Celular/fisiologia , Quebras de DNA de Cadeia Dupla , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Endopeptidases/metabolismo , Proteínas de Fluorescência Verde , Proteínas Nucleares/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Saccharomycetales , Securina , Separase , Sirolimo/farmacologia
11.
Nat Cell Biol ; 8(9): 1032-4, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16892052

RESUMO

DNA double-strand breaks (DSB) can arise during DNA replication, or after exposure to DNA-damaging agents, and their correct repair is fundamental for cell survival and genomic stability. Here, we show that the Smc5-Smc6 complex is recruited to DSBs de novo to support their repair by homologous recombination between sister chromatids. In addition, we demonstrate that Smc5-Smc6 is necessary to suppress gross chromosomal rearrangements. Our findings show that the Smc5-Smc6 complex is essential for genome stability as it promotes repair of DSBs by error-free sister-chromatid recombination (SCR), thereby suppressing inappropriate non-sister recombination events.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Dano ao DNA , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Troca de Cromátide Irmã , DNA/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Instabilidade Genômica , Saccharomyces cerevisiae/genética
12.
Vaccines (Basel) ; 11(10)2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37896988

RESUMO

Inducing humoral and cytotoxic mucosal immunity at the sites of pathogen entry has the potential to prevent the infection from getting established. This is different from systemic vaccination, which protects against the development of systemic symptoms. The field of mucosal vaccination has seen fewer technological advances compared to nucleic acid and subunit vaccine advances for injectable vaccine platforms. The advent of the next-generation adenoviral vectors has given a boost to mucosal vaccine research. Basic research into the mechanisms regulating innate and adaptive mucosal immunity and the discovery of effective and safe mucosal vaccine adjuvants will continue to improve mucosal vaccine design. The results from clinical trials of inhaled COVID-19 vaccines demonstrate their ability to induce the proliferation of cytotoxic T cells and the production of secreted IgA and IgG antibodies locally, unlike intramuscular vaccinations. However, these mucosal vaccines induce systemic immune responses at par with systemic vaccinations. This review summarizes the function of the respiratory mucosa-associated lymphoid tissue and the advantages that the adenoviral vectors provide as inhaled vaccine platforms.

13.
Invest Ophthalmol Vis Sci ; 64(7): 39, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37389545

RESUMO

Since long before the first approval of gene therapy for retinal disease, ocular gene therapy has captured the hopes of patients, clinicians, and scientists alike. Indeed, the retina provides a unique system for studying and treating ocular diseases, and it holds the distinction as the first tissue targeted by an approved gene therapy for inherited disorders in the United States. There are many methods for addressing genetic diseases in the eyes using a wide range of potential delivery systems and vectors. However, despite the immense progress over the last several decades, both old and new challenges remain, such as the long-term effects of treatments, immunogenicity, targeting, and manufacturing. This review provides a discussion of the history of ocular gene therapy, the various gene therapy approaches, methods to deliver a gene directly to ocular tissues (including both routes of administration and vectors), challenges to ocular gene therapy, the current clinical trial landscape, and future directions of the field.


Assuntos
Degeneração Retiniana , Humanos , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Retina , Terapia Genética
14.
J Cell Biol ; 178(2): 209-18, 2007 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-17635934

RESUMO

Double-strand break (DSB) damage in yeast and mammalian cells induces the rapid ATM (ataxia telangiectasia mutated)/ATR (ataxia telangiectasia and Rad3 related)-dependent phosphorylation of histone H2AX (gamma-H2AX). In budding yeast, a single endonuclease-induced DSB triggers gamma-H2AX modification of 50 kb on either side of the DSB. The extent of gamma-H2AX spreading does not depend on the chromosomal sequences. DNA resection after DSB formation causes the slow, progressive loss of gamma-H2AX from single-stranded DNA and, after several hours, the Mec1 (ATR)-dependent spreading of gamma-H2AX to more distant regions. Heterochromatic sequences are only weakly modified upon insertion of a 3-kb silent HMR locus into a gamma-H2AX-covered region. The presence of heterochromatin does not stop the phosphorylation of chromatin more distant from the DSB. In mouse embryo fibroblasts, gamma-H2AX distribution shows that gamma-H2AX foci increase in size as chromatin becomes more accessible. In yeast, we see a high level of constitutive gamma-H2AX in telomere regions in the absence of any exogenous DNA damage, suggesting that yeast chromosome ends are transiently detected as DSBs.


Assuntos
Dano ao DNA , Heterocromatina/metabolismo , Histonas/metabolismo , Mamíferos/metabolismo , Saccharomyces cerevisiae/metabolismo , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Cromossomos de Mamíferos , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Histonas/química , Histonas/genética , Mamíferos/genética , Camundongos , Mutação , Fosforilação , Saccharomyces cerevisiae/genética
15.
Front Immunol ; 13: 940715, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36177016

RESUMO

The world has responded to the COVID-19 pandemic with unprecedented speed and vigor in the mass vaccination campaigns, targeted to reduce COVID-19 severity and mortality, reduce the pressure on the healthcare system, re-open society, and reduction in disease mortality and morbidity. Here we review the preclinical and clinical development of BBV152, a whole virus inactivated vaccine and an important tool in the fight to control this pandemic. BBV152, formulated with a TLR7/8 agonist adjuvant generates a Th1-biased immune response that induces high neutralization efficacy against different SARS-CoV-2 variants of concern and robust long-term memory B- and T-cell responses. With seroconversion rates as high as 98.3% in vaccinated individuals, BBV152 shows 77.8% and 93.4% protection from symptomatic COVID-19 disease and severe symptomatic COVID-19 disease respectively. Studies in pediatric populations show superior immunogenicity (geometric mean titer ratio of 1.76 compared to an adult) with a seroconversion rate of >95%. The reactogenicity and safety profiles were comparable across all pediatric age groups between 2-18 yrs. as in adults. Like most approved vaccines, the BBV152 booster given 6 months after full vaccination, reverses a waning immunity, restores the neutralization efficacy, and shows synergy in a heterologous prime-boost study with about 3-fold or 300% increase in neutralization titers against multiple SARS-CoV-2 variants of concern. Based on the interim Phase III data, BBV152 received full authorization for adults and emergency use authorization for children from ages 6 to 18 years in India. It is also licensed for emergency use in 14 countries globally. Over 313 million vaccine doses have already been administered in India alone by April 18th, 2022.


Assuntos
COVID-19 , SARS-CoV-2 , Adjuvantes Imunológicos , Adolescente , Adulto , COVID-19/prevenção & controle , Vacinas contra COVID-19/efeitos adversos , Criança , Pré-Escolar , Humanos , Pandemias/prevenção & controle , Receptor 7 Toll-Like , Desenvolvimento de Vacinas , Vacinas de Produtos Inativados/efeitos adversos
16.
J Cancer ; 13(6): 1933-1944, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35399717

RESUMO

In this study, we evaluated the ability of negatively charged bio-degradable nanoparticles, ONP- 302, to inhibit tumor growth. Therapeutic treatment with ONP-302 in vivo resulted in a marked delay in tumor growth in three different syngeneic tumor models in immunocompetent mice. ONP- 302 efficacy persisted with depletion of CD8+ T cells in immunocompetent mice and also was effective in immune deficient mice. Examination of ONP-302 effects on components of the tumor microenvironment (TME) were explored. ONP-302 treatment caused a gene expression shift in TAMs toward the pro-inflammatory M1 type and substantially inhibited the expression of genes associated with the pro-tumorigenic function of CAFs. ONP-302 also induced apoptosis in CAFs in the TME. Together, these data support further development of ONP-302 as a novel first-in- class anti-cancer therapeutic that can be used as a single-agent as well as in combination therapies for the treatment of solid tumors due to its ability to modulate the TME.

17.
Nat Commun ; 12(1): 346, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436641

RESUMO

Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Linfócitos do Interstício Tumoral/imunologia , Mastócitos/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Melanoma/imunologia , Melanoma/patologia , Melanoma/terapia , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/metabolismo , Sunitinibe/farmacologia , Sunitinibe/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
19.
Nat Commun ; 11(1): 473, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980600

RESUMO

A variant at amino acid 47 in human TP53 exists predominantly in individuals of African descent. P47S human and mouse cells show increased cancer risk due to defective ferroptosis. Here, we show that this ferroptotic defect causes iron accumulation in P47S macrophages. This high iron content alters macrophage cytokine profiles, leads to higher arginase level and activity, and decreased nitric oxide synthase activity. This leads to more productive intracellular bacterial infections but is protective against malarial toxin hemozoin. Proteomics of macrophages reveal decreased liver X receptor (LXR) activation, inflammation and antibacterial defense in P47S macrophages. Both iron chelators and LXR agonists improve the response of P47S mice to bacterial infection. African Americans with elevated saturated transferrin and serum ferritin show higher prevalence of the P47S variant (OR = 1.68 (95%CI 1.07-2.65) p = 0.023), suggestive of its role in iron accumulation in humans. This altered macrophage phenotype may confer an advantage in malaria-endemic sub-Saharan Africa.


Assuntos
Ferro/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , África Subsaariana , Negro ou Afro-Americano/genética , Animais , Infecções Bacterianas/etiologia , Infecções Bacterianas/genética , Infecções Bacterianas/metabolismo , Ferritinas/sangue , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Ferroptose/fisiologia , Variação Genética , Hemeproteínas/toxicidade , Humanos , Listeriose/etiologia , Receptores X do Fígado/agonistas , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Malária/genética , Malária/metabolismo , Camundongos , Camundongos Transgênicos , Transferrina/metabolismo
20.
Curr Opin Immunol ; 60: 19-29, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31112765

RESUMO

Primary T cell immunodeficiency and HIV-infected patients are plagued by non-viral infections caused by bacteria, fungi, and parasites, suggesting an important and underappreciated role for T lymphocytes in controlling microbes. Here, we review recent studies showing that killer lymphocytes use the antimicrobial cytotoxic granule pore-forming peptide granulysin, induced by microbial exposure, to permeabilize cholesterol-poor microbial membranes and deliver death-inducing granzymes into these pathogens. Granulysin and granzymes cause microptosis, programmed cell death in microbes, by inducing reactive oxygen species and destroying microbial antioxidant defenses and disrupting biosynthetic and central metabolism pathways required for their survival, including protein synthesis, glycolysis, and the Krebs cycle.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Bactérias/imunologia , Fungos/imunologia , Células Matadoras Naturais/imunologia , Parasitos/imunologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa