Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 611(7935): 405-412, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36323780

RESUMO

Solid tumours are innervated by nerve fibres that arise from the autonomic and sensory peripheral nervous systems1-5. Whether the neo-innervation of tumours by pain-initiating sensory neurons affects cancer immunosurveillance remains unclear. Here we show that melanoma cells interact with nociceptor neurons, leading to increases in their neurite outgrowth, responsiveness to noxious ligands and neuropeptide release. Calcitonin gene-related peptide (CGRP)-one such nociceptor-produced neuropeptide-directly increases the exhaustion of cytotoxic CD8+ T cells, which limits their capacity to eliminate melanoma. Genetic ablation of the TRPV1 lineage, local pharmacological silencing of nociceptors and antagonism of the CGRP receptor RAMP1 all reduced the exhaustion of tumour-infiltrating leukocytes and decreased the growth of tumours, nearly tripling the survival rate of mice that were inoculated with B16F10 melanoma cells. Conversely, CD8+ T cell exhaustion was rescued in sensory-neuron-depleted mice that were treated with local recombinant CGRP. As compared with wild-type CD8+ T cells, Ramp1-/- CD8+ T cells were protected against exhaustion when co-transplanted into tumour-bearing Rag1-deficient mice. Single-cell RNA sequencing of biopsies from patients with melanoma revealed that intratumoral RAMP1-expressing CD8+ T cells were more exhausted than their RAMP1-negative counterparts, whereas overexpression of RAMP1 correlated with a poorer clinical prognosis. Overall, our results suggest that reducing the release of CGRP from tumour-innervating nociceptors could be a strategy to improve anti-tumour immunity by eliminating the immunomodulatory effects of CGRP on cytotoxic CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos , Melanoma , Nociceptores , Animais , Camundongos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Melanoma/imunologia , Melanoma/patologia , Nociceptores/fisiologia , Células Receptoras Sensoriais/metabolismo , Neuritos/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Taxa de Sobrevida , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Genes RAG-1/genética , Humanos , Biópsia , Prognóstico
2.
J Vis Exp ; (184)2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35848838

RESUMO

Somatosensory neurons have evolved to detect noxious stimuli and activate defensive reflexes. By sharing means of communication, nociceptor neurons also tune host defenses by controlling the activity of the immune system. The communication between these systems is mostly adaptive, helping to protect homeostasis, it can also lead to, or promote, the onset of chronic diseases. Both systems co-evolved to allow for such local interaction, as found in primary and secondary lymphoid tissues and mucosa. Recent studies have demonstrated that nociceptors directly detect and respond to foreign antigens, immune cell-derived cytokines, and microbes. Nociceptor activation not only results in pain hypersensitivity and itching, but lowers the nociceptor firing threshold, leading to the local release of neuropeptides. The peptides that are produced by, and released from, the peripheral terminals of nociceptors can block the chemotaxis and polarization of lymphocytes, controlling the localization, duration, and type of inflammation. Recent evidence shows that sensory neurons interact with innate immune cells via cell-cell contact, for example, engaging group 2D (NKG2D) receptors on natural killer (NK) cells. Given that NK cells express the cognate receptors for various nociceptor-produced mediators, it is conceivable that nociceptors use neuropeptides to control the activity of NK cells. Here, we devise a co-culture method to study nociceptor neuron-NK cell interactions in a dish. Using this approach, we found that lumbar nociceptor neurons decrease NK cell cytokine expression. Overall, such a reductionist method could be useful to study how tumor-innervating neurons control the anticancer function of NK cells and how NK cells control the elimination of injured neurons.


Assuntos
Neuropeptídeos , Nociceptores , Citocinas/metabolismo , Humanos , Células Matadoras Naturais , Neuropeptídeos/metabolismo , Nociceptores/metabolismo , Dor , Células Receptoras Sensoriais/metabolismo
3.
Cell Rep Med ; 3(3): 100534, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35492876

RESUMO

The cross-presenting capacity of dendritic cells (DCs) can be limited by non-specific degradation during endosome maturation. To bypass this limitation, we present in this study a new Accum-based formulation designed to promote endosome-to-cytosol escape. Treatment of primary DCs with Accum linked to the xenoantigen ovalbumin (OVA) triggers endosomal damages and enhances protein processing. Despite multiple challenges using ascending doses of tumor cells, DC prophylactic vaccination results in complete protection due to increased levels of effector CD4 and CD8 T cells as well as high production of pro-inflammatory mediators. When combined with anti-PD-1, therapeutic vaccination using both syngeneic and allogeneic Accum-OVA-pulsed DCs triggers potent anti-tumoral responses. The net outcome culminates in increased CD11c, CD8, and NK infiltration along with a high CD8/Treg ratio. These highly favorable therapeutic effects highlight the promising potential of Accum as a distinct and potent technology platform suitable for the design of next generation cell cancer vaccines.


Assuntos
Vacinas Anticâncer , Células Dendríticas , Deriva e Deslocamento Antigênicos , Linfócitos T CD8-Positivos , Endossomos , Ovalbumina
4.
J Allergy Clin Immunol ; 147(6): 2330-2342, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33453289

RESUMO

BACKGROUND: Lung nociceptor neurons amplify immune cell activity and mucus metaplasia in response to an inhaled allergen challenge in sensitized mice. OBJECTIVE: We sought to identify the cellular mechanisms by which these sensory neurons are activated subsequent to allergen exposure. METHODS: We used calcium microscopy and electrophysiologic recording to assess whether vagal neurons directly respond to the model allergen ovalbumin (OVA). Next, we generated the first nociceptor-specific FcεR1γ knockdown (TRPV1Cre::FcεR1γfl/fl) mice to assess whether this targeted invalidation would affect the severity of allergic inflammation in response to allergen challenges. RESULTS: Lung-innervating jugular nodose complex ganglion neurons express the high-affinity IgE receptor FcεR1, the levels of which increase in OVA-sensitized mice. FcεR1γ-expressing vagal nociceptor neurons respond directly to OVA complexed with IgE with depolarization, action potential firing, calcium influx, and neuropeptide release. Activation of vagal neurons by IgE-allergen immune complexes, through the release of substance P from their peripheral terminals, directly amplifies TH2 cell influx and polarization in the airways. Allergic airway inflammation is decreased in TRPV1Cre::FcεR1γfl/fl mice and in FcεR1α-/- mice into which bone marrow has been transplanted. Finally, increased in vivo circulating levels of IgE following allergen sensitization enhances the responsiveness of FcεR1 to immune complexes in both mouse jugular nodose complex ganglion neurons and human induced pluripotent stem cell-derived nociceptors. CONCLUSIONS: Allergen sensitization triggers a feedforward inflammatory loop between IgE-producing plasma cells, FcεR1-expressing vagal sensory neurons, and TH2 cells, which helps to both initiate and amplify allergic airway inflammation. These data highlight a novel target for reducing allergy, namely, FcεR1γ expressed by nociceptors.


Assuntos
Expressão Gênica , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Receptores de IgE/genética , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Alérgenos/imunologia , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Predisposição Genética para Doença , Hipersensibilidade/genética , Hipersensibilidade/patologia , Camundongos , Camundongos Knockout , Neurônios/imunologia , Neurônios/metabolismo , Nociceptores/metabolismo , Ovalbumina/efeitos adversos , Ovalbumina/imunologia , Receptores de IgE/metabolismo , Mucosa Respiratória/patologia , Substância P/metabolismo , Nervo Vago
5.
Cell Rep Med ; 2(12): 100455, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-35028603

RESUMO

Dendritic cells (DCs) excel at cross-presenting antigens, but their effectiveness as cancer vaccine is limited. Here, we describe a vaccination approach using mesenchymal stromal cells (MSCs) engineered to express the immunoproteasome complex (MSC-IPr). Such modification instills efficient antigen cross-presentation abilities associated with enhanced major histocompatibility complex class I and CD80 expression, de novo production of interleukin-12, and higher chemokine secretion. This cross-presentation capacity of MSC-IPr is highly dependent on their metabolic activity. Compared with DCs, MSC-IPr hold the ability to cross-present a vastly different epitope repertoire, which translates into potent re-activation of T cell immunity against EL4 and A20 lymphomas and B16 melanoma tumors. Moreover, therapeutic vaccination of mice with pre-established tumors efficiently controls cancer growth, an effect further enhanced when combined with antibodies targeting PD-1, CTLA4, LAG3, or 4-1BB under both autologous and allogeneic settings. Therefore, MSC-IPr constitute a promising subset of non-hematopoietic antigen-presenting cells suitable for designing universal cell-based cancer vaccines.


Assuntos
Vacinas Anticâncer/imunologia , Linfoma/imunologia , Melanoma Experimental/imunologia , Células-Tronco Mesenquimais/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Engenharia de Proteínas , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Reprogramação Celular , Células Dendríticas/imunologia , Feminino , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade , Camundongos Endogâmicos C57BL , Fosforilação Oxidativa , Fenótipo , Vacinação
6.
Front Pharmacol ; 11: 237, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32231565

RESUMO

Phenotypic screening is an ideal strategy for the discovery of novel bioactive molecules. Using a customized high-throughput screening (HTS) assay employing primary T lymphocytes, we screened a small library of 4,398 compounds with unknown biological function/target to identify compounds eliciting immunomodulatory properties and discovered a sulfonyl-containing hit, we named InhiTinib. This compound inhibited interferon (IFN)-gamma production and proliferation of primary CD3+ T cells without inducing cell death. In contrast, InhiTinib triggered apoptosis in several murine and human cancer cell lines. Besides, the compound was well tolerated by immunocompetent mice, triggered tumor regression in animals with pre-established EL4 T-cell lymphomas, and prolonged the overall survival of mice harboring advanced tumors. Altogether, our data demonstrate the anti-cancer properties of InhiTinib, which can henceforth bridge to wider-scale biochemical and clinical tests following further in-depth pharmacodynamic studies.

7.
J Immunol ; 203(11): 2887-2898, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31659013

RESUMO

Dendritic cells (DCs) are critical players in skin homeostasis. A subset of mannose receptor (CD206)-expressing monocyte-derived DCs was found in skin, and their migratory counterpart is present in skin-draining lymph nodes (sdLNs). Skin CD206+ DCs were shown to upregulate MHC class II (MHCII) progressively, raising the question of whether this feature affects their biology. In this study, we assessed the role of MHCII regulation in the development and migration of these cells in mouse models expressing differential MHCII levels. Using CD206 as a surrogate marker, we found that skin CD206+ DCs develop in an MHCII-independent manner. However, their migration to sdLNs was affected by overexpression rather than absence or lower expression of MHCII. Accordingly, B16 tumor growth was exacerbated in mice overexpressing MHCII in the absence of ubiquitination. Mechanistically, CD206+ DCs from these mice showed decreased IRF4 and CCR7 expression. LPS, which is known to promote monocyte-derived DC recruitment to sdLNs, partially improved these defects. However, GM-CSF delivery restored CD206+ DC migration by promoting IRF4 expression. Collectively, these data show that MHCII downregulation is crucial for IRF4-dependent migration of CD206+ DCs to sdLNs in health and disease.


Assuntos
Movimento Celular , Células Dendríticas/metabolismo , Regulação para Baixo , Antígenos de Histocompatibilidade Classe II/metabolismo , Lectinas Tipo C/metabolismo , Linfonodos/metabolismo , Lectinas de Ligação a Manose/metabolismo , Receptores de Superfície Celular/metabolismo , Pele/metabolismo , Ubiquitinação , Animais , Receptor de Manose , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Front Neurosci ; 13: 25, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30766472

RESUMO

Diabetes is a common condition characterized by persistent hyperglycemia. High blood sugar primarily affects cells that have a limited capacity to regulate their glucose intake. These cells include capillary endothelial cells in the retina, mesangial cells in the renal glomerulus, Schwann cells, and neurons of the peripheral and central nervous systems. As a result, hyperglycemia leads to largely intractable complications such as retinopathy, nephropathy, hypertension, and neuropathy. Diabetic pain neuropathy is a complex and multifactorial disease that has been associated with poor glycemic control, longer diabetes duration, hypertension, advanced age, smoking status, hypoinsulinemia, and dyslipidemia. While many of the driving factors involved in diabetic pain are still being investigated, they can be broadly classified as either neuron -intrinsic or -extrinsic. In neurons, hyperglycemia impairs the polyol pathway, leading to an overproduction of reactive oxygen species and reactive nitrogen species, an enhanced formation of advanced glycation end products, and a disruption in Na+/K+ ATPase pump function. In terms of the extrinsic pathway, hyperglycemia leads to the generation of both overactive microglia and microangiopathy. The former incites a feed-forward inflammatory loop that hypersensitizes nociceptor neurons, as observed at the onset of diabetic pain neuropathy. The latter reduces neurons' access to oxygen, glucose and nutrients, prompting reductions in nociceptor terminal expression and losses in sensation, as observed in the later stages of diabetic pain neuropathy. Overall, microglia can be seen as potent and long-lasting amplifiers of nociceptor neuron activity, and may therefore constitute a potential therapeutic target in the treatment of diabetic pain neuropathy.

9.
Mult Scler Relat Disord ; 14: 29-31, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28619427

RESUMO

BACKGROUND: Soluble CD40 ligand (sCD154) is a proinflammatory and prothrombotic ligand belonging to the tumor necrosis factor family. It has been shown by a variety of studies that sCD154 is elevated in the serum of patients afflicted with system autoimmune diseases. The aim of our study was to address whether sCD154 is increased in disease affected by Multiple Sclerosis (MS). METHODS: Twenty MS patients who have been newly diagnosed as clinically definite multiple sclerosis (CDMS) along with twenty age and sex matched healthy individuals were recruited for this study. Serum cCD154 was measured by Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: The results showed no statistically meaningful difference between newly diagnosed MS patients (3.07ng/ul±0.66) and control group (2.95ng/ul±0.79; p=0.62) CONCLUSION: Regarding our study, it seems that soluble CD40 ligand derived from serum is not correlated with early stage of MS disease.


Assuntos
Ligante de CD40/sangue , Esclerose Múltipla/sangue , Adulto , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Esclerose Múltipla/fisiopatologia , Índice de Gravidade de Doença , Adulto Jovem
10.
J Neuroimmunol ; 273(1-2): 120-3, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24984830

RESUMO

Autotaxin (ATX) is an enzyme producing lysophosphatidic acid (LPA) from lysophosphatidyl choline (LPC) and it is up-regulated in inflammatory conditions such as various cancers, arthritis and multiple sclerosis (MS). Numerous studies have shown that the LPA signaling gives rise to angiogenesis, mitosis, cell proliferation and cytokine secretion. On the one hand, an increasing body of evidence suggests that blockade of ATX has anti-inflammatory properties in a variety of diseases. The aim of this study was to measure the enzyme activity of ATX in cerebrospinal fluid (CSF) and serum of patients with MS using an enzymatic photometric method. Twenty definite relapsing remitting MS patients along with 20 patients with other neurological diseases (OND) were recruited. The results showed that ATX activity was significantly higher (p value<0.0001) in MS patients than those patients diagnosed with OND. It is possible that inhibition of the ATX may decrease the rate of MS relapses/progression.


Assuntos
Esclerose Múltipla/líquido cefalorraquidiano , Diester Fosfórico Hidrolases/líquido cefalorraquidiano , Adulto , Feminino , Humanos , Masculino , Esclerose Múltipla/sangue , Esclerose Múltipla/complicações , Doenças do Sistema Nervoso/etiologia , Diester Fosfórico Hidrolases/sangue , Punção Espinal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA