Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 987
Filtrar
1.
Nature ; 623(7989): 1053-1061, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37844613

RESUMO

Inflammation is a hallmark of cancer1. In patients with cancer, peripheral blood myeloid expansion, indicated by a high neutrophil-to-lymphocyte ratio, associates with shorter survival and treatment resistance across malignancies and therapeutic modalities2-5. Whether myeloid inflammation drives progression of prostate cancer in humans remain unclear. Here we show that inhibition of myeloid chemotaxis can reduce tumour-elicited myeloid inflammation and reverse therapy resistance in a subset of patients with metastatic castration-resistant prostate cancer (CRPC). We show that a higher blood neutrophil-to-lymphocyte ratio reflects tumour myeloid infiltration and tumour expression of senescence-associated mRNA species, including those that encode myeloid-chemoattracting CXCR2 ligands. To determine whether myeloid cells fuel resistance to androgen receptor signalling inhibitors, and whether inhibiting CXCR2 to block myeloid chemotaxis reverses this, we conducted an investigator-initiated, proof-of-concept clinical trial of a CXCR2 inhibitor (AZD5069) plus enzalutamide in patients with metastatic CRPC that is resistant to androgen receptor signalling inhibitors. This combination was well tolerated without dose-limiting toxicity and it decreased circulating neutrophil levels, reduced intratumour CD11b+HLA-DRloCD15+CD14- myeloid cell infiltration and imparted durable clinical benefit with biochemical and radiological responses in a subset of patients with metastatic CRPC. This study provides clinical evidence that senescence-associated myeloid inflammation can fuel metastatic CRPC progression and resistance to androgen receptor blockade. Targeting myeloid chemotaxis merits broader evaluation in other cancers.


Assuntos
Antagonistas de Receptores de Andrógenos , Antineoplásicos , Quimiotaxia , Resistencia a Medicamentos Antineoplásicos , Células Mieloides , Neoplasias de Próstata Resistentes à Castração , Humanos , Masculino , Quimiotaxia/efeitos dos fármacos , Progressão da Doença , Inflamação/tratamento farmacológico , Inflamação/patologia , Antígenos CD15/metabolismo , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Metástase Neoplásica , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Andrógenos/farmacologia , Antagonistas de Receptores de Andrógenos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
2.
Benef Microbes ; 14(4): 401-419, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38661366

RESUMO

The intestinal microbiota contributes to gut immune homeostasis, where short-chain fatty acids (SCFAs) function as the major mediators. We aimed to elucidate the immunomodulatory effects of acetate, propionate, and butyrate. With that in mind, we sought to characterise the expression of SCFA receptors and transporters as well as SCFAs' impact on the activation of different immune cells. Whereas all three SCFAs decreased tumour necrosis factor (TNF)-α production in activated T cells, only butyrate and propionate inhibited interferon (IFN)-γ, interleukin (IL)-17, IL-13, and IL-10 production. Butyrate and propionate inhibited the expression of the chemokine receptors CCR9 and CCR10 in activated T- and B-cells, respectively. Similarly, butyrate and propionate were effective inhibitors of IL-1ß, IL-6, TNF-α, and IL-10 production in myeloid cells upon lipopolysaccharide and R848 stimulation. Acetate was less efficient at inhibiting cytokine production except for IFN-α. Moreover, SCFAs inhibited the production of IL-6 and TNF-α in monocytes, myeloid dendritic cells (mDC), and plasmacytoid dendritic cells (pDC), whereas acetate effects were relatively more prominent in pDCs. In monocytes and mDCs, acetate was a less efficient inhibitor, but it was equally effective in inhibiting pDCs activation. We also studied the ability of SCFAs to induce trained immunity or tolerance. Butyrate and propionate - but not acetate - prevented Toll-like receptor-mediated activation in SCFA-trained cells, as demonstrated by a reduced production of IL-6 and TNF-α. Our findings indicate that butyrate and propionate are equally efficient in inhibiting the adaptive and innate immune response and did not induce trained immunity. The findings may be explained by differential SCFA receptor and transporter expression profiles of the immune cells.


Assuntos
Citocinas , Ácidos Graxos Voláteis , Tolerância Imunológica , Imunidade Inata , Linfócitos T , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia , Humanos , Imunidade Inata/efeitos dos fármacos , Citocinas/metabolismo , Citocinas/imunologia , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Butiratos/farmacologia , Células Mieloides/imunologia , Células Mieloides/efeitos dos fármacos , Propionatos/farmacologia , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Monócitos/imunologia , Monócitos/efeitos dos fármacos
3.
Am J Manag Care ; 28(16 Suppl): S323-S328, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36548523

RESUMO

Multiple sclerosis (MS) is a chronic, immune-mediated, neurodegenerative condition that results in progressive accumulation of disability over the course of the disease. MS presents heterogeneously, and, as the disease progresses, patients develop a range of physical and neurologic problems that include reduced mobility, cognitive impairment, weakness, fatigue, pain, and defects in speech or vision. Economically, MS is costly, including both direct costs stemming from clinical care and medications and the indirect costs of productivity losses. These costs pose a substantial burden to patients, families, caregivers, employers, and society. There are 21 approved disease-modifying therapies for MS across several drug classes. The importance of early MS treatment has been confirmed, and progress has been made in the treatment of relapsing-remitting MS, although this progress has not been replicated for progressive presentations of the disease. Ongoing research continues to elucidate the exact mechanisms of disease in MS as well as potential new treatment strategies that may better address current gaps, such as disability progression in secondary progressive MS without activity. One of the novel pathways under investigation is the inhibition of Bruton tyrosine kinase, a cytoplasmic tyrosine kinase, which is expressed in B cells and other potentially targetable hematopoietic lineage cells. This review examines emerging hypotheses that targeting both B cells and myeloid cells within the periphery and central nervous system could yield clinical effects in key areas of MS pathophysiology that are currently unaddressed.


Assuntos
Tirosina Quinase da Agamaglobulinemia , Esclerose Múltipla , Humanos , Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Tirosina Quinase da Agamaglobulinemia/metabolismo , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/enzimologia , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/enzimologia , Redes e Vias Metabólicas , Linfócitos B/efeitos dos fármacos , Linfócitos B/enzimologia , Células Mieloides/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
4.
Proc Natl Acad Sci U S A ; 119(14): e2111804119, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35353625

RESUMO

The receptor for colony stimulating factor 1 (CSF-1R) is important for the survival and function of myeloid cells that mediate pathology during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). CSF-1 and IL-34, the ligands of CSF-1R, have similar bioactivities but distinct tissue and context-dependent expression patterns, suggesting that they have different roles. This could be the case in EAE, given that CSF-1 expression is up-regulated in the CNS, while IL-34 remains constitutively expressed. We found that targeting CSF-1 with neutralizing antibody halted ongoing EAE, with efficacy superior to CSF-1R inhibitor BLZ945, whereas IL-34 neutralization had no effect, suggesting that pathogenic myeloid cells were maintained by CSF-1. Both anti­CSF-1 and BLZ945 treatment greatly reduced the number of monocyte-derived cells and microglia in the CNS. However, anti­CSF-1 selectively depleted inflammatory microglia and monocytes in inflamed CNS areas, whereas BLZ945 depleted virtually all myeloid cells, including quiescent microglia, throughout the CNS. Anti­CSF-1 treatment reduced the size of demyelinated lesions and microglial activation in the gray matter. Lastly, we found that bone marrow­derived immune cells were the major mediators of CSF-1R­dependent pathology, while microglia played a lesser role. Our findings suggest that targeting CSF-1 could be effective in ameliorating MS pathology, while preserving the homeostatic functions of myeloid cells, thereby minimizing risks associated with ablation of CSF-1R­dependent cells.


Assuntos
Encefalomielite Autoimune Experimental , Fator Estimulador de Colônias de Macrófagos , Esclerose Múltipla , Animais , Benzotiazóis/farmacologia , Benzotiazóis/uso terapêutico , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Ácidos Picolínicos/farmacologia , Ácidos Picolínicos/uso terapêutico , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores
5.
Inflamm Res ; 71(2): 169-182, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34999919

RESUMO

Ethyl pyruvate (EP) has potent influence on redox processes, cellular metabolism, and inflammation. It has been intensively studied in numerous animal models of systemic and organ-specific disorders whose pathogenesis involves a strong immune component. Here, basic chemical and biological properties of EP are discussed, with an emphasis on its redox and metabolic activity. Further, its influence on myeloid and T cells is considered, as well as on intracellular signaling beyond its effect on immune cells. Also, the effects of EP on animal models of chronic inflammatory and autoimmune disorders are presented. Finally, a possibility to apply EP as a treatment for such diseases in humans is discussed. Scientific papers cited in this review were identified using the PubMed search engine that relies on the MEDLINE database. The reference list covers the most important findings in the field in the past twenty years.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Inflamação/tratamento farmacológico , Piruvatos/uso terapêutico , Animais , Modelos Animais de Doenças , Humanos , Células Mieloides/efeitos dos fármacos , Piruvatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
6.
J Neurooncol ; 156(3): 625-634, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35037156

RESUMO

PURPOSE: Nearly 10% of patients with adult diffuse glioma develop clinically significant myelotoxicity while on temozolomide (TMZ) leading to treatment interruptions. This study aimed to assess single nucleotide polymorphisms (SNPs) in the O6-methylguanine-DNA methyltransferase (MGMT) gene in adults with biopsy-proven diffuse glioma who develop TMZ-induced myelotoxicity and correlate their presence with severity and duration of such toxicity. METHODS: This study assessed 33 adults treated with TMZ for diffuse glioma who developed ≥ grade 2 thrombocytopenia and/or ≥ grade 3 neutropenia. Genomic DNA was extracted from peripheral blood cells for MGMT SNP analysis after written informed consent. TMZ-induced severe myelotoxicity (≥ grade 3) was correlated with three specified SNPs commonly seen in the MGMT gene (L84F, I143V/K178R) using chi-square test or Fischer's exact test as appropriate. RESULTS: Of the 33 adults, 24 (72.7%) experienced ≥ grade 3 thrombocytopenia and/or neutropenia, while 9 (27.3%) developed grade 2 thrombocytopenia only. The variant T allele of L84F was expressed in 28.7% (19/66) of analyzed alleles, which was substantially higher than previously reported for South Asian ancestry. The variant G allele of I143V/K178R was expressed in 9.3% (6/64) of analyzed alleles. Of which 3 patients showed statistically significant association with prolonged myelosuppression for > 2 months (p = 0.03). No significant correlation was established between the mentioned SNPs and severe myelotoxicity. CONCLUSIONS: There is substantially higher frequency of variant T allele (L84F) in Indian patients than previously reported for South Asians. The presence of specific SNPs in the MGMT gene correlates with prolonged duration but not severity of TMZ-induced myelotoxicity.


Assuntos
Neoplasias Encefálicas , Metilases de Modificação do DNA , Enzimas Reparadoras do DNA , Glioma , Temozolomida , Proteínas Supressoras de Tumor , Adulto , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Glioma/tratamento farmacológico , Glioma/genética , Humanos , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Testes Farmacogenômicos , Polimorfismo de Nucleotídeo Único , Temozolomida/efeitos adversos , Proteínas Supressoras de Tumor/genética
7.
Mol Pharm ; 19(1): 246-257, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34816721

RESUMO

Gastric cancer (GC) is a common cancer worldwide, with high incidence and mortality rates. Therefore, early and precise diagnosis is critical to improving GC prognosis. Tumor-associated myeloid cells infiltrate the tumor microenvironment (TME) and can produce immunosuppressive effects in the early stage of the tumor. The surface integrin receptor CD11b is widely expressed in the specific subsets of myeloid cells, and it has the characteristics of high abundance, high specificity, and high potential for targeted immunotherapy. In this study, two strategies for labeling anti-CD11b, including 89Zr-DFO-anti-CD11b and pretargeted imaging (68Ga-NOTA-polypeptide-PEG11-Tz/anti-CD11b-TCO), were used to evaluate the value of early diagnosis of GC and confirm the advantages of the pretargeted strategy for the diagnosis of GC. Pretargeted molecular probe 68Ga-NOTA-polypeptide-PEG11-Tz was synthesized. The binding affinity of the Tz-radioligand to CD11b was evaluated in vitro, and its blood pharmacokinetic test was performed in vivo. Moreover, the anti-CD11b antibody was conjugated with a p-isothiocyanatobenzyl-desferrioxamine (SCN-DFO) chelator and radiolabeled with zirconium-89. Biodistribution and positron-emission computed tomography imaging experiments were performed in MGC-803 tumor-bearing model mice to evaluate the value of the early diagnosis of GC. Histological evaluation of MGC-803 tumors was conducted to confirm the infiltration of the GC TME with CD11b+ myeloid cells. 68Ga-NOTA-polypeptide-PEG11-Tz was successfully radiosynthesized, with the radiochemical purity above 95%, as confirmed by reversed-phase high-performance liquid chromatography. The radioligand showed favorable stability in normal saline and phosphate-buffered saline, good affinity to RAW264.7 cells, and rapid blood clearance in mice. The results of biodistribution and imaging experiments using the pretargeted method showed that the tumor/muscle ratios were 5.17 ± 2.98, 5.94 ± 1.46, and 4.46 ± 2.73 at the pretargeting intervals of 24, 48, and 72 h, respectively. The experimental results using the method of the directly labeling antibody (89Zr-DFO-anti-CD11b) showed that, despite radioactive accumulation in the tumor, there was a higher level of radioactive accumulation in normal tissues. The tumor/muscle ratios were 1.09 ± 0.67, 1.66 ± 0.95, 2.94 ± 1.24, 3.64 ± 1.21, and 3.55 ± 1.64 at 1, 24, 48, 72, and 120 h. The current research proved the value of 68Ga-NOTA-polypeptide-PEG11-Tz/anti-CD11b-TCO in the diagnosis of GC using the pretargeted strategy. Compared to 89Zr-DFO-anti-CD11b, the image contrast achieved by the pretargeted strategy was relatively improved, and the background accumulation of the probe was relatively low. These advantages can improve the diagnostic efficiency for GC and provide supporting evidence for radioimmunotherapy targeting CD11b receptors.


Assuntos
Antígeno CD11b/metabolismo , Química Click/métodos , Células Mieloides/metabolismo , Radioisótopos , Neoplasias Gástricas/metabolismo , Zircônio , Animais , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Mieloides/efeitos dos fármacos , Transplante de Neoplasias , Compostos Organometálicos , Tomografia por Emissão de Pósitrons/métodos , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/terapia , Microambiente Tumoral
8.
Anticancer Drugs ; 33(2): 117-123, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34561996

RESUMO

Platinum is reported to have adjuvant immune properties, whether oxaliplatin (OXA) could be utilized to synergize with anti-programmed cell death-1 (PD-1) antibody or anti-NKG2D (natural-killer group 2, member D) antibody is investigated. Subcutaneous A549 lung cancer and murine Lewis lung carcinoma (LLC) models were constructed, which were further intravenously injected with platinum-based drugs or concomitant administrated with anti-PD-1 antibody and or anti-NKG2D antibody. The tumor volume and the proportion of myeloid cells (CD45+CD11b+), CD3+T cells and NK (NK1.1+) cells were detected. The relative expression of chemokine (C-X-C motif) ligand 9 (CXCL9), CXCL10 and CXCL11 and C-X-C motif chemokine receptor 3 (CXCR3) was detected with the ELISA, western blot and flow cytometry. The three platinum drugs (cisplatin, DDP; carboplatin, CBP; OXA) showed similar effects to inhibit A549 tumor growth in immune-deficient mice. While OXA exhibited better antitumor efficacy in wild-type mice bearing LLC with downregulated myeloid cells proportion, upregulated concentration of CXCL9, CXCL10 and CXCL11, and upregulated proportion and CXCR3 expression on T cells and NK cells. OXA combined with anti-PD1 or anti-NKG2D synergistically improved tumor growth inhibition and survival. The combination of OXA to anti-PD1 and anti-NKG2D antibodies will provide the most appropriate treatment benefit. Oxaliplatin promotes T cells and NK cells infiltration through the CXCL9/10/11-CXCR3 axis to enhance anti-PD1 or anti-NKG2D immunotherapy in lung cancer.


Assuntos
Antineoplásicos/farmacologia , Quimiocinas CXC/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Células A549 , Animais , Antígenos de Superfície , Antineoplásicos/administração & dosagem , Carboplatina/administração & dosagem , Carboplatina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Combinação de Medicamentos , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Ligantes , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/efeitos dos fármacos , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina/farmacologia , Linfócitos T , Carga Tumoral/efeitos dos fármacos
9.
Bioengineered ; 12(2): 12332-12344, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34895041

RESUMO

Isoflurane-induced neurotoxicity has attracted much interest. Recent studies suggest that isoflurane causes microglial activation, resulting in an inflammatory response and microglial insult. Maprotiline is a novel drug that has been licensed as an antidepressant with considerable anti-inflammatory activity. However, it is still unknown whether maprotiline possesses a protective effect against isoflurane-induced microglial insult. Here, we found that maprotiline ameliorated isoflurane-caused reduction in BV2 microglial cell viability and lactate dehydrogenase (LDH) release. Maprotiline mitigated isoflurane-induced oxidative stress by inhibiting reactive oxygen species (ROS) production and increasing superoxide dismutase (SOD) activity. Isoflurane-induced expression and production of inflammatory markers including tumor necrosis factor (TNF-α), interleukin (IL)-1ß, cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2) were decreased in maprotiline-treated cells. Maprotiline inhibited the mRNA and protein levels of Iba1, a marker of microglial activation, in isoflurane-induced BV2 cells. Maprotiline treatment restored isoflurane-induced reduction of TREM2 in BV2 microglial cells. In addition, the knockdown of TREM2 abolished the beneficial effects of maprotiline against isoflurane. Collectively, maprotiline exerted protective effects against isoflurane-caused oxidative stress, inflammatory response, and cell injury via regulating TREM2. These findings show that maprotiline prevented the isoflurane-induced microglial activation, indicating that maprotiline might be used as an optimal therapeutic agent for preventing the isoflurane-caused neurotoxicity.


Assuntos
Isoflurano/farmacologia , Maprotilina/farmacologia , Glicoproteínas de Membrana/metabolismo , Microglia/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Receptores Imunológicos/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Inflamação/metabolismo , L-Lactato Desidrogenase/metabolismo , Camundongos , Microglia/metabolismo , Células Mieloides/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo
10.
Int J Mol Sci ; 22(22)2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34830245

RESUMO

Although histamine is a well-known itch mediator, histamine H1-receptor blockers often lack efficacy in chronic itch. Recent molecular and cellular based studies have shown that non-histaminergic mediators, such as proteases, neuropeptides and cytokines, along with their cognate receptors, are involved in evocation and modulation of itch sensation. Many of these molecules are produced and secreted by immune cells, which act on sensory nerve fibers distributed in the skin to cause itching and sensitization. This understanding of the connections between immune cell-derived mediators and sensory nerve fibers has led to the development of new treatments for itch. This review summarizes current knowledge of immune cell-derived itch mediators and neuronal response mechanisms, and discusses therapeutic agents that target these systems.


Assuntos
Anti-Inflamatórios/uso terapêutico , Histamina/imunologia , Fatores Imunológicos/uso terapêutico , Prurido/imunologia , Receptores Histamínicos H1/imunologia , Células Receptoras Sensoriais/imunologia , Anticorpos Monoclonais/uso terapêutico , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Citocinas/metabolismo , Expressão Gênica , Histamina/metabolismo , Antagonistas dos Receptores Histamínicos/uso terapêutico , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/patologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/patologia , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/imunologia , Neuropeptídeos/metabolismo , Peptídeo Hidrolases/imunologia , Peptídeo Hidrolases/metabolismo , Inibidores de Proteases/uso terapêutico , Prurido/tratamento farmacológico , Prurido/genética , Prurido/patologia , Receptores Histamínicos H1/genética , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/patologia , Pele/efeitos dos fármacos , Pele/imunologia , Pele/inervação , Pele/patologia
11.
Cells ; 10(11)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34831209

RESUMO

Nutritional intake impacts the human epigenome by directing epigenetic pathways in normal cell development via as yet unknown molecular mechanisms. Consequently, imbalance in the nutritional intake is able to dysregulate the epigenetic profile and drive cells towards malignant transformation. Here we present a novel epigenetic effect of the essential nutrient, NAD. We demonstrate that impairment of DNMT1 enzymatic activity by NAD-promoted ADP-ribosylation leads to demethylation and transcriptional activation of the CEBPA gene, suggesting the existence of an unknown NAD-controlled region within the locus. In addition to the molecular events, NAD- treated cells exhibit significant morphological and phenotypical changes that correspond to myeloid differentiation. Collectively, these results delineate a novel role for NAD in cell differentiation, and indicate novel nutri-epigenetic strategies to regulate and control gene expression in human cells.


Assuntos
Diferenciação Celular , Metilação de DNA/genética , NAD/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Desmetilação do DNA/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Neoplasias/genética , Neoplasias/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
13.
Nat Commun ; 12(1): 5981, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34645812

RESUMO

The acidic tumor microenvironment in melanoma drives immune evasion by up-regulating cyclic adenosine monophosphate (cAMP) in tumor-infiltrating monocytes. Here we show that the release of non-toxic concentrations of an adenylate cyclase (AC) inhibitor from poly(sarcosine)-block-poly(L-glutamic acid γ-benzyl ester) (polypept(o)id) copolymer micelles restores antitumor immunity. In combination with selective, non-therapeutic regulatory T cell depletion, AC inhibitor micelles achieve a complete remission of established B16-F10-OVA tumors. Single-cell sequencing of melanoma-infiltrating immune cells shows that AC inhibitor micelles reduce the number of anti-inflammatory myeloid cells and checkpoint receptor expression on T cells. AC inhibitor micelles thus represent an immunotherapeutic measure to counteract melanoma immune escape.


Assuntos
Inibidores de Adenilil Ciclases/farmacologia , Adenilil Ciclases/genética , Antineoplásicos/farmacologia , AMP Cíclico/antagonistas & inibidores , Melanoma Experimental/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Inibidores de Adenilil Ciclases/síntese química , Adenilil Ciclases/imunologia , Animais , Antineoplásicos/síntese química , Compostos de Benzil/química , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Ésteres , Feminino , Expressão Gênica , Humanos , Imunidade Inata/efeitos dos fármacos , Injeções Intralesionais , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Micelas , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/patologia , Peptídeos/química , Ácido Poliglutâmico/química , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Sarcosina/análogos & derivados , Sarcosina/química , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Carga Tumoral/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
14.
Int Immunopharmacol ; 101(Pt B): 108233, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34653730

RESUMO

The diversity of immune responses in allergic diseases is critically mediated by dendritic cells (DCs), including myeloid and plasmacytoid DCs. Allergen inhalation increased the release of IL-33 from patients with allergic rhinitis (AR), which affecting the downstream cells by binding to its receptor (ST2). However, the effects of inhaled allergens on the expression of ST2 by DCs and IL-33 on the function of mDCs are unknown. The levels of ST2+mDCs and ST2+pDCs in the blood from patients with AR and healthy subjects were examined using flow cytometry. Moreover, the patients were challenged using the allergens and the levels of ST2+mDCs and ST2+pDCs were investigated at different time points. We found that there were higher levels of ST2+ mDCs and ST2+ pDCs in patients with AR, and these levels were further increased 0.5 h after allergen inhalation. Additionally, the type 2 immune response was upregulated after challenge. IL-33 treatment increased the expression of ST2 on mDCs. Our study demonstrated that ST2 was upregulated on DCs after allergen inhalation and that mDCs responded directly to IL-33 through ST2, suggesting that the IL-33/ST2 axis might play an important role in the pathogenesis of allergic rhinitis by DCs.


Assuntos
Alérgenos/toxicidade , Células Dendríticas/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Células Mieloides/efeitos dos fármacos , Rinite Alérgica/metabolismo , Administração por Inalação , Adulto , Células Dendríticas/metabolismo , Feminino , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Interleucina-33 , Masculino , Células Mieloides/metabolismo
15.
Biomed Pharmacother ; 143: 112211, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34649344

RESUMO

Lipopolysaccharide (LPS) is the major component of the outer membrane of Gram-negative bacteria and is usually administrated to establish models of inflammation. Artesunate (ART), a water-soluble artemisinin derivative, displays multiple pharmacological actions against tumors, viral infections, and inflammation, and has been used as a therapeutic weapon against malaria. In this study, our aim was to evaluate whether ART pretreatment is capable of preventing inflammation induced by LPS. BALB/c mice were treated with 100 mg/kg of ART i.p. for 7 days followed by a single dose of LPS. ART pretreatment led to an improvement in clinical score, prevented alterations in biochemical markers, and reestablished the platelet counts. Flow cytometry analysis showed that ART protected the inflammation mainly by reducing the percentage of M1 macrophages while increasing M2 macrophages and a reestablishment of classical monocytes in the BM. In the spleen, ART pretreatment increased N2 neutrophils, myeloid-derived suppressor cells (MDSC), and regulatory T cells, the latter was also increased in peripheral blood. In addition, a marked decrease in inflammatory cytokines and chemokines was observed in the ART treated group. Our data suggest that ART prevents inflammation, reducing tissue damage and restoring homeostasis.


Assuntos
Anti-Inflamatórios/farmacologia , Artesunato/farmacologia , Inflamação/prevenção & controle , Células Mieloides/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fenótipo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
16.
Mol Syst Biol ; 17(10): e10387, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34664389

RESUMO

We need to effectively combine the knowledge from surging literature with complex datasets to propose mechanistic models of SARS-CoV-2 infection, improving data interpretation and predicting key targets of intervention. Here, we describe a large-scale community effort to build an open access, interoperable and computable repository of COVID-19 molecular mechanisms. The COVID-19 Disease Map (C19DMap) is a graphical, interactive representation of disease-relevant molecular mechanisms linking many knowledge sources. Notably, it is a computational resource for graph-based analyses and disease modelling. To this end, we established a framework of tools, platforms and guidelines necessary for a multifaceted community of biocurators, domain experts, bioinformaticians and computational biologists. The diagrams of the C19DMap, curated from the literature, are integrated with relevant interaction and text mining databases. We demonstrate the application of network analysis and modelling approaches by concrete examples to highlight new testable hypotheses. This framework helps to find signatures of SARS-CoV-2 predisposition, treatment response or prioritisation of drug candidates. Such an approach may help deal with new waves of COVID-19 or similar pandemics in the long-term perspective.


Assuntos
COVID-19/imunologia , Biologia Computacional/métodos , Bases de Dados Factuais , SARS-CoV-2/imunologia , Software , Antivirais/uso terapêutico , COVID-19/genética , COVID-19/virologia , Gráficos por Computador , Citocinas/genética , Citocinas/imunologia , Mineração de Dados/estatística & dados numéricos , Regulação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/virologia , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/imunologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/virologia , Mapeamento de Interação de Proteínas , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Tratamento Farmacológico da COVID-19
17.
Front Immunol ; 12: 738586, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691045

RESUMO

V-domain Ig suppressor of T cell activation (VISTA) is a novel coinhibitory immune checkpoint molecule that maintains immune homeostasis. The present study explored the role of VISTA in human and murine inflammatory tissues of apical periodontitis (AP). VISTA was upregulated in inflammatory tissues of human AP. In mice, the expression of VISTA gradually increased with the development of mouse experimental apical periodontitis (MAP), the CD3+ T cells, CD11b+ myeloid cells, and FOXP3+ regulatory T cells also gradually accumulated. Moreover, a blockade of VISTA using a mouse in vivo anti-VISTA antibody aggravated periapical bone loss and enhanced the infiltration of immune cells in an experimental mouse periapical periodontitis model. The collective results suggest that VISTA serves as a negative regulator of the development and bone loss of apical periodontitis.


Assuntos
Perda do Osso Alveolar/metabolismo , Processo Alveolar/efeitos dos fármacos , Anticorpos/toxicidade , Proteínas de Membrana/antagonistas & inibidores , Células Mieloides/efeitos dos fármacos , Periodontite Periapical/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Adulto , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/patologia , Processo Alveolar/imunologia , Processo Alveolar/metabolismo , Animais , Antígenos B7/metabolismo , Estudos de Casos e Controles , Modelos Animais de Doenças , Humanos , Masculino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Células Mieloides/imunologia , Células Mieloides/metabolismo , Periodontite Periapical/imunologia , Periodontite Periapical/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Adulto Jovem
18.
J Hematol Oncol ; 14(1): 142, 2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34496935

RESUMO

BACKGROUND: Tumor microenvironment (TME) is a dynamic cellular milieu to promote tumor angiogenesis, growth, proliferation, and metastasis, while derailing the host anti-tumor response. TME impedes bispecific antibody (BsAb) or chimeric antigen receptor (CAR)-driven T cells infiltration, survival, and cytotoxic efficacy. Modulating tumor infiltrating myeloid cells (TIMs) could potentially improve the efficacy of BsAb. METHODS: We evaluated the effects of TIM modulation on BsAb-driven T cell infiltration into tumors, their persistence, and in vivo anti-tumor response. Anti-GD2 BsAb and anti-HER2 BsAb built on IgG-[L]-scFv platform were tested against human cancer xenografts in BALB-Rag2-/-IL-2R-γc-KO (BRG) mice. Depleting antibodies specific for polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC), monocytic MDSC (M-MDSC), and tumor associated macrophage (TAM) were used to study the role of each TIM component. Dexamethasone, an established anti-inflammatory agent, was tested for its effect on TIMs. RESULTS: BsAb-driven T cells recruited myeloid cells into human tumor xenografts. Each TIM targeting therapy depleted cells of interest in blood and in tumors. Depletion of PMN-MDSCs, M-MDSCs, and particularly TAMs was associated with enhanced T cell infiltration into tumors, significantly improving tumor control and survival in multiple cancer xenograft models. Dexamethasone premedication depleted monocytes in circulation and TAMs in tumors, enhanced BsAb-driven T cell infiltration, and anti-tumor response with survival benefit. CONCLUSION: Reducing TIMs markedly enhanced anti-tumor effects of BsAb-based T cell immunotherapy by improving intratumoral T cell infiltration and persistence. TAM depletion was more effective than PMN- or M-MDSCs depletion at boosting the anti-tumor response of T cell engaging BsAb.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Células Mieloides/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos
19.
Oncogene ; 40(42): 6059-6070, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34556813

RESUMO

Glioblastoma (GBM) is the most common and malignant type of intracranial tumors with poor prognosis. Accumulating evidence suggests that phenotypic alterations of infiltrating myeloid cells in the tumor microenvironment are important for GBM progression. Conventional tumor immunotherapy commonly targets T-cells, while innate immunity as a therapeutic target is an emerging field. Targeting infiltrating myeloid cells that induce immune suppression in the TME provides a novel direction to improve the prognosis of patients with GBM. The factors released by tumor cells recruit myeloid cells into tumor bed and reprogram infiltrating myeloid cells into immunostimulatory/immunosuppressive phenotypes. Reciprocally, infiltrating myeloid cells, especially microglia/macrophages, regulate GBM progression and affect therapeutic efficacy. Herein, we revisited biological characteristics and functions of infiltrating myeloid cells and discussed the recent advances in immunotherapies targeting infiltrating myeloid cells in GBM. With an evolving understanding of the complex interactions between infiltrating myeloid cells and tumor cells in the tumor microenvironment, we will expand novel immunotherapeutic regimens targeting infiltrating myeloid cells in GBM treatment and improve the outcomes of GBM patients.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Células Mieloides/patologia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Progressão da Doença , Glioblastoma/tratamento farmacológico , Glioblastoma/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunidade Inata , Imunoterapia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Fenótipo , Prognóstico , Microambiente Tumoral
20.
Cell ; 184(19): 4981-4995.e14, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34464586

RESUMO

Poor tumor infiltration, development of exhaustion, and antigen insufficiency are common mechanisms that limit chimeric antigen receptor (CAR)-T cell efficacy. Delivery of pattern recognition receptor agonists is one strategy to improve immune function; however, targeting these agonists to immune cells is challenging, and off-target signaling in cancer cells can be detrimental. Here, we engineer CAR-T cells to deliver RN7SL1, an endogenous RNA that activates RIG-I/MDA5 signaling. RN7SL1 promotes expansion and effector-memory differentiation of CAR-T cells. Moreover, RN7SL1 is deployed in extracellular vesicles and selectively transferred to immune cells. Unlike other RNA agonists, transferred RN7SL1 restricts myeloid-derived suppressor cell (MDSC) development, decreases TGFB in myeloid cells, and fosters dendritic cell (DC) subsets with costimulatory features. Consequently, endogenous effector-memory and tumor-specific T cells also expand, allowing rejection of solid tumors with CAR antigen loss. Supported by improved endogenous immunity, CAR-T cells can now co-deploy peptide antigens with RN7SL1 to enhance efficacy, even when heterogenous CAR antigen tumors lack adequate neoantigens.


Assuntos
Fatores Imunológicos/farmacologia , RNA/farmacologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Antígenos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proteína DEAD-box 58/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Imunidade/efeitos dos fármacos , Imunocompetência , Memória Imunológica , Imunoterapia , Interferons/metabolismo , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Peptídeos/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...