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










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 43(3): 339-361, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38238476

RESUMO

Hematopoietic stem cell (HSC) divisional fate and function are determined by cellular metabolism, yet the contribution of specific cellular organelles and metabolic pathways to blood maintenance and stress-induced responses in the bone marrow remains poorly understood. The outer mitochondrial membrane-localized E3 ubiquitin ligase MITOL/MARCHF5 (encoded by the Mitol gene) is known to regulate mitochondrial and endoplasmic reticulum (ER) interaction and to promote cell survival. Here, we investigated the functional involvement of MITOL in HSC maintenance by generating MX1-cre inducible Mitol knockout mice. MITOL deletion in the bone marrow resulted in HSC exhaustion and impairment of bone marrow reconstitution capability in vivo. Interestingly, MITOL loss did not induce major mitochondrial dysfunction in hematopoietic stem and progenitor cells. In contrast, MITOL deletion induced prolonged ER stress in HSCs, which triggered cellular apoptosis regulated by IRE1α. In line, dampening of ER stress signaling by IRE1α inihibitor KIRA6 partially rescued apoptosis of long-term-reconstituting HSC. In summary, our observations indicate that MITOL is a principal regulator of hematopoietic homeostasis and protects blood stem cells from cell death through its function in ER stress signaling.


Assuntos
Endorribonucleases , Proteínas Serina-Treonina Quinases , Camundongos , Animais , Proteínas Serina-Treonina Quinases/genética , Apoptose , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Células-Tronco Hematopoéticas/metabolismo
2.
Nature ; 623(7986): 397-405, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37914940

RESUMO

Microglia are specialized brain-resident macrophages that arise from primitive macrophages colonizing the embryonic brain1. Microglia contribute to multiple aspects of brain development, but their precise roles in the early human brain remain poorly understood owing to limited access to relevant tissues2-6. The generation of brain organoids from human induced pluripotent stem cells recapitulates some key features of human embryonic brain development7-10. However, current approaches do not incorporate microglia or address their role in organoid maturation11-21. Here we generated microglia-sufficient brain organoids by coculturing brain organoids with primitive-like macrophages generated from the same human induced pluripotent stem cells (iMac)22. In organoid cocultures, iMac differentiated into cells with microglia-like phenotypes and functions (iMicro) and modulated neuronal progenitor cell (NPC) differentiation, limiting NPC proliferation and promoting axonogenesis. Mechanistically, iMicro contained high levels of PLIN2+ lipid droplets that exported cholesterol and its esters, which were taken up by NPCs in the organoids. We also detected PLIN2+ lipid droplet-loaded microglia in mouse and human embryonic brains. Overall, our approach substantially advances current human brain organoid approaches by incorporating microglial cells, as illustrated by the discovery of a key pathway of lipid-mediated crosstalk between microglia and NPCs that leads to improved neurogenesis.


Assuntos
Encéfalo , Colesterol , Células-Tronco Pluripotentes Induzidas , Microglia , Células-Tronco Neurais , Neurogênese , Organoides , Animais , Humanos , Camundongos , Encéfalo/citologia , Encéfalo/metabolismo , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Microglia/citologia , Microglia/metabolismo , Organoides/citologia , Organoides/metabolismo , Colesterol/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Axônios , Proliferação de Células , Ésteres/metabolismo , Gotículas Lipídicas/metabolismo
3.
Cell Rep ; 42(6): 112599, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37279110

RESUMO

Therapeutic neoantigen cancer vaccines have limited clinical efficacy to date. Here, we identify a heterologous prime-boost vaccination strategy using a self-assembling peptide nanoparticle TLR-7/8 agonist (SNP) vaccine prime and a chimp adenovirus (ChAdOx1) vaccine boost that elicits potent CD8 T cells and tumor regression. ChAdOx1 administered intravenously (i.v.) had 4-fold higher antigen-specific CD8 T cell responses than mice boosted by the intramuscular (i.m.) route. In the therapeutic MC38 tumor model, i.v. heterologous prime-boost vaccination enhances regression compared with ChAdOx1 alone. Remarkably, i.v. boosting with a ChAdOx1 vector encoding an irrelevant antigen also mediates tumor regression, which is dependent on type I IFN signaling. Single-cell RNA sequencing of the tumor myeloid compartment shows that i.v. ChAdOx1 reduces the frequency of immunosuppressive Chil3 monocytes and activates cross-presenting type 1 conventional dendritic cells (cDC1s). The dual effect of i.v. ChAdOx1 vaccination enhancing CD8 T cells and modulating the TME represents a translatable paradigm for enhancing anti-tumor immunity in humans.


Assuntos
Linfócitos T CD8-Positivos , Vacinação , Humanos , Camundongos , Animais , Imunidade Adaptativa , Vetores Genéticos , Adjuvantes Imunológicos
4.
Sci Immunol ; 7(78): eadd3330, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36525505

RESUMO

Langerhans cell histiocytosis (LCH) is a potentially fatal neoplasm characterized by the aberrant differentiation of mononuclear phagocytes, driven by mitogen-activated protein kinase (MAPK) pathway activation. LCH cells may trigger destructive pathology yet remain in a precarious state finely balanced between apoptosis and survival, supported by a unique inflammatory milieu. The interactions that maintain this state are not well known and may offer targets for intervention. Here, we used single-cell RNA-seq and protein analysis to dissect LCH lesions, assessing LCH cell heterogeneity and comparing LCH cells with normal mononuclear phagocytes within lesions. We found LCH discriminatory signatures pointing to senescence and escape from tumor immune surveillance. We also uncovered two major lineages of LCH with DC2- and DC3/monocyte-like phenotypes and validated them in multiple pathological tissue sites by high-content imaging. Receptor-ligand analyses and lineage tracing in vitro revealed Notch-dependent cooperativity between DC2 and DC3/monocyte lineages during expression of the pathognomonic LCH program. Our results present a convergent dual origin model of LCH with MAPK pathway activation occurring before fate commitment to DC2 and DC3/monocyte lineages and Notch-dependent cooperativity between lineages driving the development of LCH cells.


Assuntos
Histiocitose de Células de Langerhans , Neoplasias , Humanos , Linhagem da Célula , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/patologia , Diferenciação Celular , Monócitos/metabolismo
5.
Cell ; 185(23): 4317-4332.e15, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-36302380

RESUMO

Therapeutic cancer vaccines are designed to increase tumor-specific T cell immunity. However, suppressive mechanisms within the tumor microenvironment (TME) may limit T cell function. Here, we assessed how the route of vaccination alters intratumoral myeloid cells. Using a self-assembling nanoparticle vaccine that links tumor antigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we treated tumor-bearing mice subcutaneously (SNP-SC) or intravenously (SNP-IV). Both routes generated antigen-specific CD8+ T cells that infiltrated tumors. However, only SNP-IV mediated tumor regression, dependent on systemic type I interferon at the time of boost. Single-cell RNA-sequencing revealed that intratumoral monocytes expressing an immunoregulatory gene signature (Chil3, Anxa2, Wfdc17) were reduced after SNP-IV boost. In humans, the Chil3+ monocyte gene signature is enriched in CD16- monocytes and associated with worse outcomes. Our results show that the generation of tumor-specific CD8+ T cells combined with remodeling of the TME is a promising approach for tumor immunotherapy.


Assuntos
Vacinas Anticâncer , Microambiente Tumoral , Humanos , Camundongos , Animais , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Imunoterapia/métodos , Antígenos de Neoplasias , Vacinação/métodos , Adjuvantes Imunológicos
6.
Immunity ; 54(9): 2101-2116.e6, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34469775

RESUMO

Tissue macrophages are immune cells whose phenotypes and functions are dictated by origin and niches. However, tissues are complex environments, and macrophage heterogeneity within the same organ has been overlooked so far. Here, we used high-dimensional approaches to characterize macrophage populations in the murine liver. We identified two distinct populations among embryonically derived Kupffer cells (KCs) sharing a core signature while differentially expressing numerous genes and proteins: a major CD206loESAM- population (KC1) and a minor CD206hiESAM+ population (KC2). KC2 expressed genes involved in metabolic processes, including fatty acid metabolism both in steady-state and in diet-induced obesity and hepatic steatosis. Functional characterization by depletion of KC2 or targeted silencing of the fatty acid transporter Cd36 highlighted a crucial contribution of KC2 in the liver oxidative stress associated with obesity. In summary, our study reveals that KCs are more heterogeneous than anticipated, notably describing a subpopulation wired with metabolic functions.


Assuntos
Antígenos CD36/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Estresse Oxidativo/fisiologia , Animais , Camundongos
7.
J Exp Med ; 218(9)2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34279540

RESUMO

Inflammatory skin diseases including atopic dermatitis (AD) and psoriasis (PSO) are underpinned by dendritic cell (DC)-mediated T cell responses. Currently, the heterogeneous human cutaneous DC population is incompletely characterized, and its contribution to these diseases remains unclear. Here, we performed index-sorted single-cell flow cytometry and RNA sequencing of lesional and nonlesional AD and PSO skin to identify macrophages and all DC subsets, including the newly described mature LAMP3+BIRC3+ DCs enriched in immunoregulatory molecules (mregDC) and CD14+ DC3. By integrating our indexed data with published skin datasets, we generated a myeloid cell universe of DC and macrophage subsets in healthy and diseased skin. Importantly, we found that CD14+ DC3s increased in PSO lesional skin and co-produced IL1B and IL23A, which are pathological in PSO. Our study comprehensively describes the molecular characteristics of macrophages and DC subsets in AD and PSO at single-cell resolution, and identifies CD14+ DC3s as potential promoters of inflammation in PSO.


Assuntos
Dermatite Atópica/patologia , Interleucina-1beta/metabolismo , Subunidade p19 da Interleucina-23/metabolismo , Células de Langerhans/patologia , Psoríase/patologia , Dermatite Atópica/metabolismo , Expressão Gênica , Redes Reguladoras de Genes , Humanos , Interleucina-15/metabolismo , Células de Langerhans/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/citologia , Psoríase/metabolismo , Análise de Célula Única
8.
Nat Immunol ; 22(1): 41-52, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33139915

RESUMO

Personalized cancer vaccines are a promising approach for inducing T cell immunity to tumor neoantigens. Using a self-assembling nanoparticle vaccine that links neoantigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we show how the route and dose alter the magnitude and quality of neoantigen-specific CD8+ T cells. Intravenous vaccination (SNP-IV) induced a higher proportion of TCF1+PD-1+CD8+ T cells as compared to subcutaneous immunization (SNP-SC). Single-cell RNA sequencing showed that SNP-IV induced stem-like genes (Tcf7, Slamf6, Xcl1) whereas SNP-SC enriched for effector genes (Gzmb, Klrg1, Cx3cr1). Stem-like cells generated by SNP-IV proliferated and differentiated into effector cells upon checkpoint blockade, leading to superior antitumor response as compared to SNP-SC in a therapeutic model. The duration of antigen presentation by dendritic cells controlled the magnitude and quality of CD8+ T cells. These data demonstrate how to optimize antitumor immunity by modulating vaccine parameters for specific generation of effector or stem-like CD8+ T cells.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Fator 1-alfa Nuclear de Hepatócito/análise , Nanopartículas , Animais , Apresentação de Antígeno , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Feminino , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Vacinação
9.
Cell ; 183(2): 377-394.e21, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32976798

RESUMO

We employed scRNA sequencing to extensively characterize the cellular landscape of human liver from development to disease. Analysis of ∼212,000 cells representing human fetal, hepatocellular carcinoma (HCC), and mouse liver revealed remarkable fetal-like reprogramming of the tumor microenvironment. Specifically, the HCC ecosystem displayed features reminiscent of fetal development, including re-emergence of fetal-associated endothelial cells (PLVAP/VEGFR2) and fetal-like (FOLR2) tumor-associated macrophages. In a cross-species comparative analysis, we discovered remarkable similarity between mouse embryonic, fetal-liver, and tumor macrophages. Spatial transcriptomics further revealed a shared onco-fetal ecosystem between fetal liver and HCC. Furthermore, gene regulatory analysis, spatial transcriptomics, and in vitro functional assays implicated VEGF and NOTCH signaling in maintaining onco-fetal ecosystem. Taken together, we report a shared immunosuppressive onco-fetal ecosystem in fetal liver and HCC. Our results unravel a previously unexplored onco-fetal reprogramming of the tumor ecosystem, provide novel targets for therapeutic interventions in HCC, and open avenues for identifying similar paradigms in other cancers and disease.


Assuntos
Carcinoma Hepatocelular/patologia , Células Endoteliais/metabolismo , Microambiente Tumoral/genética , Adulto , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Receptor 2 de Folato/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Fígado/patologia , Neoplasias Hepáticas/genética , Macrófagos/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Transcriptoma/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Immunity ; 51(3): 573-589.e8, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31474513

RESUMO

Human mononuclear phagocytes comprise phenotypically and functionally overlapping subsets of dendritic cells (DCs) and monocytes, but the extent of their heterogeneity and distinct markers for subset identification remains elusive. By integrating high-dimensional single-cell protein and RNA expression data, we identified distinct markers to delineate monocytes from conventional DC2 (cDC2s). Using CD88 and CD89 for monocytes and HLA-DQ and FcεRIα for cDC2s allowed for their specific identification in blood and tissues. We also showed that cDC2s could be subdivided into phenotypically and functionally distinct subsets based on CD5, CD163, and CD14 expression, including a distinct subset of circulating inflammatory CD5-CD163+CD14+ cells related to previously defined DC3s. These inflammatory DC3s were expanded in systemic lupus erythematosus patients and correlated with disease activity. These findings further unravel the heterogeneity of DC subpopulations in health and disease and may pave the way for the identification of specific DC subset-targeting therapies.


Assuntos
Biomarcadores/sangue , Células Dendríticas/imunologia , Inflamação/sangue , Inflamação/imunologia , Leucócitos Mononucleares/imunologia , Fagócitos/imunologia , Antígenos CD/sangue , Antígenos CD/imunologia , Células Cultivadas , Citometria de Fluxo/métodos , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Monócitos/imunologia , Fenótipo , Análise de Célula Única
11.
J Cell Physiol ; 233(11): 8908-8918, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29797576

RESUMO

Umbelliprenin (UMB) has shown various pharmacological properties in vitro. We investigated the antineoplastic and immunostimulatory effects of UMB in 4T1 mammary-tumor-bearing mice. Two-hundred microliter of UMB (12.5 mg/ml) was intraperitoneally administrated to healthy and tumor-bearing female Balb/c mice for a period of 18 days. Data was analyzed using GraphPad Prism 5 software for Windows (version 5, La Jolla, CA). UMB caused a significant decrease in tumor size (P < 0.01). Serum interferon gamma (IFNγ) was augmented in both healthy and tumor-bearing animals (P < 0.01), and IL-4 declined in healthy animals (P < 0.01) treated with UMB. Expressions of Ki-67, VEGF, CD31, MMP2, MMP9, VCAM1, and NF-κB were significantly decreased in tumors from UMB-treated animals (P < 0.001), whereas E-Cadherin and TNFR1 expressions were markedly increased (P < 0.001). The rates of liver and lung metastases in UMB-administrated animals were smaller compared to the control. UMB can potently inhibit tumor growth, angiogenesis, metastasis, and inflammation and potentiate an antitumor immune response in vivo. However, further investigations are required to evaluate the UMB mechanisms of action in cancerous cells.


Assuntos
Inflamação/tratamento farmacológico , Neoplasias Mamárias Animais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Umbeliferonas/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Inflamação/sangue , Inflamação/genética , Inflamação/patologia , Interferon gama/sangue , Neoplasias Mamárias Animais/sangue , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neovascularização Patológica/sangue , Neovascularização Patológica/genética , Neovascularização Patológica/patologia
12.
Eur J Nutr ; 57(3): 1025-1044, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28229276

RESUMO

PURPOSE: Chlorella vulgaris (CV) has exhibited immune-enhancing and protective activities against cancer and infections. However, there is an increasing concern about the use of Chlorella species in human, regarding its various molecules with antigenic features found in infectious microorganisms. Our goal was to investigate the impact of higher concentrations of CV on tumor growth in spontaneous mouse mammary tumor (SMMT) models. METHODS: Balb/c mice were daily given CV powder at doses of 0, 200, or 300 mg/kg for 42 days (CONTROL, CV200, and CV300 groups, respectively; n = 6/group). On day 14, the SMMT was inoculated. Tumor volume (TV) and body weight (BW) were monitored on 5-day intervals following tumor challenge. On day 43, blood, spleen, lungs, and tumor tissues were collected. Histopathological examinations on lungs and tumor tissues were performed following hematoxylin-eosin staining. Intratumor expression of 27 genes was assessed by real-time PCR. Total IgG, IFNγ, and IL-4 levels in serum and spleen culture supernatant were measured by ELISA. RESULTS: The TV/BW index showed significant increase in the CV200 group compared to the CONTROL (p = 0.047). The CV200 tumors exhibited more malignant phenotype, higher angiogenesis rate, and lower peritumoral neutrophil and macrophage-to-lymphocyte infiltration ratio compared to the CONTROL. Serum concentrations of IFNγ, IL-4, and IgG were declined, and the spleen IFNγ and IgG production was higher in the CV200 compared to the CONTROL. The IL-1ß, IL-10, TGFß1, FOXP3, HO-1, Gr1, CD11b, PCNA, LCN2, iNOS2, VEGFR2, CD31, and CD105L expressions were markedly increased in the CV200 tumors compared to the CONTROL (p = 0.001, 0.002, 0.006, 0.021, 0.004, 0.030, 0.016, 0.031, 0.025, 0.008, 0.014, 0.022, and 0.037, respectively). The changes in cytokine, IgG and gene expression values considerably correlated with tumor size, as well as with each other. CONCLUSIONS: Our data provided evidence that C. vulgaris at a specific dose (200 mg/kg) promoted tumor growth in a mammary tumor model. This consequence might reflect an immune derangement in favor of developing a protumor microenvironment. However, this hypothesis needs to be further investigated in future.


Assuntos
Carcinoma Ductal de Mama/imunologia , Chlorella vulgaris/imunologia , Imunossupressores/efeitos adversos , Interferon gama/sangue , Neoplasias Mamárias Experimentais/imunologia , Probióticos/efeitos adversos , Baço/imunologia , Animais , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/prevenção & controle , Carcinoma Ductal de Mama/secundário , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica , Imunoglobulina G/análise , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Interferon gama/antagonistas & inibidores , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Probióticos/administração & dosagem , Probióticos/uso terapêutico , Baço/metabolismo , Baço/patologia , Carga Tumoral , Microambiente Tumoral
13.
Gene ; 634: 29-36, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28844667

RESUMO

BACKGROUND: Oxidative stress is closely linked to inflammation in neurodegenerative diseases. We aimed to investigate the expression of redox system genes in Multiple Sclerosis (MS) patients either exposed or not exposed to conventional treatments. METHODS: Forty-four MS patients were divided into three groups: newly diagnosed (Group 1), receiving interferon (Group 2) and receiving immunosuppressive drugs (Group 3). Also, 15 healthy controls were enrolled. The mRNA expression of TRX1, TXNRD1, TRX2, TXNRD2, TXNIP, and APEX1 genes in peripheral blood mononuclear cells (PBMCs) was assessed by relative quantitative real-time PCR. Also, serum level of Trx1 was measured by ELISA. RESULTS: Serum level of Trx1 in the newly diagnosed MS patients was significantly higher compared to the healthy controls (P=0.013). Likewise, TRX1 and APEX1 expressions were significantly higher in the newly diagnosed patients compared to controls (P=0.003 and P=0.042), patients under interferon treatment (P=0.003 and P=0.013), and patients received immunosuppressants (P=0.001 and P=0.025). Furthermore, TXNIP expression in MS patients (either group 1, group 2, or group 3) was significantly lower than that in the control group (P=0.017, P=0.002, and P=0.022 respectively). The expression of TXNRD1, TRX2, and TXNRD2 did not show any significant difference between the control and the MS patient (P>0.05). CONCLUSIONS: Our data showed that redox system elements are differentially expressed in newly diagnosed MS patients, or patients receiving either interferon or immunosuppressive treatments. However, much more studies are required to confirm our findings and clarify the underlying mechanisms.


Assuntos
Proteínas de Transporte/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Esclerose Múltipla/genética , Tiorredoxinas/genética , Adolescente , Adulto , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Interferons/farmacologia , Interferons/uso terapêutico , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Tiorredoxinas/sangue , Adulto Jovem
14.
J Immunoassay Immunochem ; 38(3): 333-342, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27929756

RESUMO

BACKGROUND: We hypothesized that Tumor cell lysate (TCL) prepared from spontaneous mouse mammary tumor (SMMT) may elicit IgG production by spleen mononuclear cells (SMCs) in ex vivo. METHODS: The SMCs from healthy mice (HM, n = 6) and four-week SMMT-bearing mice (TBM, n = 6) was cultured in presence of TCL and mitogen for 42 hr at 37°C, separately. Serum and SMCs culture supernatant levels of IFNγ, IL-4, and total IgG were measured using special ELISA kits. RESULTS: Serum IgG level of TBM was significantly higher than that of HM group (P = 0.019), while serum IFNγ and IL-4 did not differ between two groups (P > 0.05). Mitogen significantly induced ex vivo production of both IFNγ (P = 0.013) and IL-4 (P = 0.015) by SMCs from HM group, and only IL-4 (P = 0.049) by SMCs from TBM group. In contrast, TCL increased ex vivo production of IgG by SMCs from both HM (P = 0.034) and TBM (P = 0.016) groups. The ex vivo IgG revealed a moderate positive correlation with tumor size (r = 0.578, P = 0.422). CONCLUSION: It seems that TCL prepared from SMMT are potent inducers of IgG production. This may propose TCL as a potential tool for monitoring of humoral immunity in animal models.


Assuntos
Imunoglobulina G/biossíntese , Neoplasias Mamárias Animais/imunologia , Baço/citologia , Baço/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/sangue , Interleucina-4/sangue , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas
15.
Oncol Lett ; 12(5): 3845-3855, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27895739

RESUMO

Gene expression profiling has been suggested to predict breast cancer outcome. The prognostic value of the 8q22-24 position in breast cancer remains to be elucidated. The present study evaluated expression patterns of the genes located at this position in metastatic and non-metastatic breast cancer. A total of 85 patients with recurrent/metastatic (n=15) and non-metastatic (n=70) early-stage, estrogen receptor-positive and lymph node-negative breast tumors were included. In addition, 15 normal breast tissue samples were used as controls. Demographic and clinical features were recorded. Subsequently, mRNA copy numbers of exostosin glycosyltransferase 1 (EXT1), WNT1 inducible signaling pathway protein 1 (WISP1), ATPase family, AAA domain containing 2 (ATAD2), TSP-like 5 (TSPYL5), metadherin (MTDH) and cyclin E2 (CCNE2) genes were measured by reverse transcription-quantitative polymerase chain reaction assay. The expression of EXT1 and WISP1 exhibited a significant decline in the metastatic breast cancer group compared to the control (P=0.015 and P=0.012, respectively). The expression of TSPYL5, MTDH and ATAD2 was significantly decreased in the metastatic (P=0.002, P=0.018 and P=0.016, respectively) and non-metastatic (P=0.038, P=0.045 and P=0.000, respectively) breast cancer groups compared with the control. The expression of CCNE2 in the metastatic and non-metastatic breast cancer groups was significantly increased compared with the control (P=0.002 and P=0.001, respectively). WISP1 expression demonstrated a correlation with patient age and tumor size, and TSPYL5 expression was correlated with lymphovascular invasion. None of the genes investigated exhibited any correlation with stage and grade of disease. The TSPYL5, MTDH, ATAD2 and CCNE2 genes may be implicated in the pathogenesis of human breast cancer, while the WISP1 and EXT1 genes may have the potential to serve as promising indicators of the risk of metastasis. However, further studies are required to validate these results.

16.
Iran Red Crescent Med J ; 18(7): e35167, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27703798

RESUMO

BACKGROUND: Resistance to chemotherapy is a growing concern, thus natural anticancer agents are drawing the attention of many scientists and clinicians. One natural anticancer agent, umbelliprenin, is a coumarin produced by many species of Ferula. OBJECTIVES: We aimed to examine the inhibitory effect of umbelliprenin on human and mouse bone marrow-derived stem cells (BMDSCs), peripheral blood mononuclear cells (PBMCs), and different cancer cell lines. MATERIALS AND METHODS: In this in vitro experimental study, the HT29, CT26, MCF-7, 4T1, A172, and GL26 cancer cells and human and mouse BMDSCs and PBMCs were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS), incubated at 37°C for 24 hours in a 5% CO2 atmosphere, and then were treated with different concentrations of umbelliprenin dissolved in dimethyl sulfoxide (DMSO) (3, 6, 12, 25, 50, 100, and 200 µg/mL) for 24, 48, and 72 hours at 37°C. Each experiment was performed in triplicate. Finally, the cell survival rate was assessed by MTT assay. The IC50 values were calculated based on the log values using GraphPad Prism version 5 software for windows (La Jolla CA, USA) and were expressed as mean ± SEM. RESULTS: Umbelliprenin inhibited the cancer cells in a concentration-dependent (P < 0.05) but not time-dependent manner (P > 0.05). The most sensitive and resistant cell lines at the 24-hour incubation time were 4T1 (IC50, 30.9 ± 3.1 µg/mL) and A172 (IC50, 51.9 ± 6.7 µg/mL); at the 48-hour incubation time: 4T1 (IC50, 30.6 ± 2.6 µg/mL) and CT26 (IC50, 53.2 ± 3.6 µg/mL); and at the 72-hour incubation time: HT29 (IC50, 37.1 ± 1.4 µg/mL) and 4T1 (IC50, 62.2 ± 4.8 µg/mL). Both human and mouse BMDSCs showed the highest resistance at the 24-hour incubation time (IC50s, 254.7 ± 21 and 204.4 ± 4.5 µg/mL, respectively) and the highest sensitivity at the 72-hour incubation time (IC50s, 120.4 ± 5 and 159.0 ± 7.3 µg/mL, respectively). The PBMCs of both human and mouse origin revealed very strong resistance to the studied concentrations of umbelliprenin (IC50s ranging from 713.5 ± 499.1 to 6651 ± 3670.7 µg/mL). CONCLUSIONS: Our findings indicate that umbelliprenin exhibits concentration-dependent inhibitory effects on various cell types; it is potentially toxic against the HT29, CT26, MCF-7, 4T1, A172, and GL26 cell lines, potentially harmful against BMDSCs, and non-toxic against PBMCs. Therefore, if our results are approved in the future, umbelliprenin can be an appropriate candidate for developing treatments against different cancers.

17.
Korean J Gastroenterol ; 65(4): 215-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25896155

RESUMO

BACKGROUND/AIMS: Several clinical trials have revealed various advantages for probiotics in inflammatory bowel disease (IBD). The aim of this study was to further investigate the effects of probiotic yogurt consumption on gut microbiota in patients with this disease. METHODS: A total of 305 participants were divided into three groups; group A (IBD patients receiving probiotic yogurt; n=105), group B (IBD patients receiving placebo; n=105), and control group (healthy individuals receiving probiotic yogurt; n=95). Stool samples were collected both before and after 8 weeks of intervention; and population of Lactobacillus, Bifidobacterium and Bacteroides in the stool specimens was measured by Taqman real-time PCR method. RESULTS: By the end of the intervention, no significant variations in the mean weight and body mass index were observed between three groups (p>0.05). However, the mean numbers of Lactobacillus, Bifidobacterium, and Bacteroides in group A were significantly increased compared to group B (p<0.001, p<0.001, and p< 0.01, respectively). There were also significant differences in the mean numbers of either of three bacteria between group A and the healthy control group; however, these differences between two groups were observed both at baseline and the end of the intervention. CONCLUSIONS: Consumption of probiotic yogurt by patients with IBD may help to improve intestinal function by increasing the number of probiotic bacteria in the intestine and colon. However, many more studies are required in order to prove the concept.


Assuntos
Doenças Inflamatórias Intestinais/tratamento farmacológico , Probióticos/uso terapêutico , Adulto , Bacteroides/genética , Bifidobacterium/genética , DNA Bacteriano/análise , Método Duplo-Cego , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal , Humanos , Intestinos/microbiologia , Lactobacillus/genética , Masculino , Pessoa de Meia-Idade , Efeito Placebo , Reação em Cadeia da Polimerase em Tempo Real
18.
Asian Pac J Cancer Prev ; 15(19): 8295-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25339020

RESUMO

BACKGROUND: H pylori is the main causative agent of Gastric cancer and chronic gastritis. Genetic diversity of H. pylori has major contribution in its pathogenesis. We investigated the prevalence of oipA and iceA1/iceA2 positive strains of H. pylori among patients with gastric cancer and gastritis. MATERIALS AND METHODS: Sampling performed by means of endoscopy from 86 patients. DNA was extracted from tissue samples using DNA extraction kit. PCR assay was performed and products were monitored by Agarose Gel Electrophoresis. RESULTS: Urease Test and 16S rRNA PCR did not show significant differences in detection of H. pylori. The frequency of iceA1 allele in patients with gastric cancer was significantly higher than those with gastritis (p<0.05). However, there was no significant difference in prevalence of oipA and iceA2 genes among the two groups of patients (p>0.05). CONCLUSIONS: The iceA1 gene, but the oipA and iceA2 genes , is associated with H. pylori-induced gastric cancer. However, confirmatory studies must be performed in future.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Biomarcadores Tumorais/genética , Infecções por Helicobacter/genética , Neoplasias Gástricas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Gastrite/genética , Gastrite/patologia , Genótipo , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Reação em Cadeia da Polimerase , Prognóstico , RNA Ribossômico 16S/genética , Neoplasias Gástricas/patologia , Urease/metabolismo
19.
Tumour Biol ; 35(5): 4757-64, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24425107

RESUMO

Transforming growth factor ß1 (TGFß1) is suggested to be involved in the pathogenesis of and in complications with breast cancer (BC). Polymorphisms in TGFß1 gene (TGFß1) have been suggested by many investigators to have a role in susceptibility to BC; however, many discordant data have been reported. Considering the role of ethnic variations, we performed an association study between TGFß1 polymorphisms and BC among Iranian women. We sequenced DNA samples of 110 BC and 110 normal control women for the exons and their adjacent intronic regions of TGFß1 using PCR. The allele, genotype, and haplotype frequencies were calculated using PowerMarker V3.25 and R 3.0.2 softwares. Ten single nucleotide polymorphisms (SNPs) were detected. Statistical analysis on the frequency of seven most frequent SNPs, including the three coding SNPs (cSNPs) revealed no significant difference between BC and control women. Moreover, among 11 constructed haplotypes, "GTGCCGC" was significantly different between two study groups. In conclusion, we found no association between the studied SNPs of TGFß1 and BC among Iranian women, but a possible association between "GTGCCGC" haplotype and BC was seen. However, further studies are suggested to clarify this association.


Assuntos
Neoplasias da Mama/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Fator de Crescimento Transformador beta1/genética , Adolescente , Adulto , Neoplasias da Mama/etiologia , Feminino , Haplótipos , Humanos , Íntrons , Pessoa de Meia-Idade , Risco
20.
Iran J Neurol ; 12(4): 136-43, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24250923

RESUMO

BACKGROUND: Recent studies have demonstrated controversial results and somewhat increased frequency of multiple sclerosis (MS). We reevaluated the files of MS patients from Qom Province of Iran in order to investigate the epidemiology of the disease. METHODS: Demographic and clinical records of 592 MS patients were reviewed, which included; age, sex, date of birth, marital and occupation status, presenting symptoms, time of onset, type and family history of MS, and history of autoimmune or other diseases. RESULTS: At the time of our study, 11 patients had died, and 581 were alive with a total female-to-male ratio of 3.4. The mean age of onset of the disease was 34.25 ± 9.01 for all the patients. 11.2% of patients had positive family history of MS. The majority of patients (80.1%) showed relapsing-remitting (RR) pattern. The prevalence of MS was calculated as 50.4/100000 for Qom. CONCLUSION: Qom is located within a high risk zone of MS. Although we found evidences about the role of environmental factors, geographical distribution, and etcetera, many more studies need to be performed in this respect.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...