Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Immunohorizons ; 6(12): 790-806, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36480485

RESUMO

Owing to ease of access and high yield, most murine myeloid-derived suppressor cell (MDSC) knowledge comes from the study of spleen-derived MDSCs rather than those isolated from the tumor. Although several studies have identified subtle differences in suppressive function between these MDSCs, a recent report demonstrated that the whole peripheral myeloid compartment poorly reflects myeloid populations found at the tumor. We confirm and extend these observations by presenting data that indicate extensive differences exist between peripheral and tumor MDSCs, suggesting that it may be inappropriate to use spleen MDSCs as surrogates for studying tumor MDSCs. Using cytospins, we observed that tumor MDSCs have undergone a morphologic shift from immature myeloid cell forms commonly seen in bone marrow (BM) and spleen MDSCs and acquired mature myeloid cell characteristics. Spleen and BM monocyte-like MDSCs (M-MDSCs) readily responded to differentiation signals for multiple myeloid cell types whereas tumor M-MDSCs had remarkably reduced cellular plasticity. At the time of isolation, M-MDSCs from BM or spleen have little to no T cell suppressive activity whereas those from the tumor possess immediate and efficient T cell suppressive function. Finally, microarray analysis revealed that the transcriptomes of tumor and spleen M-MDSCs possessed >4500 differentially expressed transcripts. We conclude that tumor M-MDSCs are more differentiated and mature, and that they are morphologically, genetically, and functionally distinct from spleen and BM M-MDSCs. These observations have important implications for the design of anti-MDSC therapies and suggest that preclinical studies using nontumor MDSCs could lead to results not applicable to tumor MDSCs.


Assuntos
Células Supressoras Mieloides , Neoplasias , Animais , Camundongos , Monócitos , Diferenciação Celular
2.
Adv Exp Med Biol ; 1390: 155-167, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36107318

RESUMO

Vitamin D is a conditionally required nutrient that can either be obtained from skin synthesis following UVB exposure from the diet. Once in the body, it is metabolized to produce the endocrine hormone, 1,25 dihydroxyvitamin D (1,25(OH)2D), that regulates gene expression in target tissues by interacting with a ligand-activated transcription factor, the vitamin D receptor (VDR). The first, and most responsive, vitamin D target tissue is the intestine. The classical intestinal role for vitamin D is the control of calcium metabolism through the regulation of intestinal calcium absorption. However, studies clearly show that other functions of the intestine are regulated by the molecular actions of 1,25(OH)2 D that are mediated through the VDR. This includes enhancing gut barrier function, regulation of intestinal stem cells, suppression of colon carcinogenesis, and inhibiting intestinal inflammation. While research demonstrates that there are both classical, calcium-regulating and non-calcium regulating roles for vitamin D in the intestine, the challenge facing biomedical researchers is how to translate these findings in ways that optimize human intestinal health.


Assuntos
Intestinos , Receptores de Calcitriol , Vitamina D , Cálcio/metabolismo , Hormônios , Humanos , Ligantes , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Fatores de Transcrição , Vitamina D/metabolismo , Vitaminas
3.
J Biol Chem ; 298(8): 102213, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35779631

RESUMO

1,25-dihydroxyvitamin D (VD) regulates intestinal calcium absorption in the small intestine (SI) and also reduces risk of colonic inflammation and cancer. However, the intestine compartment-specific target genes of VD signaling are unknown. Here, we examined VD action across three functional compartments of the intestine using RNA-seq to measure VD-induced changes in gene expression and Chromatin Immunoprecipitation with next generation sequencing to measure vitamin D receptor (VDR) genomic binding. We found that VD regulated the expression of 55 shared transcripts in the SI crypt, SI villi, and in the colon, including Cyp24a1, S100g, Trpv6, and Slc30a10. Other VD-regulated transcripts were unique to the SI crypt (162 up, 210 down), villi (199 up, 63 down), or colon (102 up, 28 down), but this did not correlate with mRNA levels of the VDR. Furthermore, bioinformatic analysis identified unique VD-regulated biological functions in each compartment. VDR-binding sites were found in 70% of upregulated genes from the colon and SI villi but were less common in upregulated genes from the SI crypt and among downregulated genes, suggesting some transcript-level VD effects are likely indirect. Consistent with this, we show that VD regulated the expression of other transcription factors and their downstream targets. Finally, we demonstrate that compartment-specific VD-mediated gene expression was associated with compartment-specific VDR-binding sites (<30% of targets) and enrichment of intestinal transcription factor-binding motifs within VDR-binding peaks. Taken together, our data reveal unique spatial patterns of VD action in the intestine and suggest novel mechanisms that could account for compartment-specific functions of this hormone.


Assuntos
Receptores de Calcitriol , Vitamina D , Animais , Genômica , Intestinos , Camundongos , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacologia , Vitamina D3 24-Hidroxilase/genética
4.
Physiol Genomics ; 53(11): 486-508, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34612061

RESUMO

Human intestinal epithelial organoids (enteroids and colonoids) are tissue cultures used for understanding the physiology of the human intestinal epithelium. Here, we explored the effect on the transcriptome of common variations in culture methods, including extracellular matrix substrate, format, tissue segment, differentiation status, and patient heterogeneity. RNA-sequencing datasets from 276 experiments performed on 37 human enteroid and colonoid lines from 29 patients were aggregated from several groups in the Texas Medical Center. DESeq2 and gene set enrichment analysis (GSEA) were used to identify differentially expressed genes and enriched pathways. PERMANOVA, Pearson's correlation, and dendrogram analysis of the data originally indicated three tiers of influence of culture methods on transcriptomic variation: substrate (collagen vs. Matrigel) and format (3-D, transwell, and monolayer) had the largest effect; segment of origin (duodenum, jejunum, ileum, colon) and differentiation status had a moderate effect; and patient heterogeneity and specific experimental manipulations (e.g., pathogen infection) had the smallest effect. GSEA identified hundreds of pathways that varied between culture methods, such as IL1 cytokine signaling enriched in transwell versus monolayer cultures and E2F target genes enriched in collagen versus Matrigel cultures. The transcriptional influence of the format was furthermore validated in a synchronized experiment performed with various format-substrate combinations. Surprisingly, large differences in organoid transcriptome were driven by variations in culture methods such as format, whereas experimental manipulations such as infection had modest effects. These results show that common variations in culture conditions can have large effects on intestinal organoids and should be accounted for when designing experiments and comparing results between laboratories. Our data constitute the largest RNA-seq dataset interrogating human intestinal epithelial organoids.


Assuntos
Técnicas de Cultura de Células/métodos , Colo/metabolismo , Meios de Cultura/farmacologia , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Organoides/metabolismo , Transcriptoma/efeitos dos fármacos , Calcitriol/farmacologia , Colágeno/metabolismo , Colágeno/farmacologia , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Meios de Cultura/química , Combinação de Medicamentos , Escherichia coli , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Laminina/metabolismo , Laminina/farmacologia , Organoides/virologia , Proteoglicanas/metabolismo , Proteoglicanas/farmacologia , RNA-Seq/métodos , Transcriptoma/genética , Viroses/metabolismo , Viroses/virologia , Vírus
5.
Mol Cell Biol ; 41(1)2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33139494

RESUMO

Although vitamin D is critical for the function of the intestine, most studies have focused on the duodenum. We show that transgenic expression of the vitamin D receptor (VDR) only in the distal intestine of VDR null mice (KO/TG mice) results in the normalization of serum calcium and rescue of rickets. Although it had been suggested that calcium transport in the distal intestine involves a paracellular process, we found that the 1,25-dihydroxyvitamin D3 [1,25(OH)2D3]-activated genes in the proximal intestine associated with active calcium transport (Trpv6, S100g, and Atp2b1) are also induced by 1,25(OH)2D3 in the distal intestine of KO/TG mice. In addition, Slc30a10, encoding a manganese efflux transporter, was one of the genes most induced by 1,25(OH)2D3 in both proximal and distal intestine. Both villus and crypt were found to express Vdr and VDR target genes. RNA sequence (RNA-seq) analysis of human enteroids indicated that the effects of 1,25(OH)2D3 observed in mice are conserved in humans. Using Slc30a10-/- mice, a loss of cortical bone and a marked decrease in S100g and Trpv6 in the intestine was observed. Our findings suggest an interrelationship between vitamin D and intestinal Mn efflux and indicate the importance of distal intestinal segments to vitamin D action.


Assuntos
Calcitriol/genética , Mucosa Intestinal/metabolismo , Intestinos/fisiologia , Animais , Calcitriol/metabolismo , Cálcio/metabolismo , Genômica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Vitamina D/farmacologia
7.
Am J Respir Cell Mol Biol ; 63(1): 79-91, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32135073

RESUMO

Vitamin D deficiency (VDD) during pregnancy is associated with increased respiratory morbidities and risk for chronic lung disease after preterm birth. However, the direct effects of maternal VDD on perinatal lung structure and function and whether maternal VDD increases the susceptibility of lung injury due to hyperoxia are uncertain. In the present study, we sought to determine whether maternal VDD is sufficient to impair lung structure and function and whether VDD increases the impact of hyperoxia on the developing rat lung. Four-week-old rats were fed VDD chow and housed in a room shielded from ultraviolet A/B light to achieve 25-hydroxyvitamin D concentrations <10 ng/ml at mating and throughout lactation. Lung structure was assessed at 2 weeks for radial alveolar count, mean linear intercept, pulmonary vessel density, and lung function (lung compliance and resistance). The effects of hyperoxia for 2 weeks after birth were assessed after exposure to fraction of inspired oxygen of 0.95. At 2 weeks, VDD offspring had decreased alveolar and vascular growth and abnormal airway reactivity and lung function. Impaired lung structure and function in VDD offspring were similar to those observed in control rats exposed to postnatal hyperoxia alone. Maternal VDD causes sustained abnormalities of distal lung growth, increases in airway hyperreactivity, and abnormal lung mechanics during infancy. These changes in VDD pups were as severe as those measured after exposure to postnatal hyperoxia alone. We speculate that antenatal disruption of vitamin D signaling increases the risk for late-childhood respiratory disease.


Assuntos
Hiperóxia/complicações , Complacência Pulmonar/fisiologia , Lesão Pulmonar/etiologia , Pulmão/fisiopatologia , Deficiência de Vitamina D/complicações , Vitamina D/análogos & derivados , Animais , Animais Recém-Nascidos , Feminino , Hiperóxia/metabolismo , Pulmão/metabolismo , Lesão Pulmonar/metabolismo , Gravidez , Ratos , Vitamina D/metabolismo
8.
Cancer Prev Res (Phila) ; 12(6): 343-356, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31028080

RESUMO

We tested whether lifelong modification of vitamin D signaling can alter the progression of early prostate carcinogenesis in studies using mice that develop high-grade prostatic intraepithelial neoplasia that is similar to humans. Two tissue-limited models showed that prostate vitamin D receptor (VDR) loss increased prostate carcinogenesis. In another study, we fed diets with three vitamin D3 levels (inadequate = 25 IU/kg diet, adequate for bone health = 150 IU/kg, or high = 1,000 IU/kg) and two calcium levels (adequate for bone health = 0.5% and high = 1.5%). Dietary vitamin D caused a dose-dependent increase in serum 25-hydroxyvitamin D levels and a reduction in the percentage of mice with adenocarcinoma but did not improve bone mass. In contrast, high calcium suppressed serum 1,25-dihydroxyvitamin D levels and improved bone mass but increased the incidence of adenocarcinoma. Analysis of the VDR cistrome in RWPE1 prostate epithelial cells revealed vitamin D-mediated regulation of multiple cancer-relevant pathways. Our data support the hypothesis that the loss of vitamin D signaling accelerates the early stages of prostate carcinogenesis, and our results suggest that different dietary requirements may be needed to support prostate health or maximize bone mass. SIGNIFICANCE: This work shows that disrupting vitamin D signaling through diet or genetic deletion increases early prostate carcinogenesis through multiple pathways. Higher-diet vitamin D levels are needed for cancer than bone.


Assuntos
Adenocarcinoma/prevenção & controle , Carcinogênese/metabolismo , Neoplasias da Próstata/prevenção & controle , Receptores de Calcitriol/metabolismo , Vitamina D/metabolismo , Vitaminas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Apoptose , Carcinogênese/patologia , Proliferação de Células , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Células Tumorais Cultivadas
11.
Endocrinology ; 158(6): 1951-1963, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28368514

RESUMO

Low vitamin D status potentiates experimental colitis, but the vitamin D-responsive cell in colitis has not been defined. We hypothesized that vitamin D has distinct roles in colonic epithelial cells and in nonepithelial cells during colitis. We tested this hypothesis by using mice with vitamin D receptor (VDR) deletion from colon epithelial cells (CEC-VDRKO) or nonintestinal epithelial cells (NEC-VDRKO). Eight-week-old mice were treated with 1.35% dextran sulfate sodium (DSS) for 5 days and then euthanized 2 or 10 days after removal of DSS. DSS induced body weight loss and increased disease activity index and spleen size. This response was increased in NEC-VDRKO mice but not CEC-VDRKO mice. DSS-induced colon epithelial damage and immune cell infiltration scores were increased in both mouse models. Although the epithelium healed between 2 and 10 days after DSS administration in control and CEC-VDRKO mice, epithelial damage remained high in NEC-VDRKO mice 10 days after removal of DSS, indicating delayed epithelial healing. Gene expression levels for the proinflammatory, M1 macrophage (Mɸ) cytokines tumor necrosis factor-α, nitric oxide synthase 2, and interleukin-1ß were significantly elevated in the colon of NEC-VDRKO mice at day 10. In vitro experiments in murine peritoneal Mɸs demonstrated that 1,25 dihydroxyvitamin D directly inhibited M1 polarization, facilitated M2 polarization, and regulated Mɸ phenotype switching toward the M2 and away from the M1 phenotype. Our data revealed unique protective roles for vitamin D signaling during colitis in the colon epithelium as well as nonepithelial cells in the colon microenvironment (i.e., modulation of Mɸ biology).


Assuntos
Colite/genética , Citoproteção/genética , Receptores de Calcitriol/fisiologia , Animais , Colite/induzido quimicamente , Colite/prevenção & controle , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Calcitriol/genética , Transdução de Sinais/genética
12.
J Steroid Biochem Mol Biol ; 173: 194-201, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28104492

RESUMO

High vitamin D status is associated with reduced colon cancer risk but these studies ignore the diversity in the molecular etiology of colon cancer. RAS activating mutations are common in colon cancer and they activate pro-proliferative signaling pathways. We examined the impact of RAS activating mutations on 1,25 dihydroxyvitamin D (1,25(OH)2D)-mediated gene expression in cultured colon and intestinal cell lines. Transient transfection of Caco-2 cells with a constitutively active mutant K-RAS (G12 V) significantly reduced 1,25(OH)2D-induced activity of both a human 25-hydroxyvitamin D, 24 hydroxyase (CYP24A1) promoter-luciferase and an artificial 3X vitamin D response element (VDRE) promoter-luciferase reporter gene. Young Adult Mouse Colon (YAMC) and Rat Intestinal Epithelial (RIE) cell lines with stable expression of mutant H-RAS had suppressed 1,25(OH)2D-mediated induction of CYP24A1 mRNA. The RAS effects were associated with lower Vitamin D receptor (VDR) mRNA and protein levels in YAMC and RIE cells and they could be partially reversed by VDR overexpression. RAS-mediated suppression of VDR levels was not due to either reduced VDR mRNA stability or increased VDR gene methylation. However, chromatin accessibility to the VDR gene at the proximal promoter (-300bp), an enhancer region at -6kb, and an enhancer region located in exon 3 was significantly reduced in RAS transformed YAMC cells (YAMC-RAS). These data show that constitutively active RAS signaling suppresses 1,25(OH)2D-mediated gene transcription in colon epithelial cells by reducing VDR gene transcription but the mechanism for this suppression is not yet known. These data suggest that cancers with RAS-activating mutations may be less responsive to vitamin D mediated treatment or chemoprevention.


Assuntos
Neoplasias do Colo/genética , Mucosa Intestinal/citologia , Receptores de Calcitriol/genética , Ativação Transcricional , Vitamina D/análogos & derivados , Proteínas ras/metabolismo , Animais , Células CACO-2 , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Mutação , Ratos , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Vitamina D/genética , Vitamina D/metabolismo , Vitamina D3 24-Hidroxilase/genética , Vitamina D3 24-Hidroxilase/metabolismo , Proteínas ras/genética
13.
Dig Dis Sci ; 61(4): 1069-79, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26631394

RESUMO

BACKGROUND: Animal models are an important tool to understand intestinal biology. Our laboratory previously generated C57BL/6-Tg(Car1-cre)5Flt transgenic mice (CAC) with large-intestine-specific Cre recombinase (Cre) expression as a model to study colon health. AIM: To expand the utility of the CAC mouse model by determining the impact of chemically induced colitis on CAC transgene expression. METHODS: CAC mice were crossed to Rosa reporter mice (Rosa26R (flox/flox) ) with a lox-STOP-lox signal controlling ß-galactosidase (ßgal) expression and then further crossed with Apc(CKO/CKO) mice in some experiments to delete Apc alleles (Apc (Δ580) ). Initially, 8-week-old CAC(Tg/WT);Rosa26R (flox/WT) ;Apc (Δ580/WT) mice were treated with dextran sulfate sodium (DSS) in drinking water (5 days, 0, 0.65, 1.35, or 2.0 %). Colon tissue damage and ßgal labeling were analyzed 10 day after stopping DSS. Next, 8-week-old CAC(Tg/WT);Rosa26R(flox/flox) mice were treated with 0 or 1.35 % DSS, and colonic ßgal labeling was assessed at 30 day post-DSS treatment. Finally, 10-week-old CAC(Tg/WT);Apc (Δ580/WT) mice were treated with DSS (0 or 2 %) for 5 days and colonic tumors were analyzed at 20 weeks. RESULTS: CAC(Tg/WT);Rosa26R (flox/WT) ;Apc (Δ580/WT) mice had a DSS dose-dependent increase in colon epithelial damage that correlated with increased epithelial ßgal labeling at 10 days (r (2) = 0.9, ß = 0.75). The ßgal labeling in CAC(Tg/WT);Rosa26R(flox/flox) mice colon remained high at 30 days, especially in the crypts of the healed ulcer. DSS also increased colon tumor incidence and multiplicity in CAC(Tg/WT);Apc (Δ580/WT) mice. CONCLUSIONS: DSS-mediated epithelial damage induces a persistent, Cre-mediated recombination of floxed alleles in CAC mice. This enables the examination of gene function in colon epithelium during experimental colitis and colitis-induced colon cancer.


Assuntos
Colite/genética , Animais , Colite/metabolismo , Colite/patologia , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Integrases , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Recombinação Genética , Transgenes , beta-Galactosidase/metabolismo
14.
J Steroid Biochem Mol Biol ; 148: 103-10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25597951

RESUMO

In colon cancer, adenomatous polyposis coli (APC) inactivating gene mutations increase nuclear ß-catenin levels and stimulate proliferation. In vitro, 1,25 dihydroxyvitamin D (1,25(OH)2D), suppresses ß-catenin-mediated gene transcription by inducing vitamin D receptor (VDR)-ß-catenin interactions. We examined whether acute treatment with 1,25(OH)2D could suppress ß-catenin-mediated gene transcription in the hyperplastic colonic lesions of mice with colon-specific deletion of both APC gene alleles (CAC; APC(Δ580/Δ580)). At four weeks of age, CAC; APC(Δ580/Δ580) and control mice were injected with vehicle or 1,25(OH)2D (1µg/kg body weight) once a day for three days and then killed six hours after the last injection. mRNA levels of ß-catenin target genes were elevated in the colon of CAC; APC(Δ580/Δ580) mice. 1,25(OH)2D increased 25 hydroxyvitamin D-24 hydroxylase mRNA levels in the colon of CAC; APC(Δ580/Δ580) and control mice indicating the treatments activated the VDR. However, 1,25(OH)2D had no effect on either ß-catenin target gene mRNA levels or the proliferation index in CAC; APC(Δ580/Δ580) or control mice. VDR mRNA and protein levels were lower (-65% and -90%) in the colon of CAC; APC(Δ580/Δ580) mice compared to control mice, suggesting loss of colon responsiveness to vitamin D. Consistent with this, vitamin D-induced expression of transient receptor potential cation channel, subfamily V, member 6 mRNA was reduced in the colon of CAC; APC(Δ580/Δ580) mice. Our data show that short term exposure to 1,25(OH)2D does not suppress colonic ß-catenin signaling in vivo. This article is part of a special issue entitled '17th Vitamin D Workshop'.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Calcitriol/farmacologia , Colo/metabolismo , Doenças do Colo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , RNA Mensageiro/genética , Receptores de Calcitriol/genética , Vitamina D3 24-Hidroxilase/genética , beta Catenina/genética , Animais , Colo/efeitos dos fármacos , Colo/patologia , Doenças do Colo/tratamento farmacológico , Doenças do Colo/patologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Vitaminas/farmacologia
15.
Immunity ; 41(2): 296-310, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25065623

RESUMO

Intestinal microbial metabolites are conjectured to affect mucosal integrity through an incompletely characterized mechanism. Here we showed that microbial-specific indoles regulated intestinal barrier function through the xenobiotic sensor, pregnane X receptor (PXR). Indole 3-propionic acid (IPA), in the context of indole, is a ligand for PXR in vivo, and IPA downregulated enterocyte TNF-α while it upregulated junctional protein-coding mRNAs. PXR-deficient (Nr1i2(-/-)) mice showed a distinctly "leaky" gut physiology coupled with upregulation of the Toll-like receptor (TLR) signaling pathway. These defects in the epithelial barrier were corrected in Nr1i2(-/-)Tlr4(-/-) mice. Our results demonstrate that a direct chemical communication between the intestinal symbionts and PXR regulates mucosal integrity through a pathway that involves luminal sensing and signaling by TLR4.


Assuntos
Intestinos/imunologia , Receptores de Esteroides/imunologia , Junções Íntimas/imunologia , Receptor 4 Toll-Like/imunologia , Junções Aderentes/genética , Junções Aderentes/imunologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anticorpos/imunologia , Complexo CD3/imunologia , Células CACO-2 , Linhagem Celular , Feminino , Células HEK293 , Humanos , Indóis , Indometacina/farmacologia , Inflamação/imunologia , Intestinos/microbiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/imunologia , Receptor de Pregnano X , Interferência de RNA , RNA Mensageiro , RNA Interferente Pequeno , Receptores de Esteroides/genética , Traumatismo por Reperfusão/imunologia , Transdução de Sinais/imunologia , Junções Íntimas/genética , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/biossíntese
16.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G249-59, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24875098

RESUMO

Colorectal cancer is a heterogeneous disease that is one of the major causes of cancer death in the U.S. There is evidence that lifestyle factors like diet can modulate the course of this disease. Demonstrating the benefit and mechanism of action of dietary interventions against colon cancer will require studies in preclinical models. Many mouse models have been developed to study colon cancer but no single model can reflect all types of colon cancer in terms of molecular etiology. In addition, many models develop only low-grade cancers and are confounded by development of the disease outside of the colon. This review will discuss how mice can be used to model human colon cancer and it will describe a variety of new mouse models that develop colon-restricted cancer as well as more advanced phenotypes for studies of late-state disease.


Assuntos
Quimioprevenção/métodos , Neoplasias Colorretais/prevenção & controle , Dieta , Animais , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dieta/efeitos adversos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Fenótipo , Comportamento de Redução do Risco , Especificidade da Espécie
17.
Cancer Lett ; 347(2): 191-5, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24632531

RESUMO

Several genetically engineered mouse (GEM) models of colorectal cancer have been developed and are a mainstay in our efforts to identify means of preventing and treating this disease. Many of these models involve a germline disruption of the adenomatous polyposis coli (Apc) tumor suppressor gene and share the limitation that the great preponderance of tumors appear in the small rather than large intestine. In recent years efforts have been made to increase the similarity of these models to human sporadic colorectal cancer by disrupting Apc in a tissue-specific fashion using the Cre-Lox system so that the genetic aberrations are confined to the colonic epithelium. These models have shown great promise but reproducible and high penetrance colon-specific tumorigenesis has not yet been achieved without invasive techniques to introduce the Cre enzyme. We therefore sought to create a new model with high penetrance colon-specific tumorigenesis but without the need for exogenous Cre administration. We utilized existing mice possessing a conditional knock out for the Apc gene and a latent activated Kras allele and crossed them with mice expressing Cre recombinase solely in the large intestine. Using this approach we generated mice that developed 1-9 colonic adenomas per mouse (average 4.3) but without any tumors in the small intestine or cecum. No invasive tumors were observed. Despite the apparent lack of invasion, the geographical correctness, complete penetrance and intermediate tumor burden make this model a promising addition to our toolkit for the study of colorectal cancer treatment and prevention.


Assuntos
Neoplasias do Colo/patologia , Genes APC , Genes ras , Integrases/fisiologia , Mutação , Animais , Sequência de Bases , Neoplasias do Colo/genética , Primers do DNA , Camundongos
18.
Cancer Metastasis Rev ; 32(1-2): 39-61, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23076650

RESUMO

Colorectal cancer is a heterogeneous disease that afflicts a large number of people in the USA. The use of animal models has the potential to increase our understanding of carcinogenesis, tumor biology, and the impact of specific molecular events on colon biology. In addition, animal models with features of specific human colorectal cancers can be used to test strategies for cancer prevention and treatment. In this review, we provide an overview of the mechanisms driving human cancer, we discuss the approaches one can take to model colon cancer in animals, and we describe a number of specific animal models that have been developed for the study of colon cancer. We believe that there are many valuable animal models to study various aspects of human colorectal cancer. However, opportunities for improving upon these models exist.


Assuntos
Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Animais , Modelos Animais de Doenças , Humanos , Metástase Neoplásica , Fenótipo
19.
Am J Physiol Regul Integr Comp Physiol ; 302(11): R1282-96, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22461179

RESUMO

The aim of this study was to identify genes that influence iron regulation under varying dietary iron availability. Male and female mice from 20+ BXD recombinant inbred strains were fed iron-poor or iron-adequate diets from weaning until 4 mo of age. At death, the spleen, liver, and blood were harvested for the measurement of hemoglobin, hematocrit, total iron binding capacity, transferrin saturation, and liver, spleen and plasma iron concentration. For each measure and diet, we found large, strain-related variability. A principal-components analysis (PCA) was performed on the strain means for the seven parameters under each dietary condition for each sex, followed by quantitative trait loci (QTL) analysis on the factors. Compared with the iron-adequate diet, iron deficiency altered the factor structure of the principal components. QTL analysis, combined with PosMed (a candidate gene searching system) published gene expression data and literature citations, identified seven candidate genes, Ptprd, Mdm1, Picalm, lip1, Tcerg1, Skp2, and Frzb based on PCA factor, diet, and sex. Expression of each of these is cis-regulated, significantly correlated with the corresponding PCA factor, and previously reported to regulate iron, directly or indirectly. We propose that polymorphisms in multiple genes underlie individual differences in iron regulation, especially in response to dietary iron challenge. This research shows that iron management is a highly complex trait, influenced by multiple genes. Systems genetics analysis of iron homeostasis holds promise for developing new methods for prevention and treatment of iron deficiency anemia and related diseases.


Assuntos
Anemia Ferropriva/genética , Deficiências de Ferro , Análise de Variância , Anemia Ferropriva/sangue , Anemia Ferropriva/metabolismo , Animais , Peso Corporal , Feminino , Expressão Gênica , Hemoglobinas/análise , Homeostase/genética , Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Análise Multivariada , Polimorfismo Genético , Locos de Características Quantitativas
20.
Biochem J ; 441(1): 61-76, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22168439

RESUMO

The population-based association between low vitamin D status and increased cancer risk can be inconsistent, but it is now generally accepted. These relationships link low serum 25OHD (25-hydroxyvitamin D) levels to cancer, whereas cell-based studies show that the metabolite 1,25(OH)2D (1,25-dihydroxyvitamin D) is a biologically active metabolite that works through vitamin D receptor to regulate gene transcription. In the present review we discuss the literature relevant to the molecular events that may account for the beneficial impact of vitamin D on cancer prevention or treatment. These data show that although vitamin D-induced growth arrest and apoptosis of tumour cells or their non-neoplastic progenitors are plausible mechanisms, other chemoprotective mechanisms are also worthy of consideration. These alternative mechanisms include enhancing DNA repair, antioxidant protection and immunomodulation. In addition, other cell targets, such as the stromal cells, endothelial cells and cells of the immune system, may be regulated by 1,25(OH)2D and contribute to vitamin D-mediated cancer prevention.


Assuntos
Neoplasias/etiologia , Deficiência de Vitamina D/complicações , Vitamina D/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Neoplasias/prevenção & controle , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Vitamina D/uso terapêutico , Vitaminas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA