Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Commun Med (Lond) ; 4(1): 128, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956268

RESUMEN

BACKGROUND: Human carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) is an inhibitory cell surface protein that functions through homophilic and heterophilic ligand binding. Its expression on immune cells in human tumors is poorly understood. METHODS: An antibody that distinguishes human CEACAM1 from other highly related CEACAM family members was labeled with 159Tb and inserted into a panel of antibodies that included specificity for programmed cell death protein 1 (PD1) and PD-L1, which are targets of immunotherapy, to gain a data-driven immune cell atlas using cytometry by time-of-flight (CyTOF). A detailed inventory of CEACAM1, PD1, and PD-L1 expression on immune cells in metastatic lesions to lymph node or soft tissues and peripheral blood samples from patients with treatment-naive and -resistant melanoma as well as peripheral blood samples from healthy controls was performed. RESULTS: CEACAM1 is absent or at low levels on healthy circulating immune cells but is increased on immune cells in peripheral blood and tumors of melanoma patients. The majority of circulating PD1-positive NK cells, innate T cells, B cells, monocytic cells, dendritic cells, and CD4+ T cells in the peripheral circulation of treatment-resistant disease co-express CEACAM1 and are demonstrable as discrete populations. CEACAM1 is present on distinct types of cells that are unique to the tumor microenvironment and exhibit expression levels that are highest in treatment resistance; this includes tumor-infiltrating CD8+ T cells. CONCLUSIONS: To the best of our knowledge, this work represents the first comprehensive atlas of CEACAM1 expression on immune cells in a human tumor and reveals an important correlation with treatment-resistant disease. These studies suggest that agents targeting CEACAM1 may represent appropriate partners for PD1-related pathway therapies.


Some proteins, such as programmed cell death protein 1 (PD1), can stop the immune system from attacking cancer cells, allowing cancers to grow. Therapies targeting these proteins can be highly effective, but tumors can become resistant. It is important to identify factors involved in this resistance to develop improved cancer therapies. Human carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) is a protein that inhibits an immune response and its levels have been associated with poor patient outcomes. We applied a method that allows for the detection of proteins on a single cell to uncover CEACAM1 patterns in melanoma. We found that increased CEACAM1 expression levels on multiple different immune cell types was associated with tumors that were resistant to therapy. These findings may help us to understand the role of CEACAM1 in cancer and to develop better cancer therapies.

2.
Mol Cell Biol ; 44(7): 261-272, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38828991

RESUMEN

The protein tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) plays an important role in modulating glucose and lipid homeostasis. We previously suggested a potential role of SHP-1 in the regulation of peroxisome proliferator-activated receptor γ2 (PPARγ2) expression and activity but the mechanisms were unexplored. PPARγ2 is the master regulator of adipogenesis, but how its activity is regulated by tyrosine phosphorylation is largely unknown. Here, we found that SHP-1 binds to PPARγ2 primarily via its N-terminal SH2-domain. We confirmed the phosphorylation of PPARγ2 on tyrosine-residue 78 (Y78), which was reduced by SHP-1 in vitro resulting in decreased PPARγ2 stability. Loss of SHP-1 led to elevated, agonist-induced expression of the classical PPARγ2 targets FABP4 and CD36, concomitant with increased lipid content in cells expressing PPARγ2, an effect blunted by abrogation of PPARγ2 phosphorylation. Collectively, we discovered that SHP-1 affects the stability of PPARγ2 through dephosphorylation thereby influencing adipogenesis.


Asunto(s)
Adipogénesis , PPAR gamma , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , PPAR gamma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Fosforilación , Humanos , Animales , Ratones , Antígenos CD36/metabolismo , Antígenos CD36/genética , Células HEK293 , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Estabilidad Proteica , Células 3T3-L1 , Dominios Homologos src , Unión Proteica
3.
Oncogene ; 41(11): 1563-1575, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35091681

RESUMEN

We investigated the role of the NFE2L3 transcription factor in inflammation-induced colorectal cancer. Our studies revealed that Nfe2l3-/- mice exhibit significantly less inflammation in the colon, reduced tumor size and numbers, and skewed localization of tumors with a more pronounced decrease of tumors in the distal colon. CIBERSORT analysis of RNA-seq data from normal and tumor tissue predicted a reduction in mast cells in Nfe2l3-/- animals, which was confirmed by toluidine blue staining. Concomitantly, the transcript levels of Il33 and Rab27a, both important regulators of mast cells, were reduced and increased, respectively, in the colorectal tumors of Nfe2l3-/- mice. Furthermore, we validated NFE2L3 binding to the regulatory sequences of the IL33 and RAB27A loci in human colorectal carcinoma cells. Using digital spatial profiling, we found that Nfe2l3-/- mice presented elevated FOXP3 and immune checkpoint markers CTLA4, TIM3, and LAG3, suggesting an increase in Treg counts. Staining for CD3 and FOXP3 confirmed a significant increase in immunosuppressive Tregs in the colon of Nfe2l3-/- animals. Also, Human Microbiome Project (HMP2) data showed that NFE2L3 transcript levels are higher in the rectum of ulcerative colitis patients. The observed changes in the tumor microenvironment provide new insights into the molecular differences regarding colon cancer sidedness. This may be exploited for the treatment of early-onset colorectal cancer as this emerging subtype primarily displays distal/left-sided tumors.


Asunto(s)
Neoplasias Colorrectales , Microambiente Tumoral , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Neoplasias Colorrectales/genética , Factores de Transcripción Forkhead , Humanos , Inflamación/genética , Interleucina-33 , Ratones , Linfocitos T Reguladores , Microambiente Tumoral/genética
4.
J Immunol ; 204(8): 2285-2294, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32169849

RESUMEN

Neutrophils promote tumor growth and metastasis at multiple stages of cancer progression. One mechanism through which this occurs is via release of neutrophil extracellular traps (NETs). We have previously shown that NETs trap tumor cells in both the liver and the lung, increasing their adhesion and metastasis following postoperative complications. Multiple studies have since shown that NETs play a role in tumor progression and metastasis. NETs are composed of nuclear DNA-derived web-like structures decorated with neutrophil-derived proteins. However, it is unknown which, if any, of these NET-affiliated proteins is responsible for inducing the metastatic phenotype. In this study, we identify the NET-associated carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) as an essential element for this interaction. Indeed, blocking CEACAM1 on NETs, or knocking it out in a murine model, leads to a significant decrease in colon carcinoma cell adhesion, migration and metastasis. Thus, this work identifies NET-associated CEACAM1 as a putative therapeutic target to prevent the metastatic progression of colon carcinoma.


Asunto(s)
Antígenos CD/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Neutrófilos/inmunología , Células A549 , Animales , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Células HT29 , Humanos , Ratones , Neutrófilos/patología
5.
Sci Rep ; 9(1): 18897, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31827213

RESUMEN

The mechanisms linking chronic inflammation of the gut (IBD) and increased colorectal cancer susceptibility are poorly understood. IBD risk is influenced by genetic factors, including the IBD5 locus (human 5q31), that harbors the IRF1 gene. A cause-to-effect relationship between chronic inflammation and colorectal cancer, and a possible role of IRF1 were studied in Irf1-/- mice in a model of colitis-associated colorectal cancer (CA-CRC) induced by azoxymethane and dextran sulfate. Loss of Irf1 causes hyper-susceptibility to CA-CRC, with early onset and increased number of tumors leading to rapid lethality. Transcript profiling (RNA-seq) and immunostaining of colons shows heightened inflammation and enhanced enterocyte proliferation in Irf1-/- mutants, prior to appearance of tumors. Considerable infiltration of leukocytes is seen in Irf1-/- colons at this early stage, and is composed primarily of proinflammatory Gr1+ Cd11b+ myeloid cells and other granulocytes, as well as CD4+ lymphoid cells. Differential susceptibility to CA-CRC of Irf1-/- vs. B6 controls is fully transferable through hematopoietic cells as observed in bone marrow chimera studies. Transcript signatures seen in Irf1-/- mice in response to AOM/DSS are enriched in clinical specimens from patients with IBD and with colorectal cancer. In addition, IRF1 expression in the colon is significantly decreased in late stage colorectal cancer (stages 3, 4) and is associated with poorer prognosis. This suggests that partial or complete loss of IRF1 expression alters the type, number, and function of immune cells in situ during chronic inflammation, possibly via the creation of a tumor-promoting environment.


Asunto(s)
Colitis/metabolismo , Neoplasias Colorrectales/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Animales , Colitis/complicaciones , Colitis/genética , Colitis/patología , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Factor 1 Regulador del Interferón/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados
7.
Oncotarget ; 8(61): 104330-104346, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29262644

RESUMEN

We have shown that carcinoembryonic antigen cell adhesion molecule 1 long isoform (CEACAM1-L) expression in MC38 metastatic colorectal cancer (CRC) cells results in liver metastasis inhibition via CCL2 and STAT3 signaling. But other molecular mechanisms orchestrating CEACAM1-L-mediated metastasis inhibition remain to be defined. We screened a panel of mouse and human CRC cells and evaluated their metastatic outcome after CEACAM1 overexpression or downregulation. An unbiased transcript profiling and a phospho-receptor tyrosine kinase screen comparing MC38 CEACAM1-L-expressing and non-expressing (CT) CRC cells revealed reduced ephrin type-A receptor 2 (EPHA2) expression and activity. An EPHA2-specific inhibitor reduced EPHA2 downstream signaling in CT cells similar to that in CEACAM1-L cells with decreased proliferation and migration. Human CRC patients exhibiting high CEACAM1 in combination with low EPHA2 expression benefited from longer time to first recurrence/metastasis compared to those with high EPHA2 expression. With the added interaction of CEACAM6, we denoted that CEACAM1 high- and EPHA2 low-expressing patient samples with lower CEACAM6 expression also exhibited a longer time to first recurrence/metastasis. In HT29 human CRC cells, down-regulation of CEACAM1 along with CEA and CEACAM6 up-regulation led to higher metastatic burden. Overall, CEACAM1-L expression in poorly differentiated CRC can inhibit liver metastasis through cell context-dependent EPHA2-mediated signaling. However, CEACAM1's role should be considered in the presence of other CEACAM family members.

8.
Clin Exp Metastasis ; 34(5): 351-361, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28758175

RESUMEN

Portal vein embolization (PVE) can be required to stimulate liver regeneration before hepatectomy for colorectal liver metastasis (CRCLM), however PVE may also trigger CRCLM progression in patients initially exhibiting chemotherapy response. Using RNA-seq, we aimed to determine the molecular networks involved in metastatic progression in this context. A prospective study including all CRCLM patients undergoing PVE prior to hepatectomy was conducted. Paired biopsies of metastatic lesions were obtained prior to and after PVE and total RNA was isolated and used to prepare Illumina rRNA-depleted TruSeq stranded cDNA libraries for HiSeq 100 bp paired-end sequencing. Patients were classified with progression of disease (PDPVE) or stable disease (SDPVE) post-PVE using 3D-CT tumor volumetric analysis. RESULTS: Twenty patients were included, 13 (65.0%) in the PDPVE group (median 58.0% (18.6-234.3) increase in tumor volume) and 7 (35.0%) in the SDPVE group exhibiting continuous chemotherapy response (median -14.3% (-40.8 to -2.8) decrease in tumor volume) (p < 0.0001). Our results showed that progressive CRCLM after PVE undergo gene expression changes that indicate activation of core cancer pathways (IL-17 (p = 5.94 × 10-03), PI3K (p = 8.71 × 10-03), IL6 and IGF-1 signaling pathways), consistent with changes driven by cytokines and growth factors. Differential expression analysis in a paired model of progression (EdgeR, DeSeq) identified significantly dysregulated genes in the PDPVE group (FOS, FOSB, RAB20, IRS2). CONCLUSION: Differentially expressed genes and pathways with known links to cancer and metastasis were identified post-PVE in patients with disease progression. Highlighting these molecular changes is a crucial first step towards development of targeted therapeutic strategies that may mitigate the effects of PVE on tumor growth.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Embolización Terapéutica , Neoplasias Hepáticas/metabolismo , Anciano , Quimioterapia Adyuvante , Neoplasias Colorrectales/secundario , Neoplasias Colorrectales/terapia , Terapia Combinada , Progresión de la Enfermedad , Femenino , Hepatectomía , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Regeneración Hepática , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular , Estudios Prospectivos , Análisis de Secuencia de ARN , Transcriptoma
9.
Oncoimmunology ; 6(7): e1328336, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28811966

RESUMEN

CEACAM1 is an extensively studied cell surface molecule with established functions in multiple cancer types, as well as in various compartments of the immune system. Due to its multi-faceted role as a recently appreciated immune checkpoint inhibitor and tumor marker, CEACAM1 is an attractive target for cancer immunotherapy. Herein, we highlight CEACAM1's function in various immune compartments and cancer types, including in the context of metastatic disease. This review outlines CEACAM1's role as a therapeutic target for cancer treatment in light of these properties.

10.
Mol Carcinog ; 56(3): 1030-1040, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27597531

RESUMEN

The common R653Q variant (∼20% homozygosity in Caucasians) in the synthetase domain of the folate-metabolizing enzyme MTHFD1 reduces purine synthesis. Although this variant does not appear to affect risk for colorectal cancer, we questioned whether it would affect growth of colorectal tumors. We induced tumor formation in a mouse model for MTHFD1-synthetase deficiency (Mthfd1S+/- ) using combined administration of azoxymethane (AOM) and dextran sodium sulfate (DSS) in male and female wild-type and Mthfd1S+/- mice. Tumor size was significantly smaller in MthfdS+/- mice, particularly in males. A reduction in the proliferation of MthfdS+/- mouse embryonic fibroblast cell lines, compared with wild-type lines, was also observed. Tumor number was not influenced by genotype. The amount of inflammation observed within tumors from male Mthfd1S+/- mice was lower than that in wild-type mice. Gene expression analysis in tumor adjacent normal (pre-neoplastic) tissue identified several genes involved in proliferation (Fosb, Fos, Ptk6, Esr2, Atf3) and inflammation (Atf3, Saa1, TNF-α) that were downregulated in MthfdS+/- males. In females, MthfdS+/- genotype was not associated with these gene expression changes, or with differences in tumor inflammation. These findings suggest that the mechanisms directing tumor growth differ significantly between males and females. We suggest that restriction of purine synthesis, reduced expression of genes involved in proliferation, and/or reduced inflammation lead to slower tumor growth in MTHFD1-synthetase deficiency. These findings may have implications for CRC tumor growth and prognosis in individuals with the R653Q variant. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Aminohidrolasas/deficiencia , Neoplasias Colorrectales/patología , Formiato-Tetrahidrofolato Ligasa/deficiencia , Meteniltetrahidrofolato Ciclohidrolasa/deficiencia , Metilenotetrahidrofolato Deshidrogenasa (NADP)/deficiencia , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Antígenos de Histocompatibilidad Menor/genética , Complejos Multienzimáticos/deficiencia , Enzimas Multifuncionales/deficiencia , Polimorfismo de Nucleótido Simple , Animales , Azoximetano/efectos adversos , Proliferación Celular , Células Cultivadas , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Sulfato de Dextran/efectos adversos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones
11.
Nat Nanotechnol ; 11(11): 941-947, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27525475

RESUMEN

Oxygen-depleted hypoxic regions in the tumour are generally resistant to therapies. Although nanocarriers have been used to deliver drugs, the targeting ratios have been very low. Here, we show that the magneto-aerotactic migration behaviour of magnetotactic bacteria, Magnetococcus marinus strain MC-1 (ref. 4), can be used to transport drug-loaded nanoliposomes into hypoxic regions of the tumour. In their natural environment, MC-1 cells, each containing a chain of magnetic iron-oxide nanocrystals, tend to swim along local magnetic field lines and towards low oxygen concentrations based on a two-state aerotactic sensing system. We show that when MC-1 cells bearing covalently bound drug-containing nanoliposomes were injected near the tumour in severe combined immunodeficient beige mice and magnetically guided, up to 55% of MC-1 cells penetrated into hypoxic regions of HCT116 colorectal xenografts. Approximately 70 drug-loaded nanoliposomes were attached to each MC-1 cell. Our results suggest that harnessing swarms of microorganisms exhibiting magneto-aerotactic behaviour can significantly improve the therapeutic index of various nanocarriers in tumour hypoxic regions.


Asunto(s)
Alphaproteobacteria , Neoplasias Colorrectales/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Compuestos Férricos , Campos Magnéticos , Nanopartículas/química , Neoplasias Experimentales/tratamiento farmacológico , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Compuestos Férricos/química , Compuestos Férricos/farmacología , Células HCT116 , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Hipoxia/patología , Ratones , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Oncotarget ; 7(44): 71651-71659, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27690217

RESUMEN

The expression and activities of RNA binding proteins are frequently dysregulated in human cancer. Their roles, however, appears to be complex, with reports indicating both pro-tumorigenic and tumor suppressive functions. Here we show, using two classical mouse cancer models, that the role of KH-type RNA binding protein, Sam68, in tumor development can be influenced by the status of the p53 tumor suppressor. We demonstrate that in mice expressing wild type p53, Sam68-deficiency resulted in a higher incidence and malignancy of carcinogen-induced tumors, suggesting a tumor suppressive role for Sam68. In marked contrast, Sam68-haploinsufficiency significantly delayed the onset of tumors in mice lacking p53 and prolonged their survival, indicating that Sam68 accelerates the development of p53-deficient tumors. These findings provide considerable insight into a previously unknown relationship between Sam68 and the p53 tumor suppressor in tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Carcinogénesis , Proteínas de Unión al ARN/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Azoximetano , Haploinsuficiencia , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/análisis
13.
Oncotarget ; 7(39): 63730-63746, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27572314

RESUMEN

We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, ß-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of ß-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/ß-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate ß-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal ß-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of ß-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of ß-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antígeno Carcinoembrionario/metabolismo , Carcinoma/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/metabolismo , Animales , Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Técnicas In Vitro , Neoplasias Mamarias Experimentales/patología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal , Regulación hacia Arriba , beta Catenina/metabolismo
14.
PLoS One ; 11(4): e0153933, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100181

RESUMEN

Mutational processes and signatures that drive early tumorigenesis are centrally important for early cancer prevention. Yet, to date, biomarkers and risk factors for polyps (adenomas) that inordinately and rapidly develop into colon cancer remain poorly defined. Here, we describe surprisingly high mutational profiles through whole-genome sequence (WGS) analysis in 2 of 4 pairs of benign colorectal adenoma tissue samples. Unsupervised hierarchical clustered transcriptomic analysis of a further 7 pairs of adenomas reveals distinct mutational signatures regardless of adenoma size. Transitional single nucleotide substitutions of C:G>T:A predominate in the adenoma mutational spectrum. Strikingly, we observe mutations in the TGF-ß pathway and CEA-associated genes in 4 out of 11 adenomas, overlapping with the Wnt pathway. Immunohistochemical labeling reveals a nearly 5-fold increase in CEA levels in 23% of adenoma samples with a concomitant loss of TGF-ß signaling. We also define a functional role by which the CEA B3 domain interacts with TGFBR1, potentially inactivating the tumor suppressor function of TGF-ß signaling. Our study uncovers diverse mutational processes underlying the transition from early adenoma to cancer. This has broad implications for biomarker-driven targeting of CEA/TGF-ß in high-risk adenomas and may lead to early detection of aggressive adenoma to CRC progression.


Asunto(s)
Adenoma/genética , Antígeno Carcinoembrionario/genética , Colon/metabolismo , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Mutación/genética , Factor de Crecimiento Transformador beta/genética , Adenoma/metabolismo , Adenoma/patología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Antígeno Carcinoembrionario/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colon/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Progresión de la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
15.
Mamm Genome ; 27(5-6): 213-24, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26979842

RESUMEN

Inbred strains of mice differ in susceptibility to colitis-associated colorectal cancer (CA-CRC). We tested 10 inbred strains of mice for their response to azoxymethane/dextran sulfate sodium-induced CA-CRC and identified a bimodal inter-strain distribution pattern when tumor multiplicity was used as a phenotypic marker of susceptibility. The FVB/NJ strain was particularly susceptible showing a higher tumor burden than any other susceptible strains (12.5-week post-treatment initiation). FVB/NJ hyper-susceptibility was detected as early as 8-week post-treatment initiation with FVB/NJ mice developing 5.5-fold more tumors than susceptible A/J or resistant B6 control mice. Linkage analysis by whole genome scan in informative (FVB/NJ×C3H/HeJ)F2 mice identified a novel susceptibility locus designated as C olon c ancer s usceptibility 6 (Ccs6) on proximal mouse chromosome 6. When gender was used as a covariate, a LOD score of 5.4 was computed with the peak marker being positioned at rs13478727, 43.8 Mbp. Mice homozygous for FVB/NJ alleles at this locus had increased tumor multiplicity compared to homozygous C3H/HeJ mice. Positional candidates in this region of chromosome 6 were analyzed with respect to a possible role in carcinogenesis and a role in inflammatory response using a new epigenetic gene scoring tool (Myeloid Inflammation Score).


Asunto(s)
Colitis/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Sitios de Carácter Cuantitativo/genética , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Colitis/complicaciones , Colitis/patología , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/patología , Ligamiento Genético , Homocigoto , Humanos , Ratones , Fenotipo
16.
Gut ; 65(5): 821-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25666195

RESUMEN

OBJECTIVE: Nearly 20%-29% of patients with colorectal cancer (CRC) succumb to liver or lung metastasis and there is a dire need for novel targets to improve the survival of patients with metastasis. The long isoform of the Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1-L or CC1-L) is a key regulator of immune surveillance in primary CRC, but its role in metastasis remains largely unexplored. We have examined how CC1-L expression impacts on colon cancer liver metastasis. DESIGN: Murine MC38 transfected with CC1-L were evaluated in vitro for proliferation, migration and invasion, and for in vivo experimental liver metastasis. Using shRNA silencing or pharmacological inhibition, we delineated the role in liver metastasis of Chemokine (C-C motif) Ligand 2 (CCL2) and Signal Transducer and Activator of Transcription 3 (STAT3) downstream of CC1-L. We further assessed the clinical relevance of these findings in a cohort of patients with CRC. RESULTS: MC38-CC1-L-expressing cells exhibited significantly reduced in vivo liver metastasis and displayed decreased CCL2 chemokine secretion and reduced STAT3 activity. Down-modulation of CCL2 expression and pharmacological inhibition of STAT3 activity in MC38 cells led to reduced cell invasion capacity and decreased liver metastasis. The clinical relevance of our findings is illustrated by the fact that high CC1 expression in patients with CRC combined with some inflammation-regulated and STAT3-regulated genes correlate with improved 10-year survival. CONCLUSIONS: CC1-L regulates inflammation and STAT3 signalling and contributes to the maintenance of a less-invasive CRC metastatic phenotype of poorly differentiated carcinomas.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Animales , Diferenciación Celular , Neoplasias Colorrectales/patología , Femenino , Humanos , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Isoformas de Proteínas/fisiología , Células Tumorales Cultivadas
17.
Platelets ; 27(2): 168-77, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26196244

RESUMEN

Previous studies have implicated that the Ig-ITIM superfamily member, CEACAM1 may regulate integrin function. While CEACAM1 has been demonstrated to play a role as an inhibitory co-receptor of ITAM-associated GPVI/FcR γ-chain signaling pathways in platelets, its physiologic role in integrin αIIbß3-mediated platelet function is unclear. In this study, we investigate the functional importance of Ceacam1 in murine platelets. We show that CEACAM1 is constitutively associated with integrin αIIbß3 in resting human and mouse platelets as demonstrated by co-immunoprecipitation studies. Using Ceacam1-deficient mice, we show that they have prolonged tail bleeding times and volume of blood lost that is corrected by reconstitution with platelet Ceacam1. Ceacam1(-/-) platelets have moderate integrin αIIbß3 mediated functional defects with impaired kinetics of platelet spreading on fibrinogen and failure to retract fibrin clots in vitro. This functional integrin αIIbß3 defect could not be attributed to altered integrin αIIbß3 expression. Ceacam1(-/-) platelets displayed normal "inside-out" signaling properties as demonstrated by normal agonist-induced binding of soluble (fluorescein isothiocyanate) FITC-fibrinogen, JON/A antibody binding, and increases in cytosolic free calcium levels. This study provides direct evidence that Ceacam1 is essential for normal integrin αIIbß3-mediated platelet function and that disruption of mouse Ceacam1 induced moderate integrin αIIbß3-mediated functional defects.


Asunto(s)
Plaquetas/metabolismo , Antígeno Carcinoembrionario/sangre , Fibrina/metabolismo , Fibrinógeno/metabolismo , Integrina alfa2/sangre , Animales , Tiempo de Sangría , Coagulación Sanguínea , Plaquetas/patología , Calcio/sangre , Antígeno Carcinoembrionario/genética , Retracción del Coagulo , Fibrina/genética , Fibrinógeno/genética , Expresión Génica , Humanos , Integrina alfa2/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
18.
World J Gastroenterol ; 21(41): 11688-99, 2015 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-26556996

RESUMEN

Colitis-associated colorectal cancer (CA-CRC) is the cause of death in 10%-15% of inflammatory bowel disease (IBD) patients. CA-CRC results from the accumulation of mutations in intestinal epithelial cells and progresses through a well-characterized inflammation to dysplasia to carcinoma sequence. Quantitative estimates of overall CA-CRC risks are highly variable ranging from 2% to 40% depending on IBD severity, duration and location, with IBD duration being the most significant risk factor associated with CA-CRC development. Recently, studies have identified IBD patients with similar patterns of colonic inflammation, but that differ with respect to CA-CRC development, suggesting a role for additional non-inflammatory risk factors in CA-CRC development. One suggestion is that select IBD patients carry polymorphisms in various low penetrance disease susceptibility genes, which pre-dispose them to CA-CRC development, although these loci have proven difficult to identify in human genome-wide association studies. Mouse models of CA-CRC have provided a viable alternative for the discovery, validation and study of individual genes in CA-CRC pathology. In this review, we summarize the current CA-CRC literature with a strong focus on genetic pre-disposition and highlight an emerging role for mouse models in the search for CA-CRC risk alleles.


Asunto(s)
Transformación Celular Neoplásica/genética , Colitis/genética , Neoplasias del Colon/genética , Animales , Transformación Celular Neoplásica/inmunología , Colitis/epidemiología , Colitis/inmunología , Neoplasias del Colon/epidemiología , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Ratones , Fenotipo , Factores de Riesgo
19.
Immunity ; 43(4): 751-63, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26384545

RESUMEN

The crosstalk between inflammation and tumorigenesis is now clearly established. However, how inflammation is elicited in the metastatic environment and the corresponding contribution of innate immunity pathways in suppressing tumor growth at secondary sites are poorly understood. Here, we show that mice deficient in Nlrp3 inflammasome components had exacerbated liver colorectal cancer metastatic growth, which was mediated by impaired interleukin-18 (IL-18) signaling. Control of tumor growth was independent of differential cancer cell colonization or proliferation, intestinal microbiota effects, or tumoricidal activity by the adaptive immune system. Instead, the inflammasome-IL-18 pathway impacted maturation of hepatic NK cells, surface expression of the death ligand FasL, and capacity to kill FasL-sensitive tumors. Our results define a regulatory signaling circuit within the innate immune system linking inflammasome activation to effective NK-cell-mediated tumor attack required to suppress colorectal cancer growth in the liver.


Asunto(s)
Adenocarcinoma/secundario , Proteínas Portadoras/fisiología , Neoplasias Colorrectales/patología , Inflamasomas/fisiología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/secundario , Adenocarcinoma/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas de Unión al Calcio/deficiencia , Caspasa 1/deficiencia , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/deficiencia , Proteína Ligando Fas/fisiología , Microbioma Gastrointestinal , Inmunidad Innata , Vigilancia Inmunológica , Inflamasomas/deficiencia , Interleucina-18/fisiología , Interleucina-1beta/fisiología , Neoplasias Hepáticas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/fisiología , Quimera por Radiación , Tolerancia a Radiación , Microambiente Tumoral
20.
Nat Commun ; 6: 6217, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25692415

RESUMEN

B cells are essential for antiviral immune defence because they produce neutralizing antibodies, present antigen and maintain the lymphoid architecture. Here we show that intrinsic signalling of CEACAM1 is essential for generating efficient B-cell responses. Although CEACAM1 exerts limited influence on the proliferation of B cells, expression of CEACAM1 induces survival of proliferating B cells via the BTK/Syk/NF-κB-axis. The absence of this signalling cascade in naive Ceacam1(-/-) mice limits the survival of B cells. During systemic infection with cytopathic vesicular stomatitis virus, Ceacam1(-/-) mice can barely induce neutralizing antibody responses and die early after infection. We find, therefore, that CEACAM1 is a crucial regulator of B-cell survival, influencing B-cell numbers and protective antiviral antibody responses.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos B/citología , Antígeno Carcinoembrionario/fisiología , Animales , Anticuerpos Neutralizantes/inmunología , Linfocitos B/inmunología , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Citometría de Flujo , Regulación de la Expresión Génica , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Transducción de Señal , Bazo/metabolismo , Vesiculovirus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA