Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 59(12): 1171-1178, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35803701

RESUMEN

BACKGROUND: Lowe syndrome (LS) is an X linked disease caused by pathogenic variants in the OCRL gene that impacts approximately 1 in 500 000 children. Classic features include congenital cataract, cognitive/behavioural impairment and renal tubulopathy. METHODS: This study is a retrospective review of clinical features reported by family based survey conducted by Lowe Syndrome Association. Frequency of non-ocular clinical feature(s) of LS and their age of onset was summarised. An LS-specific therapy effectiveness scale was used to assess the response to the administered treatment. Expression of OCRL and relevant neuropeptides was measured in postmortem human brain by qPCR. Gene expression in the mouse brain was determined by reanalysis of publicly available bulk and single cell RNA sequencing. RESULTS: A total of 137 individuals (1 female, 89.1% white, median age 14 years (range 0.8-56)) were included in the study. Short stature (height <3rd percentile) was noted in 81% (n=111) individuals, and 15% (n=20) received growth hormone therapy. Undescended testis was reported in 47% (n=64), and median age of onset of puberty was 15 years. Additional features were dental problems (n=77, 56%), bone fractures (n=63, 46%), hypophosphataemia (n=60, 44%), developmental delay and behavioural issues. OCRL is expressed in human and mouse hypothalami, and in hypothalamic cell clusters expressing Ghrh, Sst, Oxt, Pomc and pituitary cells expressing Gh and Prl. CONCLUSIONS: There is a wide spectrum of the clinical phenotype of LS. Some of the features may be partly driven by the loss of function of OCRL in the hypothalamus and the pituitary.


Asunto(s)
Catarata , Síndrome Oculocerebrorrenal , Niño , Masculino , Animales , Ratones , Femenino , Humanos , Lactante , Preescolar , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fenotipo , Catarata/genética , Encéfalo/metabolismo
2.
PLoS Genet ; 11(10): e1005569, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26496357

RESUMEN

Skeletal dysplasias are a common, genetically heterogeneous cause of short stature that can result from disruptions in many cellular processes. We report the identification of the lesion responsible for skeletal dysplasia and male infertility in the spontaneous, recessive mouse mutant chagun. We determined that Poc1a, encoding protein of the centriole 1a, is disrupted by the insertion of a processed Cenpw cDNA, which is flanked by target site duplications, suggestive of a LINE-1 retrotransposon-mediated event. Mutant fibroblasts have impaired cilia formation and multipolar spindles. Male infertility is caused by defective spermatogenesis early in meiosis and progressive germ cell loss. Spermatogonial stem cell transplantation studies revealed that Poc1a is essential for normal function of both Sertoli cells and germ cells. The proliferative zone of the growth plate is small and disorganized because chondrocytes fail to re-align after cell division and undergo increased apoptosis. Poc1a and several other genes associated with centrosome function can affect the skeleton and lead to skeletal dysplasias and primordial dwarfisms. This mouse mutant reveals how centrosome dysfunction contributes to defects in skeletal growth and male infertility.


Asunto(s)
Proteínas del Citoesqueleto/genética , Enanismo/genética , Infertilidad Masculina/genética , Elementos de Nucleótido Esparcido Largo/genética , Espermatogénesis/genética , Animales , Proteínas de Ciclo Celular , Centriolos/genética , Centrosoma/metabolismo , Proteínas Cromosómicas no Histona/genética , Enanismo/patología , Humanos , Infertilidad Masculina/patología , Masculino , Meiosis/genética , Ratones , Proteínas/genética , Proteínas/metabolismo , Células de Sertoli/metabolismo , Espermatogonias/metabolismo
3.
J Pathol ; 232(1): 43-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24014111

RESUMEN

We have investigated the role of cytokine lymphotoxin in tumour-stromal interactions in human ovarian cancer. We found that lymphotoxin overexpression is commonly shared by the cancer cells of various ovarian cancer subtypes, and lymphotoxin-beta receptor (LTBR) is expressed ubiquitously in both the cancer cells and cancer-associated fibroblasts (CAFs). In monoculture, we showed that ovarian cancer cells are not the major lymphotoxin-responsive cells. On the other hand, our co-culture studies demonstrated that the cancer cell-derived lymphotoxin induces chemokine expression in stromal fibroblasts through LTBR-NF-κB signalling. Amongst the chemokines being produced, we found that fibroblast-secreted CXCL11 promotes proliferation and migration of ovarian cancer cells via the chemokine receptor CXCR3. CXCL11 is highly expressed in CAFs in ovarian cancer biopsies, while CXCR3 is found in malignant cells in primary ovarian tumours. Additionally, the overexpression of CXCR3 is significantly associated with the tumour grade and lymph node metastasis of ovarian cancer, further supporting the role of CXCR3, which interacts with CXCL11, in promoting growth and metastasis of human ovarian cancer. Taken together, these results demonstrated that cancer-cell-derived lymphotoxin mediates reciprocal tumour-stromal interactions in human ovarian cancer by inducing CXCL11 in fibroblasts. Our findings suggest that lymphotoxin-LTBR and CXCL11-CXCR3 signalling represent therapeutic targets in ovarian cancer.


Asunto(s)
Quimiocina CXCL11/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/metabolismo , Neoplasias Ováricas/patología , Receptores CXCR3/metabolismo , Transducción de Señal , Línea Celular Tumoral , Quimiocina CXCL11/genética , Técnicas de Cocultivo , Células Epiteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Hong Kong , Humanos , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/genética , Neoplasias Ováricas/metabolismo , Receptores CXCR3/genética , Microambiente Tumoral
4.
bioRxiv ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38746430

RESUMEN

Myelin regulatory factor (Myrf) is a critical transcription factor in early retinal and retinal pigment epithelial development, and human variants in MYRF are a cause for nanophthalmos. Single cell RNA sequencing (scRNAseq) was performed on Myrf conditional knockout mice ( Rx>Cre Myrf fl/fl ) at 3 developmental timepoints. Myrf was expressed specifically in the RPE, and expression was abrogated in Rx>Cre Myrf fl/fl eyes. scRNAseq analysis revealed a loss of RPE cells at all timepoints resulting from cell death. GO-term analysis in the RPE revealed downregulation of melanogenesis and anatomic structure morphogenesis pathways, which were supported by electron microscopy and histologic analysis. Novel structural target genes including Ermn and Upk3b , along with macular degeneration and inherited retinal disease genes were identified as downregulated, and a strong upregulation of TGFß/BMP signaling and effectors was observed. Regulon analysis placed Myrf downstream of Pax6 and Mitf and upstream of Sox10 in RPE differentiation. Together, these results suggest a strong role for Myrf in the RPE maturation by regulating melanogenesis, cell survival, and cell structure, in part acting through suppression of TGFß signaling and activation of Sox10 . SUMMARY STATEMENT: Myrf regulates RPE development, melanogenesis, and is important for cell structure and survival, in part through regulation of Ermn , Upk3b and Sox10, and BMP/TGFb signaling.

5.
Nat Rev Endocrinol ; 20(2): 77-92, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38102391

RESUMEN

Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Animales , Ratones , Células Madre Pluripotentes Inducidas/metabolismo , Hipófisis/metabolismo , Factores de Transcripción/metabolismo , Transducción de Señal , Diferenciación Celular
6.
Genome Med ; 16(1): 75, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822427

RESUMEN

BACKGROUND: Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS: The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS: Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS: The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.


Asunto(s)
Hipopituitarismo , Ratones Noqueados , Hipófisis , Hipopituitarismo/genética , Animales , Humanos , Hipófisis/metabolismo , Hipófisis/anomalías , Hipófisis/patología , Ratones , Fenotipo , Femenino , Masculino , Modelos Animales de Enfermedad , Secuenciación del Exoma , Displasia Septo-Óptica/genética
7.
Endocrinology ; 164(6)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37183548

RESUMEN

The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations.


Asunto(s)
Enfermedades de la Hipófisis , Adenohipófisis , Embarazo , Femenino , Ratones , Animales , Tirotropina/metabolismo , Hipófisis/metabolismo , Factores de Transcripción/metabolismo , Enfermedades de la Hipófisis/metabolismo , Inmunohistoquímica , Adenohipófisis/metabolismo , Factores de Transcripción SOXB2/metabolismo
8.
Endocrinology ; 161(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913463

RESUMEN

Dietary vitamin A is metabolized into bioactive retinoic acid (RA) in vivo and regulates the development of many embryonic tissues. RA signaling is active in the oral ectoderm-derived tissues of the neuroendocrine system, but its role there has not yet been fully explored. We show here that RA signaling is active during pituitary organogenesis and dependent on the pituitary transcription factor Prop1. Prop1-mutant mice show reduced expression of the aldehyde dehydrogenase gene Aldh1a2, which metabolizes the vitamin A-intermediate retinaldehyde into RA. To elucidate the specific function of RA signaling during neuroendocrine development, we studied a conditional deletion of Aldh1a2 and a dominant-negative mouse model of inhibited RA signaling during pituitary organogenesis. These models partially phenocopy Prop1-mutant mice by exhibiting embryonic pituitary dysmorphology and reduced hormone expression, especially thyrotropin. These findings establish the role of RA in embryonic pituitary stem cell progression to differentiated hormone cells and raise the question of gene-by-environment interactions as contributors to pituitary development and disease.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/metabolismo , Proteínas de Homeodominio/metabolismo , Hipófisis/embriología , Retinal-Deshidrogenasa/metabolismo , Tretinoina/metabolismo , Familia de Aldehído Deshidrogenasa 1/genética , Animales , Ratones Endogámicos C57BL , Organogénesis , Hipófisis/metabolismo , Retinal-Deshidrogenasa/genética , Transducción de Señal , Células Madre/metabolismo , Tirotropina/metabolismo
9.
J Mol Endocrinol ; 65(2): R35-R51, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32485670

RESUMEN

In the last 15 years, single-cell technologies have become robust and indispensable tools to investigate cell heterogeneity. Beyond transcriptomic, genomic and epigenome analyses, technologies are constantly evolving, in particular toward multi-omics, where analyses of different source materials from a single cell are combined, and spatial transcriptomics, where resolution of cellular heterogeneity can be detected in situ. While some of these techniques are still being optimized, single-cell RNAseq has commonly been used because the examination of transcriptomes allows characterization of cell identity and, therefore, unravel previously uncharacterized diversity within cell populations. Most endocrine organs have now been investigated using this technique, and this has given new insights into organ embryonic development, characterization of rare cell types, and disease mechanisms. Here, we highlight recent studies, particularly on the hypothalamus and pituitary, and examine recent findings on the pancreas and reproductive organs where many single-cell experiments have been performed.


Asunto(s)
Sistema Endocrino/citología , Análisis de la Célula Individual , Animales , Humanos , Morfogénesis , Reproducción , Procesos de Determinación del Sexo , Transcriptoma/genética
11.
J Mater Chem B ; 6(19): 3078-3084, 2018 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32254342

RESUMEN

We report on the sizable production of fluorescent nanodiamonds (FNDs) containing a near infrared (NIR) color center - namely the silicon vacancy (SiV) defect, and their first demonstration inside cells for bio-imaging. We further demonstrate a concept of multi-color bio-imaging using FNDs to investigate intercellular processes using two types of FNDs. Due to their specific spectral properties, SiV FNDs can be distinguished from common nitrogen-vacancy (NV) FNDs and show a distinct initial spreading throughout the cell interior. The reported results are the first demonstration of multi-color labeling with FNDs that are especially interesting for in vivo bio-imaging due to their stable fluorescence.

12.
Elife ; 72018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29578405

RESUMEN

The pituitary is an essential endocrine gland regulating multiple processes. Regeneration of endocrine cells is of therapeutic interest and recent studies are promising, but mechanisms of endocrine cell fate acquisition need to be better characterised. The NOTCH pathway is important during pituitary development. Here, we further characterise its role in the murine pituitary, revealing differential sensitivity within and between lineages. In progenitors, NOTCH activation blocks cell fate acquisition, with time-dependant modulation. In differentiating cells, response to activation is blunted in the POU1F1 lineage, with apparently normal cell fate specification, while POMC cells remain sensitive. Absence of apparent defects in Pou1f1-Cre; Rbpjfl/fl mice further suggests no direct role for NOTCH signalling in POU1F1 cell fate acquisition. In contrast, in the POMC lineage, NICD expression induces a regression towards a progenitor-like state, suggesting that the NOTCH pathway specifically blocks POMC cell differentiation. These results have implications for pituitary development, plasticity and regeneration. Activation of NOTCH signalling in different cell lineages of the embryonic murine pituitary uncovers an unexpected differential sensitivity, and this consequently reveals new aspects of endocrine lineages development and plasticity.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Endocrinas/fisiología , Hipófisis/fisiología , Receptores Notch/metabolismo , Animales , Regulación de la Expresión Génica , Ratones , Transducción de Señal
13.
Endocrinology ; 159(12): 3910-3924, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335147

RESUMEN

Transcription factors and signaling pathways that regulate stem cells and specialized hormone-producing cells in the pituitary gland have been the subject of intense study and have yielded a mechanistic understanding of pituitary organogenesis and disease. However, the regulation of stem cell proliferation and differentiation, the heterogeneity among specialized hormone-producing cells, and the role of nonendocrine cells in the gland remain important, unanswered questions. Recent advances in single-cell RNA sequencing (scRNAseq) technologies provide new avenues to address these questions. We performed scRNAseq on ∼13,663 cells pooled from six whole pituitary glands of 7-week-old C57BL/6 male mice. We identified pituitary endocrine and stem cells in silico, as well as other support cell types such as endothelia, connective tissue, and red and white blood cells. Differential gene expression analyses identify known and novel markers of pituitary endocrine and stem cell populations. We demonstrate the value of scRNAseq by in vivo validation of a novel gonadotrope-enriched marker, Foxp2. We present novel scRNAseq data of in vivo pituitary tissue, including data from agnostic clustering algorithms that suggest the presence of a somatotrope subpopulation enriched in sterol/cholesterol synthesis genes. Additionally, we show that incomplete transcriptome annotation can cause false negatives on some scRNAseq platforms that only generate 3' transcript end sequences, and we use in vivo data to recover reads of the pituitary transcription factor Prop1. Ultimately, scRNAseq technologies represent a significant opportunity to address long-standing questions regarding the development and function of the different populations of the pituitary gland throughout life.


Asunto(s)
Biomarcadores/análisis , Hipófisis/citología , Hipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Análisis de la Célula Individual/métodos , Células Madre/clasificación , Células Madre/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Separación Celular/métodos , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Organogénesis/genética , Hipófisis/embriología , Hormonas Hipofisarias/genética , Análisis de Secuencia de ARN/métodos , Células Madre/citología , Transcriptoma
14.
Mol Cell Endocrinol ; 439: 213-223, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27616671

RESUMEN

The hypothalamic-pituitary axes are the coordinating centers for multiple endocrine gland functions and physiological processes. Defects in the hypothalamus or pituitary gland can cause reduced growth and severe short stature, affecting approximately 1 in 4000 children, and a large percentage of cases of pituitary hormone deficiencies do not have an identified genetic cause. SOX21 is a protein that regulates hair, neural, and trophoblast stem cell differentiation. Mice lacking Sox21 have reduced growth, but the etiology of this growth defect has not been described. We studied the expression of Sox21 in hypothalamic-pituitary development and examined multiple endocrine axes in these mice. We find no evidence of reduced intrauterine growth, food intake, or physical activity, but there is evidence for increased energy expenditure in mutants. In addition, despite changes in pituitary hormone expression, hypothalamic-pituitary axes appear to be functional. Therefore, SOX21 variants may be a cause of non-endocrine short stature in humans.


Asunto(s)
Eliminación de Gen , Hipotálamo/metabolismo , Hipófisis/metabolismo , Factores de Transcripción SOXB2/genética , Animales , Animales Recién Nacidos , Metabolismo Basal , Peso Corporal , Diencéfalo/metabolismo , Femenino , Hormona del Crecimiento/metabolismo , Crecimiento y Desarrollo , Masculino , Ratones Noqueados , Especificidad de Órganos , Tiroxina/sangre
15.
Mol Cell Endocrinol ; 445: 14-26, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27650955

RESUMEN

The anterior pituitary gland is comprised of specialized cell-types that produce and secrete polypeptide hormones in response to hypothalamic input and feedback from target organs. These specialized cells arise from stem cells that express SOX2 and the pituitary transcription factor PROP1, which is necessary to establish the stem cell pool and promote an epithelial to mesenchymal-like transition, releasing progenitors from the niche. The adult anterior pituitary responds to physiological challenge by mobilizing the SOX2-expressing progenitor pool and producing additional hormone-producing cells. Knowledge of the role of signaling pathways and extracellular matrix components in these processes may lead to improvements in the efficiency of differentiation of embryonic stem cells or induced pluripotent stem cells into hormone producing cells in vitro. Advances in our basic understanding of pituitary stem cell regulation and differentiation may lead to improved diagnosis and treatment for patients with hypopituitarism.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Adenohipófisis/citología , Factores de Transcripción SOXB1/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Humanos , Adenohipófisis/metabolismo , Transducción de Señal , Células Madre/metabolismo
16.
Endocr Rev ; 37(6): 636-675, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27828722

RESUMEN

The genetic basis for combined pituitary hormone deficiency (CPHD) is complex, involving 30 genes in a variety of syndromic and nonsyndromic presentations. Molecular diagnosis of this disorder is valuable for predicting disease progression, avoiding unnecessary surgery, and family planning. We expect that the application of high throughput sequencing will uncover additional contributing genes and eventually become a valuable tool for molecular diagnosis. For example, in the last 3 years, six new genes have been implicated in CPHD using whole-exome sequencing. In this review, we present a historical perspective on gene discovery for CPHD and predict approaches that may facilitate future gene identification projects conducted by clinicians and basic scientists. Guidelines for systematic reporting of genetic variants and assigning causality are emerging. We apply these guidelines retrospectively to reports of the genetic basis of CPHD and summarize modes of inheritance and penetrance for each of the known genes. In recent years, there have been great improvements in databases of genetic information for diverse populations. Some issues remain that make molecular diagnosis challenging in some cases. These include the inherent genetic complexity of this disorder, technical challenges like uneven coverage, differing results from variant calling and interpretation pipelines, the number of tolerated genetic alterations, and imperfect methods for predicting pathogenicity. We discuss approaches for future research in the genetics of CPHD.


Asunto(s)
Genómica/métodos , Hipopituitarismo/genética , Animales , Humanos
17.
Cell Stem Cell ; 6(6): 603-15, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20569697

RESUMEN

Recent evidence suggests that a subpopulation of cancer cells, cancer stem cells (CSCs), is responsible for tumor growth in colorectal cancer. However, the role of CSCs in colorectal cancer metastasis is unclear. Here, we identified a subpopulation of CD26(+) cells uniformly present in both the primary and metastatic tumors in colorectal cancer patients with liver metastasis. Furthermore, in patients without distant metastasis at the time of presentation, the presence of CD26(+) cells in their primary tumors predicted distant metastasis on follow-up. Isolated CD26(+) cells, but not CD26(-) cells, led to development of distant metastasis when injected into the mouse cecal wall. CD26(+) cells were also associated with enhanced invasiveness and chemoresistance. Our findings have uncovered a critical role of CSCs in metastatic progression of cancer. Furthermore, the ability to predict metastasis based on analysis of CSC subsets in the primary tumor may have important clinical implication as a selection criterion for adjuvant therapy.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma/diagnóstico , Neoplasias Colorrectales/diagnóstico , Dipeptidil Peptidasa 4/biosíntesis , Neoplasias Hepáticas/diagnóstico , Células Madre Neoplásicas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores de Tumor/genética , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/fisiopatología , Carcinoma/secundario , Ensayos de Migración Celular , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Dipeptidil Peptidasa 4/genética , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Fluorouracilo/farmacología , Estudios de Seguimiento , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/fisiopatología , Neoplasias Hepáticas/secundario , Ratones , Ratones SCID , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Compuestos Organoplatinos/farmacología , Oxaliplatino , Pronóstico , ARN Interferente Pequeño/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA