Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38496665

ABSTRACT

The cloaca is a transient structure that forms in the terminal hindgut giving rise to the rectum dorsally and the urogenital sinus ventrally. Similarly, human hindgut cultures derived from human pluripotent stem cells generate human colonic organoids (HCOs) which also contain co-developing urothelial tissue. In this study, our goal was to identify pathways involved in cloacal patterning and apply this to human hindgut cultures. RNA-seq data comparing dorsal versus ventral cloaca in e10.5 mice revealed that WNT signaling was elevated in the ventral versus dorsal cloaca. Inhibition of WNT signaling in hindgut cultures biased their differentiation towards a colorectal fate. WNT activation biased differentiation towards a urothelial fate, giving rise to human urothelial organoids (HUOs). HUOs contained cell types present in human urothelial tissue. Based on our results, we propose a mechanism whereby WNT signaling patterns the ventral cloaca, prior to cloacal septation, to give rise to the urogenital sinus.

2.
Cell Stem Cell ; 30(11): 1434-1451.e9, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37922878

ABSTRACT

Most organs have tissue-resident immune cells. Human organoids lack these immune cells, which limits their utility in modeling many normal and disease processes. Here, we describe that pluripotent stem cell-derived human colonic organoids (HCOs) co-develop a diverse population of immune cells, including hemogenic endothelium (HE)-like cells and erythromyeloid progenitors that undergo stereotypical steps in differentiation, resulting in the generation of functional macrophages. HCO macrophages acquired a transcriptional signature resembling human fetal small and large intestine tissue-resident macrophages. HCO macrophages modulate cytokine secretion in response to pro- and anti-inflammatory signals and were able to phagocytose and mount a robust response to pathogenic bacteria. When transplanted into mice, HCO macrophages were maintained within the colonic organoid tissue, established a close association with the colonic epithelium, and were not displaced by the host bone-marrow-derived macrophages. These studies suggest that HE in HCOs gives rise to multipotent hematopoietic progenitors and functional tissue-resident macrophages.


Subject(s)
Pluripotent Stem Cells , Humans , Mice , Animals , Hematopoietic Stem Cells , Colon , Organoids , Macrophages
3.
Med. UIS ; 36(1): 69-88, abr. 2023.
Article in Spanish | LILACS | ID: biblio-1534833

ABSTRACT

Introducción: la disfagia es un trastorno de la deglución, el cual es habitualmente desatendido por profesionales de la salud, en especial la disfagia orofaríngea neurogénica, que es capaz de producir varios síntomas, signos y complicaciones secundarias en los pacientes. Objetivo: realizar una caracterización clínica incluyendo percepción de síntomas de disfagia en pacientes con disfagia orofaríngea neurogénica de causas neurológicas y neuromusculares en Antioquia, Colombia entre los años 2019 y 2021. Metodología: estudio transversal realizado en 80 pacientes con disfagia orofaríngea neurogénica confirmada a través de la herramienta Eating Assessment Tool-10, evaluación clínica y/o resultados de video fluoroscopia de la deglución. Resultados: 71 pacientes presentaron causas neurológicas centrales. La enfermedad cerebrovascular y la enfermedad de Parkinson fueron las etiologías más frecuentes. Solo 18% de los pacientes con causas neurológicas y 33% con causas neuromusculares reportaron tolerancia a todas las consistencias de alimentos. Mediana de 16 puntos en cuanto a autopercepción de síntomas de disfagia mediante el instrumento Eating Assessment Tool-10, con puntuaciones más altas en pacientes con presencia de gastrostomía, antecedente de neumonía, odinofagia y alteración en la oclusión mandibular al examen físico. En los pacientes con causas neurológicas hubo mayor presencia de signos motores linguales y apraxias orofaciales. Conclusión: existen características clínicas como sensación de comida pegada, dificultad para tragar alimentos sólidos, tos y ahogo al tragar, que son útiles en el reconocimiento de casos de disfagia orofaríngea, y apoyan que esta genera más síntomas que signos al examen físico en pacientes con condiciones neurológicas y neuromusculares.


Introduction: dysphagia is a swallowing disorder that is usually neglected by health professionals, especially neurogenic oropharyngeal dysphagia, which can produce various symptoms, signs and secondary complications in patients. Objective: to perform a clinical characterization, including perception of dysphagia symptoms, in patients with neurogenic oropharyngeal dysphagia of neurological and neuromuscular causes in Antioquia, Colombia between 2019 and 2021. Methodology: cross-sectional study conducted in 80 patients with neurogenic oropharyngeal dysphagia confirmed through the Eating Assessment Tool-10, clinical assessment and/ or video fluoroscopy results of swallowing. Results: 71 patients presented central neurological causes. Cerebrovascular disease and Parkinson's disease were the most frequent etiologies. Only 18% of patients with neurological causes and 33% with neuromuscular causes reported tolerance to all food consistencies. Median of 16 points in terms of self-perception of dysphagia symptoms using the Eating Assessment Tool-10, with higher scores in patients with gastrostomy, a history of pneumonia, odynophagia, and abnormal mandibular occlusion on physical examination. In patients with neurological causes, there was a greater presence of lingual motor signs and orofacial apraxia. Conclusion: there are clinical characteristics such as a sensation of stuck food, difficulty swallowing solid foods, coughing, and choking when swallowing, which are useful in recognizing cases of oropharyngeal dysphagia, and support that this generates more symptoms than signs on physical examination in patients with neurological and neuromuscular conditions.


Subject(s)
Humans , Male , Female , Deglutition Disorders , Signs and Symptoms , Nervous System Diseases , Neurologic Manifestations , Neuromuscular Diseases
4.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955755

ABSTRACT

Human intestinal organoids (HIOs) generated from human pluripotent stem cells hold great promise for modeling human development and as a possible source of tissue for transplantation. HIOs generate all of the main epithelial and mesenchymal cell types found in the developing human intestine and mature into intestinal tissue with crypts and villi following transplantation into immunocompromised mice. However, incomplete in vitro patterning and the presence of contaminating neurons could hinder their use for regenerative medicine in humans. Based on studies in model organisms, we hypothesized that the treatment of HIOs with all trans retinoic acid (ATRA) would improve their in vitro growth and patterning. We found that ATRA not only improved the patterning of HIOs, ATRA also increased organoid forming efficiency, improved epithelial growth, enriched intestinal subepithelial myofibroblasts (ISEMFs) and reduced neuronal contamination in HIOs. Taken together, our studies demonstrate how the manipulation of a single developmental signaling pathway can be used to improve the survival, patterning and cellular composition of HIOs.


Subject(s)
Organoids , Pluripotent Stem Cells , Animals , Cell Differentiation , Humans , Intestinal Mucosa/metabolism , Intestines , Mice , Tretinoin/metabolism , Tretinoin/pharmacology
5.
J Vis Exp ; (173)2021 07 09.
Article in English | MEDLINE | ID: mdl-34309606

ABSTRACT

Intestinal regional specification describes a process through which unique morphology and function are imparted to defined areas of the developing gastrointestinal (GI) tract. Regional specification in the intestine is driven by multiple developmental pathways, including the bone morphogenetic protein (BMP) pathway. Based on normal regional specification, a method to generate human colonic organoids (HCOs) from human pluripotent stem cells (hPSCs), which include human embryonic stem cells (hES) and induced pluripotent stem cells (iPSCs), was developed. A three-day induction of BMP signaling sufficiently patterns mid/hindgut tube cultures into special AT-rich sequence-binding protein 2 (SATB2)-expressing HCOs containing all of the main epithelial cell types present in human colon as well as co-developing mesenchymal cells. Omission of BMP (or addition of the BMP inhibitor NOGGIN) during this critical patterning period resulted in the formation of human intestinal organoids (HIOs). HIOs and HCOs morphologically and molecularly resemble human developing small intestine and colon, respectively. Despite the utility of HIOs and HCOs for studying human intestinal development, the generation of HIOs and HCOs is challenging. This paper presents methods for generating, maintaining, and characterizing HIOs and HCOs. In addition, the critical steps in the protocol and troubleshooting recommendations are provided.


Subject(s)
Organoids , Pluripotent Stem Cells , Cell Differentiation , Colon , Endoderm , Humans , Intestines
6.
Methods Cell Biol ; 159: 201-227, 2020.
Article in English | MEDLINE | ID: mdl-32586443

ABSTRACT

Advances in human pluripotent stem cell (hPSC) biology now allow the generation of organoids that resemble different regions of the gastrointestinal tract. Generation of region-specific organoids has been facilitated by developmental biology studies carried out in model organisms such as mouse, frog and chick. By mimicking embryonic development, hPSC-derived human colonic organoids (HCOs) can be generated through a stepwise differentiation, first into definitive endoderm (DE), then into mid/hindgut spheroids which are then patterned into posterior gut tissue which gives rise to HCOs following prolonged in vitro culture. HCOs undergo transitions similar to those observed in the developing colon of model organisms and human embryos. HCOs develop into tissue that resembles fetal colon on the basis of morphology, gene expression and presence of differentiated cell types. Generation of HCOs without the proper training or expertise can be a daunting task. Here, we describe a detailed protocol for differentiating hPSCs into HCOs, we include suggestions for troubleshooting these differentiations, and we discuss experimental design considerations. We have also highlighted the key advantages and limitations of the system.


Subject(s)
Cell Culture Techniques/methods , Colon/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation , Endoderm/cytology , Gene Expression Regulation , Human Embryonic Stem Cells/cytology , Humans , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology
7.
Gastroenterology ; 157(6): 1556-1571.e5, 2019 12.
Article in English | MEDLINE | ID: mdl-31442438

ABSTRACT

BACKGROUND & AIMS: It has been a challenge to develop fully functioning cells from human pluripotent stem cells (hPSCs). We investigated how activation of hedgehog signaling regulates derivation of enteric neural crest (NC) cells from hPSCs. METHODS: We analyzed transcriptomes of mouse and hPSC-derived enteric NCs using single-cell RNA sequencing (scRNA-seq) to identify the changes in expression associated with lineage differentiation. Intestine tissues were collected from Tg(GBS-GFP), Sufuf/f; Wnt1-cre, Ptch1+/-, and Gli3Δ699/Δ699 mice and analyzed by flow cytometry and immunofluorescence for levels of messenger RNAs encoding factors in the hedgehog signaling pathway during differentiation of enteric NCs. Human NC cells (HNK-1+p75NTR+) were derived from IMR90 and UE02302 hPSC lines. hPSCs were incubated with a hedgehog agonist (smoothened agonist [SAG]) and antagonists (cyclopamine) and analyzed for differentiation. hPSC-based innervated colonic organoids were derived from these hPSC lines and analyzed by immunofluorescence and neuromuscular coupling assay for expression of neuronal subtype markers and assessment of the functional maturity of the hPSC-derived neurons, respectively. RESULTS: Single-cell RNA sequencing analysis showed that neural fate acquisition by human and mouse enteric NC cells requires reduced expression of NC- and cell cycle-specific genes and up-regulation of neuronal or glial lineage-specific genes. Activation of the hedgehog pathway was associated with progression of mouse enteric NCs to the more mature state along the neuronal and glial lineage differentiation trajectories. Activation of the hedgehog pathway promoted development of cultured hPSCs into NCs of greater neurogenic potential by activating expression of genes in the neurogenic lineage. The hedgehog agonist increased differentiation of hPSCs into cells of the neuronal lineage by up-regulating expression of GLI2 target genes, including INSM1, NHLH1, and various bHLH family members. The hedgehog agonist increased expression of late neuronal markers and neuronal activities in hPSC-derived neurons. CONCLUSIONS: In enteric NCs from humans and mice, activation of hedgehog signaling promotes differentiation into neurons by promoting cell-state transition, expression of genes in the neurogenic lineage, and functional maturity of enteric neurons.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Cell Line , Enteric Nervous System/cytology , Gene Expression Profiling/methods , Hedgehog Proteins/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/innervation , Male , Mice , Mice, Transgenic , Neural Crest/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
9.
Front Med (Lausanne) ; 6: 297, 2019.
Article in English | MEDLINE | ID: mdl-31956653

ABSTRACT

In recent years, advances in human pluripotent stem cell (hPSC) biology have enabled the generation of gastrointestinal (GI) organoids which recapitulate aspects of normal organ development. HPSC derived gastrointestinal organoids are comprised of epithelium and mesenchyme and have a remarkable ability to self-organize and recapitulate early stages of human intestinal development. Furthermore, hPSC derived organoids can be transplanted into immunocompromised mice which allows further maturation of both the epithelium and mesenchyme. In this review, we will briefly summarize work from model systems which has elucidated mechanisms of GI patterning and how these insights have been used to guide the differentiation of hPSCs into organoids resembling small intestine and colon. We will succinctly discuss how developmental principles have been used to promote maturation of human intestinal organoids (HIOs) in vitro as well as to introduce an enteric nervous system into HIOs. We will then concisely review how organoids have been used to study human pathogens, how new genetic and bioengineering tools are being applied to organoid research, and how this integration has allowed researchers to elucidate mechanisms of human development and disease. Finally, we will briefly discuss remaining challenges in the field and how they can be addressed. HPSC derived organoids are promising new model systems which hold the potential of unlocking unknown mechanisms of human gastrointestinal development and disease.

10.
Development ; 145(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30143540

ABSTRACT

Enteroendocrine cells (EECs) are a minor cell population in the intestine yet they play a major role in digestion, satiety and nutrient homeostasis. Recently developed human intestinal organoid models include EECs, but their rarity makes it difficult to study their formation and function. Here, we used the EEC-inducing property of the transcription factor NEUROG3 in human pluripotent stem cell-derived human intestinal organoids and colonic organoids to promote EEC development in vitro An 8-h pulse of NEUROG3 expression induced expression of known target transcription factors and after 7 days organoids contained up to 25% EECs in the epithelium. EECs expressed a broad array of human hormones at the mRNA and/or protein level, including motilin, somatostatin, neurotensin, secretin, substance P, serotonin, vasoactive intestinal peptide, oxyntomodulin, GLP-1 and INSL5. EECs secreted several hormones including gastric inhibitory polypeptide (GIP), ghrelin, GLP-1 and oxyntomodulin. Injection of glucose into the lumen of organoids caused an increase in both GIP secretion and K-cell number. Lastly, we observed formation of all known small intestinal EEC subtypes following transplantation and growth of human intestinal organoids in mice.


Subject(s)
Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Pluripotent Stem Cells/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Differentiation , Hormones/metabolism , Humans , Intestines/cytology , Nerve Tissue Proteins/metabolism , Organoids/cytology , Pluripotent Stem Cells/metabolism , Time Factors , Transcription Factors/metabolism
11.
Cell Stem Cell ; 21(1): 51-64.e6, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28648364

ABSTRACT

Gastric and small intestinal organoids differentiated from human pluripotent stem cells (hPSCs) have revolutionized the study of gastrointestinal development and disease. Distal gut tissues such as cecum and colon, however, have proved considerably more challenging to derive in vitro. Here we report the differentiation of human colonic organoids (HCOs) from hPSCs. We found that BMP signaling is required to establish a posterior SATB2+ domain in developing and postnatal intestinal epithelium. Brief activation of BMP signaling is sufficient to activate a posterior HOX code and direct hPSC-derived gut tube cultures into HCOs. In vitro, HCOs express colonic markers and contained colon-specific cell populations. Following transplantation into mice, HCOs undergo morphogenesis and maturation to form tissue that exhibits molecular, cellular, and morphologic properties of human colon. Together these data show BMP-dependent patterning of human hindgut into HCOs, which will be valuable for studying diseases including colitis and colon cancer.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Colon/metabolism , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction , Animals , Colon/cytology , Heterografts , Humans , Mice , Mice, Inbred NOD , Organoids/cytology , Organoids/transplantation , Pluripotent Stem Cells/cytology
12.
Methods Mol Biol ; 1597: 167-177, 2017.
Article in English | MEDLINE | ID: mdl-28361317

ABSTRACT

Over the past several decades, developmental biologists have discovered fundamental mechanisms by which organs form in developing embryos. With this information it is now possible to generate human "organoids" by the stepwise differentiation of human pluripotent stem cells using a process that recapitulates organ development. For the gastrointestinal tract, one of the first key steps is the formation of definitive endoderm and mesoderm, a process that relies on the TGFb molecule Nodal. Endoderm is then patterned along the anterior-posterior axis, with anterior endoderm forming the foregut and posterior endoderm forming the mid and hindgut. A-P patterning of the endoderm is accomplished by the combined activities of Wnt, BMP, and FGF. High Wnt and BMP promote a posterior fate, whereas repressing these pathways promotes an anterior endoderm fate. The stomach derives from the posterior foregut and retinoic acid signaling is required for promoting a posterior foregut fate. The small and large intestine derive from the mid and hindgut, respectively.These stages of gastrointestinal development can be precisely manipulated through the temporal activation and repression of the pathways mentioned above. For example, stimulation of the Nodal pathway with the mimetic Activin A, another TGF-ß superfamily member, can trigger the differentiation of pluripotent stem cells into definitive endoderm (D'Amour et al., Nat Biotechnol 23:1534-1541, 2005). Exposure of definitive endoderm to high levels of Wnt and FGF promotes the formation of posterior endoderm and mid/hindgut tissue that expresses CDX2. Mid-hindgut spheroids that are cultured in a three-dimensional matrix form human intestinal organoids (HIOs) that are small intestinal in nature Spence et al., Nature 2011. In contrast, activation of FGF and Wnt in the presence of the BMP inhibitor Noggin promotes the formation of anterior endoderm and foregut tissues that express SOX2. These SOX2-expressing foregut spheroids can be further patterned into posterior foregut by addition of retinoic acid. Once formed, these posterior foregut spheroids can be grown in three-dimensional human gastric organoids (HGOs) that have all of the cell types of antral part of the stomach (Mc Cracken et al. 2014).Here, we describe the detailed methods for generating stomach/human gastric organoids (HGOs) and human intestinal organoids (HIOs) from human pluripotent stem cells. We first present a method for generating definitive endoderm from pluripotent stem cells followed by differentiation of definitive endoderm into either posterior foregut spheroids or mid-hindgut spheroids. We then describe how three-dimensional culturing of these spheroids results in the formation of HGOs and HIOs, respectively.


Subject(s)
Human Embryonic Stem Cells/cytology , Intestines/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Stomach/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Endoderm/cytology , Humans , Signal Transduction/physiology
13.
Development ; 144(6): 1128-1136, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28174251

ABSTRACT

Temporal manipulation of the in vitro environment and growth factors can direct differentiation of human pluripotent stem cells into organoids - aggregates with multiple tissue-specific cell types and three-dimensional structure mimicking native organs. A mechanistic understanding of early organoid formation is essential for improving the robustness of these methods, which is necessary prior to use in drug development and regenerative medicine. We investigated intestinal organoid emergence, focusing on measurable parameters of hindgut spheroids, the intermediate step between definitive endoderm and mature organoids. We found that 13% of spheroids were pre-organoids that matured into intestinal organoids. Spheroids varied by several structural parameters: cell number, diameter and morphology. Hypothesizing that diameter and the morphological feature of an inner mass were key parameters for spheroid maturation, we sorted spheroids using an automated micropipette aspiration and release system and monitored the cultures for organoid formation. We discovered that populations of spheroids with a diameter greater than 75 µm and an inner mass are enriched 1.5- and 3.8-fold for pre-organoids, respectively, thus providing rational guidelines towards establishing a robust protocol for high quality intestinal organoids.


Subject(s)
Organoids/growth & development , Tissue Engineering/methods , Cell Count , Cell Size , Cells, Cultured , Digestive System/cytology , Flow Cytometry , Humans , Organoids/cytology , Spheroids, Cellular/cytology
14.
Nat Med ; 20(11): 1310-4, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25326803

ABSTRACT

Differentiation of human pluripotent stem cells (hPSCs) into organ-specific subtypes offers an exciting avenue for the study of embryonic development and disease processes, for pharmacologic studies and as a potential resource for therapeutic transplant. To date, limited in vivo models exist for human intestine, all of which are dependent upon primary epithelial cultures or digested tissue from surgical biopsies that include mesenchymal cells transplanted on biodegradable scaffolds. Here, we generated human intestinal organoids (HIOs) produced in vitro from human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) that can engraft in vivo. These HIOs form mature human intestinal epithelium with intestinal stem cells contributing to the crypt-villus architecture and a laminated human mesenchyme, both supported by mouse vasculature ingrowth. In vivo transplantation resulted in marked expansion and maturation of the epithelium and mesenchyme, as demonstrated by differentiated intestinal cell lineages (enterocytes, goblet cells, Paneth cells, tuft cells and enteroendocrine cells), presence of functional brush-border enzymes (lactase, sucrase-isomaltase and dipeptidyl peptidase 4) and visible subepithelial and smooth muscle layers when compared with HIOs in vitro. Transplanted intestinal tissues demonstrated digestive functions as shown by permeability and peptide uptake studies. Furthermore, transplanted HIO-derived tissue was responsive to systemic signals from the host mouse following ileocecal resection, suggesting a role for circulating factors in the intestinal adaptive response. This model of the human small intestine may pave the way for studies of intestinal physiology, disease and translational studies.


Subject(s)
Intestine, Small/physiology , Models, Biological , Pluripotent Stem Cells/cytology , Adult , Animals , Cecum/surgery , Cell Line , Humans , Ileum/surgery , In Vitro Techniques , Intestine, Small/transplantation , Mice, Inbred NOD , Mice, SCID , Organoids/cytology
16.
Stem Cells ; 29(3): 430-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21425406

ABSTRACT

Ets2 has both tumor repressive and supportive functions for different types of cancer. We have investigated the role of Ets2 within intestinal epithelial cells in postnatal mouse colon development and tumorigenesis. Conditional inactivation of Ets2 within intestinal epithelial cells results in over representation of Ets2-deficient colon crypts within young and adult animals. This preferential representation is associated with an increased number of proliferative cells within the stem cell region and an increased rate of crypt fission in young mice that result in larger patches of Ets2-deficient crypts. These effects are consistent with a selective advantage of Ets2-deficient intestinal stem cells in colonizing colonic crypts and driving crypt fission. Ets2-deficient colon crypts have an increased mucosal thickness, an increased number of goblet cells, and an increased density. Mice with Ets2-deficient intestinal cells develop more colon tumors in response to treatment with azoxymethane and dextran sulfate sodium. The selective population of colon crypts, the altered differentiation state and increased sensitivity to carcinogen-induced tumors all indicate that Ets2 deficiency alters colon stem cell number or behavior. Ets2-dependent, epithelial cell-autonomous repression of intestinal tumors may contribute to protection from colon cancer of persons with increased dosage of chromosome 21.


Subject(s)
Adenoma/genetics , Adult Stem Cells/pathology , Cell Transformation, Neoplastic/genetics , Colon/cytology , Colonic Neoplasms/genetics , Proto-Oncogene Protein c-ets-2/physiology , Adenoma/pathology , Adult Stem Cells/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Down-Regulation/genetics , Down-Regulation/physiology , Female , Genetic Predisposition to Disease , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism
18.
Cancer Res ; 68(19): 7882-6, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829544

ABSTRACT

CD133 (Prominin1) is a pentaspan transmembrane glycoprotein expressed in several stem cell populations and cancers. Reactivity with an antibody (AC133) to a glycoslyated form of CD133 has been widely used for the enrichment of cells with tumor-initiating activity in xenograph transplantation assays. We have found by fluorescence-activated cell sorting that increased AC133 reactivity in human embryonic stem cells, colon cancer, and melanoma cells is correlated with increased DNA content and, reciprocally, that the least reactive cells are in the G(1)-G(0) portion of the cell cycle. Continued cultivation of cells sorted on the basis of high and low AC133 reactivity results in a normalization of the cell reactivity profiles, indicating that cells with low AC133 reactivity can generate highly reactive cells as they resume proliferation. The association of AC133 with actively cycling cells may contribute to the basis for enrichment for tumor-initiating activity.


Subject(s)
Antigens, CD/metabolism , Cell Cycle/immunology , Colonic Neoplasms/metabolism , Embryonic Stem Cells/metabolism , Glycoproteins/metabolism , Melanoma/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Antigens, CD/immunology , Caco-2 Cells , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Cell Separation/methods , Coculture Techniques , Colonic Neoplasms/pathology , DNA/analysis , Epitopes/metabolism , Gene Expression Profiling , Glycoproteins/immunology , Humans , Immunohistochemistry , Melanoma/pathology , Mice , Peptides/immunology
19.
Acta neurol. colomb ; 24(2): 81-90, abr.-jun. 2008. tab
Article in Spanish | LILACS | ID: lil-638363

ABSTRACT

Debido a la falta de una prueba reina, el diagnóstico de la Esclerosis Múltiple continúa basándose en la clínica y ayudado por laboratorio e imágenes de resonancia. Han existido varios criterios para precisar el diagnóstico, la mayoría de los cuales comparten la demostración de diseminación en tiempo y en espacio y no encontrar una mejor explicación de la clínica. Se hace un recuento histórico de los criterios desde Allison y Millard en 1954 hasta la última revisión de los criterios de McDonald realizada por un panel internacional presidido por Polman en Amsterdam en 2005. De acuerdo con éste último, nuevas lesiones en T2 al menos un mes después del comienzo, o detección de gadolíneo al menos 3 meses después y en un sitio diferente al que corresponde el evento inicial, son suficientes para demostrar diseminación en tiempo, permitiendo más rapidez y flexibilidad en el diagnóstico. Una lesión espinal puede ser equivalente a una lesión infratentorial para demostrar diseminación en espacio y la forma primaria progresiva puede diagnosticarse en ausencia de hallazgos en el LCR, si se presentan anormalidades típicas en la resonancia cerebral. En el futuro es probable que se refine el diagnóstico con mejores imágenes y con marcadores biológicos, inmunológicos o genéticos, pero siempre prevalecerá el sano criterio médico.


Subject(s)
Humans , Multiple Sclerosis , Biomarkers , Neurology
20.
Dev Biol ; 312(1): 284-99, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17977525

ABSTRACT

The Ets2 transcription factor is essential for the development of the mouse placenta and for generating signals for embryonic mesoderm and axis formation. Using a conditional targeted Ets2 allele, we show that Ets2 is essential for trophoblast stem (TS) cells self-renewal. Inactivation of Ets2 results in TS cell slower growth, increased expression of a subset of differentiation-associated genes and decreased expression of several genes implicated in TS self-renewal. Among the direct TS targets of Ets2 is Cdx2, a key master regulator of TS cell state. Thus Ets2 contributes to the regulation of multiple genes important for maintaining the undifferentiated state of TS cells and as candidate signals for embryonic development.


Subject(s)
Proto-Oncogene Protein c-ets-2/metabolism , Stem Cells/cytology , Trophoblasts/cytology , Alleles , Animals , CDX2 Transcription Factor , Cell Differentiation , Cell Line , Cell Proliferation , Colon/metabolism , Embryo Loss , Gene Expression Regulation, Developmental , Gene Targeting , Homeodomain Proteins/genetics , Humans , Integrases/metabolism , Mice , Mice, Mutant Strains , Stem Cells/metabolism , Transcription Factors/genetics , Transcription, Genetic , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...