Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.698
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 182(6): 1606-1622.e23, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32888429

RESUMEN

The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.


Asunto(s)
Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/metabolismo , Cuerpos de Nissl/metabolismo , ARN Mensajero/metabolismo , Análisis de la Célula Individual/métodos , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Relojes Circadianos/genética , Colon/citología , Colon/metabolismo , Retículo Endoplásmico Rugoso/genética , Retículo Endoplásmico Rugoso/metabolismo , Retículo Endoplásmico Rugoso/ultraestructura , Células Epiteliales/metabolismo , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Íleon/citología , Íleon/metabolismo , Inflamación/genética , Inflamación/metabolismo , Enfermedades Intestinales/genética , Enfermedades Intestinales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Cuerpos de Nissl/genética , Cuerpos de Nissl/ultraestructura , ARN Mensajero/genética , RNA-Seq , Ribosomas/metabolismo , Ribosomas/ultraestructura , Células del Estroma/metabolismo
2.
Nat Immunol ; 22(6): 699-710, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34040226

RESUMEN

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Asunto(s)
Colitis/inmunología , Mucosa Intestinal/inmunología , Listeriosis/inmunología , Macrófagos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Proliferación Celular/genética , Colitis/microbiología , Colitis/patología , Colon/citología , Colon/embriología , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Toxina Diftérica/administración & dosificación , Toxina Diftérica/inmunología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Microbioma Gastrointestinal/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Vida Libre de Gérmenes , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Mucosa Intestinal/patología , Listeriosis/microbiología , Listeriosis/patología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , RNA-Seq , Transducción de Señal/genética , Transducción de Señal/inmunología
3.
Nat Immunol ; 21(3): 343-353, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066951

RESUMEN

Gastrointestinal microbiota and immune cells interact closely and display regional specificity; however, little is known about how these communities differ with location. Here, we simultaneously assess microbiota and single immune cells across the healthy, adult human colon, with paired characterization of immune cells in the mesenteric lymph nodes, to delineate colonic immune niches at steady state. We describe distinct helper T cell activation and migration profiles along the colon and characterize the transcriptional adaptation trajectory of regulatory T cells between lymphoid tissue and colon. Finally, we show increasing B cell accumulation, clonal expansion and mutational frequency from the cecum to the sigmoid colon and link this to the increasing number of reactive bacterial species.


Asunto(s)
Colon/inmunología , Colon/microbiología , Microbioma Gastrointestinal/inmunología , Adulto , Linfocitos B/inmunología , Colon/citología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Especificidad de Órganos , RNA-Seq , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Transcriptoma
4.
Cell ; 165(7): 1708-1720, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27264604

RESUMEN

In the mammalian intestine, crypts of Leiberkühn house intestinal epithelial stem/progenitor cells at their base. The mammalian intestine also harbors a diverse array of microbial metabolite compounds that potentially modulate stem/progenitor cell activity. Unbiased screening identified butyrate, a prominent bacterial metabolite, as a potent inhibitor of intestinal stem/progenitor proliferation at physiologic concentrations. During homeostasis, differentiated colonocytes metabolized butyrate likely preventing it from reaching proliferating epithelial stem/progenitor cells within the crypt. Exposure of stem/progenitor cells in vivo to butyrate through either mucosal injury or application to a naturally crypt-less host organism led to inhibition of proliferation and delayed wound repair. The mechanism of butyrate action depended on the transcription factor Foxo3. Our findings indicate that mammalian crypt architecture protects stem/progenitor cell proliferation in part through a metabolic barrier formed by differentiated colonocytes that consume butyrate and stimulate future studies on the interplay of host anatomy and microbiome metabolism.


Asunto(s)
Bacterias/metabolismo , Butiratos/metabolismo , Colon/citología , Colon/microbiología , Microbioma Gastrointestinal , Células Madre/metabolismo , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Animales , Proliferación Celular , Intestino Delgado/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oxidación-Reducción , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Células Madre/citología , Pez Cebra
5.
Nature ; 629(8012): 669-678, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600382

RESUMEN

Interleukin 22 (IL-22) has a non-redundant role in immune defence of the intestinal barrier1-3. T cells, but not innate lymphoid cells, have an indispensable role in sustaining the IL-22 signalling that is required for the protection of colonic crypts against invasion during infection by the enteropathogen Citrobacter rodentium4 (Cr). However, the intestinal epithelial cell (IEC) subsets targeted by T cell-derived IL-22, and how T cell-derived IL-22 sustains activation in IECs, remain undefined. Here we identify a subset of absorptive IECs in the mid-distal colon that are specifically targeted by Cr and are differentially responsive to IL-22 signalling. Major histocompatibility complex class II (MHCII) expression by these colonocytes was required to elicit sustained IL-22 signalling from Cr-specific T cells, which was required to restrain Cr invasion. Our findings explain the basis for the regionalization of the host response to Cr and demonstrate that epithelial cells must elicit MHCII-dependent help from IL-22-producing T cells to orchestrate immune protection in the intestine.


Asunto(s)
Citrobacter rodentium , Colon , Células Epiteliales , Mucosa Intestinal , Linfocitos T , Animales , Femenino , Masculino , Ratones , Citrobacter rodentium/inmunología , Colon/citología , Colon/inmunología , Colon/microbiología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-22/inmunología , Interleucina-22/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/citología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Nature ; 623(7989): 1044-1052, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37993709

RESUMEN

All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1ß production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.


Asunto(s)
Presentación de Antígeno , Colon , Células Epiteliales , Interferón gamma , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Colon/citología , Colon/inmunología , Colon/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
7.
Nat Immunol ; 17(4): 441-50, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26855029

RESUMEN

Epithelial tissues continually undergo apoptosis. Commensal organisms that inhabit the epithelium influence tissue homeostasis, in which regulatory T cells (Treg cells) have a central role. However, the physiological importance of epithelial cell apoptosis and how the number of Treg cells is regulated are both incompletely understood. Here we found that apoptotic epithelial cells negatively regulated the commensal-stimulated proliferation of Treg cells. Gut commensals stimulated CX3CR1(+)CD103(-)CD11b(+) dendritic cells (DCs) to produce interferon-ß (IFN-ß), which augmented the proliferation of Treg cells in the intestine. Conversely, phosphatidylserine exposed on apoptotic epithelial cells suppressed IFN-ß production by the DCs via inhibitory signaling mediated by the cell-surface glycoprotein CD300a and thus suppressed Treg cell proliferation. Our findings reveal a regulatory role for apoptotic epithelial cells in maintaining the number of Treg cell and tissue homeostasis.


Asunto(s)
Apoptosis/inmunología , Epidermis/inmunología , Células Epiteliales/inmunología , Microbioma Gastrointestinal/inmunología , Interferón beta/inmunología , Mucosa Intestinal/inmunología , Mucosa Respiratoria/inmunología , Linfocitos T Reguladores/inmunología , Alérgenos/toxicidad , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colon/citología , Colon/inmunología , Células Dendríticas/inmunología , Dermatitis Alérgica por Contacto/etiología , Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/patología , Sulfato de Dextran/toxicidad , Células Epidérmicas , Citometría de Flujo , Inmunohistoquímica , Mucosa Intestinal/citología , Células de Langerhans/inmunología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Noqueados , Ovalbúmina/toxicidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos/genética , Mucosa Respiratoria/citología , Infecciones por Salmonella/inmunología , Salmonella typhimurium
8.
Cell ; 149(1): 146-58, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22464327

RESUMEN

Lineage mapping has identified both proliferative and quiescent intestinal stem cells, but the molecular circuitry controlling stem cell quiescence is incompletely understood. By lineage mapping, we show Lrig1, a pan-ErbB inhibitor, marks predominately noncycling, long-lived stem cells that are located at the crypt base and that, upon injury, proliferate and divide to replenish damaged crypts. Transcriptome profiling of Lrig1(+) colonic stem cells differs markedly from the profiling of highly proliferative, Lgr5(+) colonic stem cells; genes upregulated in the Lrig1(+) population include those involved in cell cycle repression and response to oxidative damage. Loss of Apc in Lrig1(+) cells leads to intestinal adenomas, and genetic ablation of Lrig1 results in heightened ErbB1-3 expression and duodenal adenomas. These results shed light on the relationship between proliferative and quiescent intestinal stem cells and support a model in which intestinal stem cell quiescence is maintained by calibrated ErbB signaling with loss of a negative regulator predisposing to neoplasia.


Asunto(s)
Colon/metabolismo , Genes Supresores de Tumor , Intestino Delgado/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Colon/citología , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Humanos , Neoplasias Intestinales/patología , Intestino Delgado/citología , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
9.
Nature ; 592(7852): 99-104, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33627870

RESUMEN

The small intestine is the main organ for nutrient absorption, and its extensive resection leads to malabsorption and wasting conditions referred to as short bowel syndrome (SBS). Organoid technology enables an efficient expansion of intestinal epithelium tissue in vitro1, but reconstruction of the whole small intestine, including the complex lymphovascular system, has remained challenging2. Here we generate a functional small intestinalized colon (SIC) by replacing the native colonic epithelium with ileum-derived organoids. We first find that xenotransplanted human ileum organoids maintain their regional identity and form nascent villus structures in the mouse colon. In vitro culture of an organoid monolayer further reveals an essential role for luminal mechanistic flow in the formation of villi. We then develop a rat SIC model by repositioning the SIC at the ileocaecal junction, where the epithelium is exposed to a constant luminal stream of intestinal juice. This anatomical relocation provides the SIC with organ structures of the small intestine, including intact vasculature and innervation, villous structures, and the lacteal (a fat-absorbing lymphatic structure specific to the small intestine). The SIC has absorptive functions and markedly ameliorates intestinal failure in a rat model of SBS, whereas transplantation of colon organoids instead of ileum organoids invariably leads to mortality. These data provide a proof of principle for the use of intestinal organoids for regenerative purposes, and offer a feasible strategy for SBS treatment.


Asunto(s)
Colon/citología , Íleon/trasplante , Mucosa Intestinal/citología , Organoides/trasplante , Regeneración , Medicina Regenerativa/métodos , Síndrome del Intestino Corto/terapia , Animales , Colon/irrigación sanguínea , Colon/inervación , Colon/cirugía , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Íleon/citología , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/inervación , Mucosa Intestinal/cirugía , Masculino , Técnicas de Cultivo de Órganos , Organoides/citología , Ratas , Ratas Endogámicas Lew , Síndrome del Intestino Corto/patología , Síndrome del Intestino Corto/cirugía
10.
Nature ; 593(7859): 405-410, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33911282

RESUMEN

Somatic mutations drive the development of cancer and may contribute to ageing and other diseases1,2. Despite their importance, the difficulty of detecting mutations that are only present in single cells or small clones has limited our knowledge of somatic mutagenesis to a minority of tissues. Here, to overcome these limitations, we developed nanorate sequencing (NanoSeq), a duplex sequencing protocol with error rates of less than five errors per billion base pairs in single DNA molecules from cell populations. This rate is two orders of magnitude lower than typical somatic mutation loads, enabling the study of somatic mutations in any tissue independently of clonality. We used this single-molecule sensitivity to study somatic mutations in non-dividing cells across several tissues, comparing stem cells to differentiated cells and studying mutagenesis in the absence of cell division. Differentiated cells in blood and colon displayed remarkably similar mutation loads and signatures to their corresponding stem cells, despite mature blood cells having undergone considerably more divisions. We then characterized the mutational landscape of post-mitotic neurons and polyclonal smooth muscle, confirming that neurons accumulate somatic mutations at a constant rate throughout life without cell division, with similar rates to mitotically active tissues. Together, our results suggest that mutational processes that are independent of cell division are important contributors to somatic mutagenesis. We anticipate that the ability to reliably detect mutations in single DNA molecules could transform our understanding of somatic mutagenesis and enable non-invasive studies on large-scale cohorts.


Asunto(s)
Células Sanguíneas/metabolismo , Diferenciación Celular/genética , Análisis Mutacional de ADN/métodos , Músculo Liso/metabolismo , Mutación , Neuronas/metabolismo , Imagen Individual de Molécula/métodos , Células Madre/metabolismo , Enfermedad de Alzheimer/genética , Células Sanguíneas/citología , División Celular , Estudios de Cohortes , Colon/citología , Epitelio/metabolismo , Granulocitos/citología , Granulocitos/metabolismo , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso/citología , Mutagénesis , Tasa de Mutación , Neuronas/citología , Células Madre/citología
11.
Nature ; 589(7841): 270-275, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33116299

RESUMEN

There is an urgent need to create novel models using human disease-relevant cells to study severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) biology and to facilitate drug screening. Here, as SARS-CoV-2 primarily infects the respiratory tract, we developed a lung organoid model using human pluripotent stem cells (hPSC-LOs). The hPSC-LOs (particularly alveolar type-II-like cells) are permissive to SARS-CoV-2 infection, and showed robust induction of chemokines following SARS-CoV-2 infection, similar to what is seen in patients with COVID-19. Nearly 25% of these patients also have gastrointestinal manifestations, which are associated with worse COVID-19 outcomes1. We therefore also generated complementary hPSC-derived colonic organoids (hPSC-COs) to explore the response of colonic cells to SARS-CoV-2 infection. We found that multiple colonic cell types, especially enterocytes, express ACE2 and are permissive to SARS-CoV-2 infection. Using hPSC-LOs, we performed a high-throughput screen of drugs approved by the FDA (US Food and Drug Administration) and identified entry inhibitors of SARS-CoV-2, including imatinib, mycophenolic acid and quinacrine dihydrochloride. Treatment at physiologically relevant levels of these drugs significantly inhibited SARS-CoV-2 infection of both hPSC-LOs and hPSC-COs. Together, these data demonstrate that hPSC-LOs and hPSC-COs infected by SARS-CoV-2 can serve as disease models to study SARS-CoV-2 infection and provide a valuable resource for drug screening to identify candidate COVID-19 therapeutics.


Asunto(s)
Antivirales/farmacología , COVID-19/virología , Colon/citología , Evaluación Preclínica de Medicamentos/métodos , Pulmón/citología , Organoides/efectos de los fármacos , Organoides/virología , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/prevención & control , Colon/efectos de los fármacos , Colon/virología , Aprobación de Drogas , Femenino , Xenoinjertos/efectos de los fármacos , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/virología , Masculino , Ratones , Organoides/citología , Organoides/metabolismo , SARS-CoV-2/genética , Estados Unidos , United States Food and Drug Administration , Tropismo Viral , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
12.
Development ; 149(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34910127

RESUMEN

Although Wnt signaling is clearly important for the intestinal epithelial homeostasis, the relevance of various sources of Wnt ligands themselves remains incompletely understood. Blocking the release of Wnt in distinct stromal cell types suggests obligatory functions of several stromal cell sources and yields different observations. The physiological contribution of epithelial Wnt to tissue homeostasis remains unclear. We show here that blocking epithelial Wnts affects colonic Reg4+ epithelial cell differentiation and impairs colonic epithelial regeneration after injury in mice. Single-cell RNA analysis of intestinal stroma showed that the majority of Wnt-producing cells were contained in transgelin (Tagln+) and smooth muscle actin α2 (Acta2+) expressing populations. We genetically attenuated Wnt production from these stromal cells using Tagln-Cre and Acta2-CreER drivers, and found that blockage of Wnt release from either epithelium or Tagln+ and Acta2+ stromal cells impaired colonic epithelial healing after chemical-induced injury. Aggregated blockage of Wnt release from both epithelium and Tagln+ or Acta2+ stromal cells drastically diminished epithelial repair, increasing morbidity and mortality. These results from two uncharacterized stromal populations suggested that colonic recovery from colitis-like injury depends on multiple Wnt-producing sources.


Asunto(s)
Actinas/metabolismo , Colitis Ulcerosa/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Proteína Wnt3A/metabolismo , Cicatrización de Heridas , Actinas/genética , Animales , Células Cultivadas , Colon/citología , Colon/metabolismo , Colon/fisiología , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Proteína Wnt3A/genética
13.
PLoS Biol ; 20(1): e3001527, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35089911

RESUMEN

CRISPR-associated nucleases are powerful tools for precise genome editing of model systems, including human organoids. Current methods describing fluorescent gene tagging in organoids rely on the generation of DNA double-strand breaks (DSBs) to stimulate homology-directed repair (HDR) or non-homologous end joining (NHEJ)-mediated integration of the desired knock-in. A major downside associated with DSB-mediated genome editing is the required clonal selection and expansion of candidate organoids to verify the genomic integrity of the targeted locus and to confirm the absence of off-target indels. By contrast, concurrent nicking of the genomic locus and targeting vector, known as in-trans paired nicking (ITPN), stimulates efficient HDR-mediated genome editing to generate large knock-ins without introducing DSBs. Here, we show that ITPN allows for fast, highly efficient, and indel-free fluorescent gene tagging in human normal and cancer organoids. Highlighting the ease and efficiency of ITPN, we generate triple fluorescent knock-in organoids where 3 genomic loci were simultaneously modified in a single round of targeting. In addition, we generated model systems with allele-specific readouts by differentially modifying maternal and paternal alleles in one step. ITPN using our palette of targeting vectors, publicly available from Addgene, is ideally suited for generating error-free heterozygous knock-ins in human organoids.


Asunto(s)
ADN/genética , Desoxirribonucleasa I/metabolismo , Sitios Genéticos , Organoides/metabolismo , Reparación del ADN por Recombinación , Coloración y Etiquetado/métodos , Alelos , Secuencia de Bases , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Colon/citología , Colon/metabolismo , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Desoxirribonucleasa I/genética , Electroporación/métodos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Técnicas de Sustitución del Gen , Vectores Genéticos , Genoma Humano , Heterocigoto , Humanos , Organoides/citología
14.
Nature ; 574(7779): 532-537, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645730

RESUMEN

The colorectal adenoma-carcinoma sequence has provided a paradigmatic framework for understanding the successive somatic genetic changes and consequent clonal expansions that lead to cancer1. However, our understanding of the earliest phases of colorectal neoplastic changes-which may occur in morphologically normal tissue-is comparatively limited, as for most cancer types. Here we use whole-genome sequencing to analyse hundreds of normal crypts from 42 individuals. Signatures of multiple mutational processes were revealed; some of these were ubiquitous and continuous, whereas others were only found in some individuals, in some crypts or during certain periods of life. Probable driver mutations were present in around 1% of normal colorectal crypts in middle-aged individuals, indicating that adenomas and carcinomas are rare outcomes of a pervasive process of neoplastic change across morphologically normal colorectal epithelium. Colorectal cancers exhibit substantially increased mutational burdens relative to normal cells. Sequencing normal colorectal cells provides quantitative insights into the genomic and clonal evolution of cancer.


Asunto(s)
Colon/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Mutación , Síntomas Prodrómicos , Recto/citología , Adenoma/genética , Adenoma/patología , Anciano , Proteína Axina/genética , Carcinoma/genética , Carcinoma/patología , Transformación Celular Neoplásica , Células Clonales/citología , Células Clonales/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Femenino , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Células Madre/citología , Células Madre/metabolismo
15.
Nature ; 567(7746): 49-55, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30814735

RESUMEN

The colonic epithelium facilitates host-microorganism interactions to control mucosal immunity, coordinate nutrient recycling and form a mucus barrier. Breakdown of the epithelial barrier underpins inflammatory bowel disease (IBD). However, the specific contributions of each epithelial-cell subtype to this process are unknown. Here we profile single colonic epithelial cells from patients with IBD and unaffected controls. We identify previously unknown cellular subtypes, including gradients of progenitor cells, colonocytes and goblet cells within intestinal crypts. At the top of the crypts, we find a previously unknown absorptive cell, expressing the proton channel OTOP2 and the satiety peptide uroguanylin, that senses pH and is dysregulated in inflammation and cancer. In IBD, we observe a positional remodelling of goblet cells that coincides with downregulation of WFDC2-an antiprotease molecule that we find to be expressed by goblet cells and that inhibits bacterial growth. In vivo, WFDC2 preserves the integrity of tight junctions between epithelial cells and prevents invasion by commensal bacteria and mucosal inflammation. We delineate markers and transcriptional states, identify a colonic epithelial cell and uncover fundamental determinants of barrier breakdown in IBD.


Asunto(s)
Colon/citología , Colon/patología , Células Epiteliales/clasificación , Células Epiteliales/citología , Salud , Enfermedades Inflamatorias del Intestino/patología , Canales Iónicos/metabolismo , Animales , Biomarcadores/análisis , Colitis Ulcerosa/genética , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Colon/microbiología , Células Epiteliales/microbiología , Células Epiteliales/patología , Predisposición Genética a la Enfermedad/genética , Células Caliciformes/citología , Células Caliciformes/metabolismo , Células Caliciformes/patología , Humanos , Concentración de Iones de Hidrógeno , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Péptidos Natriuréticos/metabolismo , Proteínas/metabolismo , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo , Células Madre/patología , Uniones Estrechas/metabolismo , Transcripción Genética , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP
16.
Nature ; 566(7743): 249-253, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700914

RESUMEN

Environmental genotoxic factors pose a challenge to the genomic integrity of epithelial cells at barrier surfaces that separate host organisms from the environment. They can induce mutations that, if they occur in epithelial stem cells, contribute to malignant transformation and cancer development1-3. Genome integrity in epithelial stem cells is maintained by an evolutionarily conserved cellular response pathway, the DNA damage response (DDR). The DDR culminates in either transient cell-cycle arrest and DNA repair or elimination of damaged cells by apoptosis4,5. Here we show that the cytokine interleukin-22 (IL-22), produced by group 3 innate lymphoid cells (ILC3) and γδ T cells, is an important regulator of the DDR machinery in intestinal epithelial stem cells. Using a new mouse model that enables sporadic inactivation of the IL-22 receptor in colon epithelial stem cells, we demonstrate that IL-22 is required for effective initiation of the DDR following DNA damage. Stem cells deprived of IL-22 signals and exposed to carcinogens escaped DDR-controlled apoptosis, contained more mutations and were more likely to give rise to colon cancer. We identified metabolites of glucosinolates, a group of phytochemicals contained in cruciferous vegetables, to be a widespread source of genotoxic stress in intestinal epithelial cells. These metabolites are ligands of the aryl hydrocarbon receptor (AhR)6, and AhR-mediated signalling in ILC3 and γδ T cells controlled their production of IL-22. Mice fed with diets depleted of glucosinolates produced only very low levels of IL-22 and, consequently, the DDR in epithelial cells of mice on a glucosinolate-free diet was impaired. This work identifies a homeostatic network protecting stem cells against challenge to their genome integrity by AhR-mediated 'sensing' of genotoxic compounds from the diet. AhR signalling, in turn, ensures on-demand production of IL-22 by innate lymphocytes directly regulating components of the DDR in epithelial stem cells.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Colon/citología , Interleucinas/farmacología , Mutágenos/farmacología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Animales , Apoptosis/efectos de los fármacos , Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Neoplasias del Colon/prevención & control , Daño del ADN , Dieta/efectos adversos , Glucosinolatos/administración & dosificación , Glucosinolatos/farmacología , Inmunidad Innata , Interleucinas/biosíntesis , Mucosa Intestinal/citología , Ligandos , Ratones , Mutágenos/administración & dosificación , Mutación/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Interleucina/metabolismo , Células Madre/citología , Linfocitos T/metabolismo , Interleucina-22
17.
J Biol Chem ; 299(8): 105068, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37468102

RESUMEN

Although it was described previously for estrogen (E2) regulation of intestinal epithelial Cl- and HCO3- secretion in sex difference, almost nothing is known about the roles of estrogen receptor (ER) subtypes in regulating E2-modulated epithelial ion transports and epithelial restitution. Here, we aimed to investigate ERα and ERß subtypes in the regulation of E2-modulated colonic epithelial HCO3- and Cl- secretion and epithelial restitution. Through physiological and biochemical studies, in combination of genetic knockdown, we showed that ERα attenuated female colonic Cl- secretion but promoted Ca2+-dependent HCO3- secretion via store-operated calcium entry (SOCE) mechanism in mice. However, ERß attenuated HCO3- secretion by inhibiting Ca2+via the SOCE and inhibiting cAMP via protein kinases. Moreover, ERα but not ERß promoted epithelial cell restitution via SOCE/Ca2+ signaling. ERα also enhanced cyclin D1, proliferating cell nuclear antigen, and ß-catenin expression in normal human colonic epithelial cells. All ERα-mediated biological effects could be attenuated by its selective antagonist and genetic knockdown. Finally, both ERα and ERß were expressed in human colonic epithelial cells and mouse colonic tissues. We therefore conclude that E2 modulates complex colonic epithelial HCO3- and Cl- secretion via ER subtype-dependent mechanisms and that ERα is specifically responsible for colonic epithelial regeneration. This study provides novel insights into the molecular mechanisms of how ERα and ERß subtypes orchestrate functional homeostasis of normal colonic epithelial cells.


Asunto(s)
Colon , Células Epiteliales , Receptor alfa de Estrógeno , Transporte Iónico , Receptores de Estrógenos , Animales , Femenino , Humanos , Ratones , Células Epiteliales/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Receptores de Estrógenos/metabolismo , Colon/citología
18.
Nat Methods ; 18(7): 829-834, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34183831

RESUMEN

We introduce a cost-effective and easily implementable scan unit that converts any camera-based microscope with optical sectioning capability into a multi-angle projection imaging system. Projection imaging reduces data overhead and accelerates imaging by a factor of >100, while also allowing users to readily view biological phenomena of interest from multiple perspectives on the fly. By rapidly interrogating the sample from just two perspectives, our method also enables real-time stereoscopic imaging and three-dimensional particle localization. We demonstrate projection imaging with spinning disk confocal, lattice light-sheet, multidirectional illumination light-sheet and oblique plane microscopes on specimens that range from organelles in single cells to the vasculature of a zebrafish embryo. Furthermore, we leverage our projection method to rapidly image cancer cell morphodynamics and calcium signaling in cultured neurons at rates up to 119 Hz as well as to simultaneously image orthogonal views of a beating embryonic zebrafish heart.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Confocal/instrumentación , Microscopía Confocal/métodos , Animales , Colon/citología , Embrión no Mamífero/citología , Femenino , Corazón/diagnóstico por imagen , Corazón/embriología , Humanos , Imagenología Tridimensional , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Ratas Sprague-Dawley , Esferoides Celulares/patología , Pez Cebra/embriología
19.
EMBO Rep ; 23(3): e53246, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34939731

RESUMEN

Regulatory T lymphocyte (Treg) homing reactions mediated by G protein-coupled receptor (GPCR)-ligand interactions play a central role in maintaining intestinal immune homeostasis by restraining inappropriate immune responses in the gastrointestinal tract. However, the origin of Treg homing to the colon remains mysterious. Here, we report that the C10ORF99 peptide (also known as CPR15L and AP57), a cognate ligand of GPR15 that controls Treg homing to the colon, originates from a duplication of the flanking CDHR1 gene and is functionally paired with GPR15 in amniotes. Evolutionary analysis and experimental data indicate that the GPR15-C10ORF99 pair is functionally conserved to mediate colonic Treg homing in amniotes and their expression patterns are positively correlated with herbivore diet in the colon. With the first herbivorous diet in early amniotes, a new biological process (herbivorous diet short-chain fatty acid-C10ORF99/GPR15-induced Treg homing colon immune homeostasis) emerged, and we propose an evolutionary model whereby GPR15-C10ORF99 functional pairing has initiated the first colonic Treg homing reaction in amniotes. Our findings also highlight that GPCR-ligand pairing leads to physiological adaptation during vertebrate evolution.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Colon/citología , Proteínas de Unión al ADN , Receptores Acoplados a Proteínas G , Linfocitos T Reguladores , Animales , Colon/inmunología , Ligandos , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfocitos T Reguladores/citología
20.
Cell ; 139(4): 802-13, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19914172

RESUMEN

The kinesin superfamily proteins (KIFs) are motor proteins that transport organelles and protein complexes in a microtubule- and ATP-dependent manner. We identified KIF26A as a new member of the murine KIFs. KIF26A is a rather atypical member as it lacks ATPase activity. Mice with a homozygous deletion of Kif26a developed a megacolon with enteric nerve hyperplasia. Kif26a-/- enteric neurons showed hypersensitivity for GDNF-Ret signaling, and we find that KIF26A suppressed GDNF-Ret signaling by direct binding and inhibition of Grb2, an essential component of GDNF/Akt/ERK signaling. We therefore propose that the unconventional kinesin KIF26A plays a key role in enteric nervous system development by repressing a cell growth signaling pathway.


Asunto(s)
Sistema Nervioso Entérico/embriología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Enfermedad de Hirschsprung/metabolismo , Cinesinas/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal , Animales , Procesos de Crecimiento Celular , Línea Celular , Colon/citología , Colon/embriología , Colon/inervación , Proteína Adaptadora GRB2/metabolismo , Cinesinas/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA