Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 37(12): e23272, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37997495

RESUMEN

Parkinson's disease (PD) is a progressive, neurodegenerative disorder with an increasing incidence, unknown etiology, and is currently incurable. Advances in understanding the pathological mechanisms at a molecular level have been slow, with little attention focused on the early prodromal phase of the disease. Consequently, the development of early-acting disease-modifying therapies has been hindered. The olfactory bulb (OB), the brain region responsible for initial processing of olfactory information, is particularly affected early in PD at both functional and molecular levels but there is little information on how the cells in this region are affected by disease. Organotypic and primary OB cultures were developed and characterized. These platforms were then used to assess the effects of 3,4-dihydroxyphenylacetylaldehyde (DOPAL), a metabolite of dopamine present in increased levels in post-mortem PD tissue and which is thought to contribute to PD pathogenesis. Our findings showed that DOPAL exposure can recapitulate many aspects of PD pathology. Oxidative stress, depolarization of mitochondrial membranes, and neurodegeneration were all induced by DOPAL addition, as were measured transcriptomic changes consistent with those reported in PD clinical studies. These olfactory models of prodromal disease lend credence to the catecholaldehyde hypothesis of PD and provide insight into the mechanisms by which the OB may be involved in disease progression.


Asunto(s)
Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , Bulbo Olfatorio/metabolismo , Sistemas Microfisiológicos , Encéfalo/metabolismo , Dopamina/metabolismo
2.
J Am Soc Nephrol ; 31(4): 701-715, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32034106

RESUMEN

BACKGROUND: Gdf15 encodes a TGF-ß superfamily member that is rapidly activated in response to stress in multiple organ systems, including the kidney. However, there has been a lack of information about Gdf15 activity and effects in normal kidney and in AKI. METHODS: We used genome editing to generate a Gdf15nuGFP-CE mouse line, removing Gdf15 at the targeted allele, and enabling direct visualization and genetic modification of Gdf15-expressing cells. We extensively mapped Gdf15 expression in the normal kidney and following bilateral ischemia-reperfusion injury, and quantified and compared renal responses to ischemia-reperfusion injury in the presence and absence of GDF15. In addition, we analyzed single nucleotide polymorphism association data for GDF15 for associations with patient kidney transplant outcomes. RESULTS: Gdf15 is normally expressed within aquaporin 1-positive cells of the S3 segment of the proximal tubule, aquaporin 1-negative cells of the thin descending limb of the loop of Henle, and principal cells of the collecting system. Gdf15 is rapidly upregulated within a few hours of bilateral ischemia-reperfusion injury at these sites and new sites of proximal tubule injury. Deficiency of Gdf15 exacerbated acute tubular injury and enhanced inflammatory responses. Analysis of clinical transplantation data linked low circulating levels of GDF15 to an increased incidence of biopsy-proven acute rejection. CONCLUSIONS: Gdf15 contributes to an early acting, renoprotective injury response, modifying immune cell actions. The data support further investigation in clinical model systems of the potential benefit from GDF15 administration in situations in which some level of tubular injury is inevitable, such as following a kidney transplant.


Asunto(s)
Lesión Renal Aguda/patología , Factor 15 de Diferenciación de Crecimiento/genética , Trasplante de Riñón , Polimorfismo Genético/genética , Daño por Reperfusión/patología , Lesión Renal Aguda/genética , Adulto , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Daño por Reperfusión/genética
3.
Am J Physiol Renal Physiol ; 319(3): F423-F435, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657158

RESUMEN

Cre-lox technology has revolutionized research in renal physiology by allowing site-specific genetic recombination in individual nephron segments. The distal convoluted tubule (DCT), consisting of distinct early (DCT1) and late (DCT2) segments, plays a central role in Na+ and K+ homeostasis. The only established Cre line targeting the DCT is Pvalb-Cre, which is limited by noninducibility, activity along DCT1 only, and activity in neurons. Here, we report the characterization of the first Cre line specific to the entire DCT. CRISPR/Cas9 targeting was used to introduce a tamoxifen-inducible IRES-Cre-ERT2 cassette downstream of the coding region of the Slc12a3 gene encoding the NaCl cotransporter (NCC). The resulting Slc12a3-Cre-ERT2 mice were crossed with R26R-YFP reporter mice, which revealed minimal leakiness with 6.3% of NCC-positive cells expressing yellow fluorescent protein (YFP) in the absence of tamoxifen. After tamoxifen injection, YFP expression was observed in 91.2% of NCC-positive cells and only in NCC-positive cells, revealing high recombination efficiency and DCT specificity. Crossing to R26R-TdTomato mice revealed higher leakiness (64.5%), suggesting differential sensitivity of the floxed site. Western blot analysis revealed no differences in abundances of total NCC or the active phosphorylated form of NCC in Slc12a3-Cre-ERT2 mice of either sex compared with controls. Plasma K+ and Mg2+ concentrations and thiazide-sensitive Na+ and K+ excretion did not differ in Slc12a3-Cre-ERT2 mice compared with controls when sex matched. These data suggest genetic modification had no obvious effect on NCC function. Slc12a3-Cre-ERT2 mice are the first line generated demonstrating inducible Cre recombinase activity along the entire DCT and will be a useful tool to study DCT function.


Asunto(s)
Túbulos Renales Distales/enzimología , Recombinasas/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Animales , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Recombinasas/genética , Simportadores del Cloruro de Sodio/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Tamoxifeno/farmacología
4.
J Am Soc Nephrol ; 29(3): 825-840, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449451

RESUMEN

The nephron is the functional unit of the kidney, but the mechanism of nephron formation during human development is unclear. We conducted a detailed analysis of nephron development in humans and mice by immunolabeling, and we compared human and mouse nephron patterning to describe conserved and divergent features. We created protein localization maps that highlight the emerging patterns along the proximal-distal axis of the developing nephron and benchmark expectations for localization of functionally important transcription factors, which revealed unanticipated cellular diversity. Moreover, we identified a novel nephron subdomain marked by Wnt4 expression that we fate-mapped to the proximal mature nephron. Significant conservation was observed between human and mouse patterning. We also determined the time at which markers for mature nephron cell types first emerge-critical data for the renal organoid field. These findings have conceptual implications for the evolutionary processes driving the diversity of mammalian organ systems. Furthermore, these findings provide practical insights beyond those gained with mouse and rat models that will guide in vitro efforts to harness the developmental programs necessary to build human kidney structures.


Asunto(s)
Diferenciación Celular , Nefronas/embriología , Nefronas/metabolismo , Células Madre/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Linaje de la Célula , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt4/metabolismo
5.
J Am Soc Nephrol ; 29(3): 806-824, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449449

RESUMEN

Cellular interactions among nephron, interstitial, and collecting duct progenitors drive mammalian kidney development. In mice, Six2+ nephron progenitor cells (NPCs) and Foxd1+ interstitial progenitor cells (IPCs) form largely distinct lineage compartments at the onset of metanephric kidney development. Here, we used the method for analyzing RNA following intracellular sorting (MARIS) approach, single-cell transcriptional profiling, in situ hybridization, and immunolabeling to characterize the presumptive NPC and IPC compartments of the developing human kidney. As in mice, each progenitor population adopts a stereotypical arrangement in the human nephron-forming niche: NPCs capped outgrowing ureteric branch tips, whereas IPCs were sandwiched between the NPCs and the renal capsule. Unlike mouse NPCs, human NPCs displayed a transcriptional profile that overlapped substantially with the IPC transcriptional profile, and key IPC determinants, including FOXD1, were readily detected within SIX2+ NPCs. Comparative gene expression profiling in human and mouse Six2/SIX2+ NPCs showed broad agreement between the species but also identified species-biased expression of some genes. Notably, some human NPC-enriched genes, including DAPL1 and COL9A2, are linked to human renal disease. We further explored the cellular diversity of mesenchymal cell types in the human nephrogenic niche through single-cell transcriptional profiling. Data analysis stratified NPCs into two main subpopulations and identified a third group of differentiating cells. These findings were confirmed by section in situ hybridization with novel human NPC markers predicted through the single-cell studies. This study provides a benchmark for the mesenchymal progenitors in the human nephrogenic niche and highlights species-variability in kidney developmental programs.


Asunto(s)
Corteza Renal/embriología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Nefronas/embriología , Animales , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular , Linaje de la Célula , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Nefronas/anatomía & histología , Nefronas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
J Am Soc Nephrol ; 29(3): 785-805, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449453

RESUMEN

Human kidney function is underpinned by approximately 1,000,000 nephrons, although the number varies substantially, and low nephron number is linked to disease. Human kidney development initiates around 4 weeks of gestation and ends around 34-37 weeks of gestation. Over this period, a reiterative inductive process establishes the nephron complement. Studies have provided insightful anatomic descriptions of human kidney development, but the limited histologic views are not readily accessible to a broad audience. In this first paper in a series providing comprehensive insight into human kidney formation, we examined human kidney development in 135 anonymously donated human kidney specimens. We documented kidney development at a macroscopic and cellular level through histologic analysis, RNA in situ hybridization, immunofluorescence studies, and transcriptional profiling, contrasting human development (4-23 weeks) with mouse development at selected stages (embryonic day 15.5 and postnatal day 2). The high-resolution histologic interactive atlas of human kidney organogenesis generated can be viewed at the GUDMAP database (www.gudmap.org) together with three-dimensional reconstructions of key components of the data herein. At the anatomic level, human and mouse kidney development differ in timing, scale, and global features such as lobe formation and progenitor niche organization. The data also highlight differences in molecular and cellular features, including the expression and cellular distribution of anchor gene markers used to identify key cell types in mouse kidney studies. These data will facilitate and inform in vitro efforts to generate human kidney structures and comparative functional analyses across mammalian species.


Asunto(s)
Riñón/embriología , Riñón/metabolismo , Organogénesis , Uréter/embriología , Animales , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Edad Gestacional , Técnicas Histológicas , Humanos , Hibridación in Situ , Riñón/anatomía & histología , Ratones , Nefronas/embriología , Nefronas/metabolismo , ARN/análisis , Uréter/metabolismo
7.
Brain Inj ; 31(11): 1521-1529, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28972405

RESUMEN

PRIMARY OBJECTIVE: Rugby is one of the few contact sports that do not mandate protective headgear, possibly because studies have shown poor efficacy for protection related to concussion pathology with existing headguards. RESEARCH DESIGN: Following innovative material technology utilization to produce headgear believed to have protective capabilities, this study examined the effects of a soft-shell headgear constructed from a novel viscoelastic material, on both behaviour and serum biomarkers after high and average impact force mild traumatic brain injuries (mTBI). METHODS AND PROCEDURES: Seventy-five male Sprague Dawley rats were divided into five groups: control, average - 37G impact, with and without headgear, and high - 106G impact, with and without headgear. Rats were sacrificed at 3 or 48 hours and serum samples were analyzed for levels of TNF-α, NEF-L, and GFAP. Animals sacrificed at 48 hours also underwent testing for balance and motor coordination, and exploratory/locomotor behaviour. MAIN OUTCOMES AND RESULTS: The novel headgear offered significant protection against mTBI symptomology and biomarkers in the group that experienced an average impact force, but only moderated protection for the animals in the high impact group. CONCLUSIONS: This innovative headgear may prevent some of the negative sequel associated with concussion pathology.


Asunto(s)
Conmoción Encefálica/prevención & control , Conmoción Encefálica/fisiopatología , Modelos Animales de Enfermedad , Dispositivos de Protección de la Cabeza , Animales , Encéfalo/metabolismo , Encéfalo/patología , Conmoción Encefálica/sangre , Conducta Exploratoria/fisiología , Proteína Ácida Fibrilar de la Glía/sangre , Masculino , Proteínas de Neurofilamentos/metabolismo , Proyectos Piloto , Desempeño Psicomotor/fisiología , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
8.
J Am Soc Nephrol ; 26(1): 81-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24904087

RESUMEN

We previously described a mesenchymal stem cell (MSC)-like population within the adult mouse kidney that displays long-term colony-forming efficiency, clonogenicity, immunosuppression, and panmesodermal potential. Although phenotypically similar to bone marrow (BM)-MSCs, kidney MSC-like cells display a distinct expression profile. FACS sorting from Hoxb7/enhanced green fluorescent protein (GFP) mice identified the collecting duct as a source of kidney MSC-like cells, with these cells undergoing an epithelial-to-mesenchymal transition to form clonogenic, long-term, self-renewing MSC-like cells. Notably, after extensive passage, kidney MSC-like cells selectively integrated into the aquaporin 2-positive medullary collecting duct when microinjected into the kidneys of neonatal mice. No epithelial integration was observed after injection of BM-MSCs. Indeed, kidney MSC-like cells retained a capacity to form epithelial structures in vitro and in vivo, and conditioned media from these cells supported epithelial repair in vitro. To investigate the origin of kidney MSC-like cells, we further examined Hoxb7(+) fractions within the kidney across postnatal development, identifying a neonatal interstitial GFP(lo) (Hoxb7(lo)) population displaying an expression profile intermediate between epithelium and interstitium. Temporal analyses with Wnt4(GCE/+):R26(tdTomato/+) mice revealed evidence for the intercalation of a Wnt4-expressing interstitial population into the neonatal collecting duct, suggesting that such intercalation may represent a normal developmental mechanism giving rise to a distinct collecting duct subpopulation. These results extend previous observations of papillary stem cell activity and collecting duct plasticity and imply a role for such cells in collecting duct formation and, possibly, repair.


Asunto(s)
Células Epiteliales/citología , Túbulos Renales Colectores/citología , Riñón/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Separación Celular , Condrocitos/citología , Colágeno/metabolismo , Perros , Transición Epitelial-Mesenquimal , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Túbulos Renales/citología , Células de Riñón Canino Madin Darby , Ratones , Osteocitos/citología , Fenotipo
9.
Development ; 139(10): 1863-73, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22510988

RESUMEN

Lengthy developmental programs generate cell diversity within an organotypic framework, enabling the later physiological actions of each organ system. Cell identity, cell diversity and cell function are determined by cell type-specific transcriptional programs; consequently, transcriptional regulatory factors are useful markers of emerging cellular complexity, and their expression patterns provide insights into the regulatory mechanisms at play. We performed a comprehensive genome-scale in situ expression screen of 921 transcriptional regulators in the developing mammalian urogenital system. Focusing on the kidney, analysis of regional-specific expression patterns identified novel markers and cell types associated with development and patterning of the urinary system. Furthermore, promoter analysis of synexpressed genes predicts transcriptional control mechanisms that regulate cell differentiation. The annotated informational resource (www.gudmap.org) will facilitate functional analysis of the mammalian kidney and provides useful information for the generation of novel genetic tools to manipulate emerging cell populations.


Asunto(s)
Sistema Urogenital/metabolismo , Animales , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Hibridación in Situ , Riñón/metabolismo , Ratones
10.
Proc Natl Acad Sci U S A ; 109(31): 12592-7, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22797898

RESUMEN

Clara cells of mammalian airways have multiple functions and are morphologically heterogeneous. Although Notch signaling is essential for the development of these cells, it is unclear how Notch influences Clara cell specification and if diversity is established among Clara cell precursors. Here we identify expression of the secretoglobin Scgb3a2 and Notch activation as early events in a program of secretory cell fate determination in developing murine airways. We show that Scgb3a2 expression in vivo is Notch-dependent at early stages and ectopically induced by constitutive Notch1 activation, and also that in vitro Notch signaling together with the pan-airway transcription factor Ttf1 (Nkx2.1) synergistically regulate secretoglobin gene transcription. Furthermore, we identified a subpopulation of secretory precursors juxtaposed to presumptive neuroepithelial bodies (NEBs), distinguished by their strong Scgb3a2 and uroplakin 3a (Upk3a) signals and reduced Ccsp (Scgb1a1) expression. Genetic ablation of Ascl1 prevented NEB formation and selectively interfered with the formation of this subpopulation of cells. Lineage labeling of Upk3a-expressing cells during development showed that these cells remain largely uncommitted during embryonic development and contribute to Clara and ciliated cells in the adult lung. Together, our findings suggest a role for Notch in the induction of a Clara cell-specific program of gene expression, and reveals that the NEB microenvironment in the developing airways is a niche for a distinct subset of Clara-like precursors.


Asunto(s)
Cuerpos Neuroepiteliales/metabolismo , Sistema Respiratorio/embriología , Nicho de Células Madre/fisiología , Células Madre/metabolismo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Cuerpos Neuroepiteliales/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Sistema Respiratorio/citología , Secretoglobinas/biosíntesis , Secretoglobinas/genética , Células Madre/citología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Am J Physiol Renal Physiol ; 307(12): F1412-26, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25339699

RESUMEN

Mesenchymal stem cells (MSCs) suppress T helper (Th)17 cell differentiation and are being clinically pursued for conditions associated with aberrant Th17 responses. Whether such immunomodulatory effects are enhanced by coadministration of MSCs with other agents is not well known. In the present study, individual and combined effects of MSCs and the vitamin D receptor (VDR) agonist paricalcitol on Th17 induction were investigated in vitro and in a mouse model of sterile kidney inflammation (unilateral ureteral obstruction). In vitro, MSCs and paricalcitol additively suppressed Th17 differentiation, although only MSCs suppressed expression of Th17-associated transcriptions factors. Combined administration of MSCs and paricalcitol resulted in an early (day 3) reduction of intrarenal CD4(+) and CD8(+) T cells, CD11b(+)/lymphocyte antigen 6G(+) neutrophils, and inflammatory (lymphocyte antigen 6C(hi)) monocytes as well as reduced transcript for IL-17 compared with untreated animals. Later (day 8), obstructed kidneys of MSC/paricalcitol double-treated mice, but not mice treated with either intervention alone, had reduced tubular injury and interstitial fibrosis as well as lower numbers of neutrophils and inflammatory monocytes and an increase in the ratio between M2 (CD206(+)) and M1 (CD206(-)) macrophages compared with control mice. Adjunctive therapy with VDR agonists may enhance the immunosuppressive properties of MSCs in the setting of pathogenic Th17-type immune responses and related inflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Ergocalciferoles/farmacología , Inmunosupresores/farmacología , Riñón/efectos de los fármacos , Trasplante de Células Madre Mesenquimatosas , Nefritis/prevención & control , Receptores de Calcitriol/agonistas , Células Th17/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Interleucina-17/genética , Interleucina-17/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Nefritis/etiología , Nefritis/inmunología , Nefritis/metabolismo , Nefritis/patología , Infiltración Neutrófila/efectos de los fármacos , Receptores de Calcitriol/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Factores de Tiempo , Obstrucción Ureteral/complicaciones
12.
Biol Reprod ; 86(2): 37, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21976597

RESUMEN

The embryonic origins of ovarian granulosa cells have been a subject of debate for decades. By tamoxifen-induced lineage tracing of Foxl2-expressing cells, we show that descendants of the bipotential supporting cell precursors in the early gonad contribute granulosa cells to a specific population of follicles in the medulla of the ovary that begin to grow immediately after birth. These precursor cells arise from the proliferative ovarian surface epithelium and enter mitotic arrest prior to upregulating Foxl2. Granulosa cells that populate the cortical primordial follicles activated in adult life derive from the surface epithelium perinatally, and enter mitotic arrest at that stage. Ingression from the surface epithelium dropped to undetectable levels by Postnatal Day 7, when most surviving oocytes were individually encapsulated by granulosa cells. These findings add complexity to the standard model of sex determination in which the Sertoli and granulosa cells of the adult testis and ovary directly stem from the supporting cell precursors of the bipotential gonad.


Asunto(s)
Linaje de la Célula , Células de la Granulosa/citología , Folículo Ovárico/citología , Ovario/embriología , Animales , Diferenciación Celular , Desarrollo Embrionario , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Células de la Granulosa/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Folículo Ovárico/metabolismo , Ovario/citología
13.
Cell Stem Cell ; 29(7): 1083-1101.e7, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35803227

RESUMEN

Human pluripotent stem-cell-derived organoids are models for human development and disease. We report a modified human kidney organoid system that generates thousands of similar organoids, each consisting of 1-2 nephron-like structures. Single-cell transcriptomic profiling and immunofluorescence validation highlighted patterned nephron-like structures utilizing similar pathways, with distinct morphogenesis, to human nephrogenesis. To examine this platform for therapeutic screening, the polycystic kidney disease genes PKD1 and PKD2 were inactivated by gene editing. PKD1 and PKD2 mutant models exhibited efficient and reproducible cyst formation. Cystic outgrowths could be propagated for months to centimeter-sized cysts. To shed new light on cystogenesis, 247 protein kinase inhibitors (PKIs) were screened in a live imaging assay identifying compounds blocking cyst formation but not overall organoid growth. Scaling and further development of the organoid platform will enable a broader capability for kidney disease modeling and high-throughput drug screens.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Quistes/metabolismo , Descubrimiento de Drogas , Humanos , Riñón/metabolismo , Organoides/metabolismo , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
14.
Open Biol ; 12(8): 220149, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35946312

RESUMEN

Organ functions are highly specialized and interdependent. Secreted factors regulate organ development and mediate homeostasis through serum trafficking and inter-organ communication. Enzyme-catalysed proximity labelling enables the identification of proteins within a specific cellular compartment. Here, we report a BirA*G3 mouse strain that enables CRE-dependent promiscuous biotinylation of proteins trafficking through the endoplasmic reticulum. When broadly activated throughout the mouse, widespread labelling of proteins was observed within the secretory pathway. Streptavidin affinity purification and peptide mapping by quantitative mass spectrometry (MS) proteomics revealed organ-specific secretory profiles and serum trafficking. As expected, secretory proteomes were highly enriched for signal peptide-containing proteins, highlighting both conventional and non-conventional secretory processes, and ectodomain shedding. Lower-abundance proteins with hormone-like properties were recovered and validated using orthogonal approaches. Hepatocyte-specific activation of BirA*G3 highlighted liver-specific biotinylated secretome profiles. The BirA*G3 mouse model demonstrates enhanced labelling efficiency and tissue specificity over viral transduction approaches and will facilitate a deeper understanding of secretory protein interplay in development, and in healthy and diseased adult states.


Asunto(s)
Modelos Genéticos , Secretoma , Animales , Biotinilación , Mamíferos , Espectrometría de Masas/métodos , Ratones , Proteómica/métodos
15.
Mult Scler ; 17(7): 808-18, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21382862

RESUMEN

BACKGROUND: Increasing evidence associates the endoplasmic reticulum (ER) stress signalling pathway as a potential treatment target in multiple sclerosis (MS). OBJECTIVE: To establish the expression profile of markers of ER stress both in demyelinating biopsy specimens and microdissected lesions in human post-mortem MS tissue. METHODS: Immunohistochemical detection of C/EBP homologous protein (CHOP), immunoglobulin heavy chain binding protein (BiP), and hypoxia marker antigen D-110 in biopsies from three patients with MS primary or secondary progressive, three patients with clinically isolated syndrome, and one patient with lesional epilepsy was carried out. Laser capture microdissection of normal, perilesion and lesion tissue from post-mortem MS tissue and non-diseased control tissue was performed, followed by real-time PCR to detect ER stress genes. RESULTS: In biopsy specimens, increased expression of the ER and hypoxic stress molecules in a range of cell types in most of the actively demyelinating lesions and perilesions was detected. Real-time PCR analysis demonstrated statistically significant elevated expression of the ER stress genes in normal-appearing white matter relative to control white matter. Moreover, significantly increased expression of CHOP was detected in the perilesion of active plaques (p < 0.01). CONCLUSIONS: Our results, showing detection of elevated expression of ER stress molecules in lesional tissue, offer compelling evidence for further investigation of the ER stress signalling pathway as a potential therapeutic target for the treatment of MS.


Asunto(s)
Química Encefálica , Enfermedades Desmielinizantes/metabolismo , Retículo Endoplásmico/química , Epilepsia/metabolismo , Esclerosis Múltiple Crónica Progresiva/metabolismo , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Adulto , Anciano , Autopsia , Biopsia , Hipoxia de la Célula , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Chaperón BiP del Retículo Endoplásmico , Epilepsia/genética , Epilepsia/patología , Femenino , Regulación de la Expresión Génica , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/genética , Humanos , Inmunohistoquímica , Masculino , Microdisección , Persona de Mediana Edad , Esclerosis Múltiple Crónica Progresiva/genética , Esclerosis Múltiple Crónica Progresiva/patología , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/patología , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Estrés Fisiológico , Factor de Transcripción CHOP/análisis , Factor de Transcripción CHOP/genética , Adulto Joven
16.
Nat Commun ; 12(1): 3641, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131121

RESUMEN

Current kidney organoids model development and diseases of the nephron but not the contiguous epithelial network of the kidney's collecting duct (CD) system. Here, we report the generation of an expandable, 3D branching ureteric bud (UB) organoid culture model that can be derived from primary UB progenitors from mouse and human fetal kidneys, or generated de novo from human pluripotent stem cells. In chemically-defined culture conditions, UB organoids generate CD organoids, with differentiated principal and intercalated cells adopting spatial assemblies reflective of the adult kidney's collecting system. Aggregating 3D-cultured nephron progenitor cells with UB organoids in vitro results in a reiterative process of branching morphogenesis and nephron induction, similar to kidney development. Applying an efficient gene editing strategy to remove RET activity, we demonstrate genetically modified UB organoids can model congenital anomalies of kidney and urinary tract. Taken together, these platforms will facilitate an enhanced understanding of development, regeneration and diseases of the mammalian collecting duct system.


Asunto(s)
Túbulos Renales Colectores/citología , Riñón/citología , Riñón/crecimiento & desarrollo , Organogénesis/fisiología , Organoides/citología , Organoides/crecimiento & desarrollo , Uréter , Sistema Urinario/citología , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Riñón/embriología , Túbulos Renales Colectores/embriología , Masculino , Ratones , Morfogénesis , Nefronas , Organogénesis/genética , Organoides/embriología , Células Madre Pluripotentes/citología , Sistema Urinario/embriología , Sistema Urinario/crecimiento & desarrollo
17.
Dev Cell ; 56(16): 2381-2398.e6, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34428401

RESUMEN

Congenital abnormalities of the kidney and urinary tract are among the most common birth defects, affecting 3% of newborns. The human kidney forms around a million nephrons from a pool of nephron progenitors over a 30-week period of development. To establish a framework for human nephrogenesis, we spatially resolved a stereotypical process by which equipotent nephron progenitors generate a nephron anlage, then applied data-driven approaches to construct three-dimensional protein maps on anatomical models of the nephrogenic program. Single-cell RNA sequencing identified progenitor states, which were spatially mapped to the nephron anatomy, enabling the generation of functional gene networks predicting interactions within and between nephron cell types. Network mining identified known developmental disease genes and predicted targets of interest. The spatially resolved nephrogenic program made available through the Human Nephrogenesis Atlas (https://sckidney.flatironinstitute.org/) will facilitate an understanding of kidney development and disease and enhance efforts to generate new kidney structures.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Nefronas/metabolismo , Transcriptoma , Animales , Humanos , Ratones , Nefronas/citología , Nefronas/embriología , Proteoma/genética , Proteoma/metabolismo , RNA-Seq , Análisis de la Célula Individual
18.
Nat Commun ; 12(1): 2382, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888706

RESUMEN

Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteómica/métodos , Proteínas Represoras/metabolismo , Coloración y Etiquetado/métodos , Animales , Animales Modificados Genéticamente , Biotina/metabolismo , Biotinilación , Ligasas de Carbono-Nitrógeno/genética , Línea Celular , Modelos Animales de Enfermedad , Drosophila , Células Madre Embrionarias , Proteínas de Escherichia coli/genética , Femenino , Humanos , Masculino , Ratones , Ingeniería de Proteínas , Transporte de Proteínas , Proteínas Represoras/genética , Espectrometría de Masas en Tándem/métodos , Teratoma/diagnóstico , Teratoma/patología
19.
Acta Neuropathol ; 119(5): 601-15, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19967542

RESUMEN

The blood brain barrier (BBB) is formed by capillary endothelial cells with inter-endothelial cell tight junctions and other cells such as pericytes and astrocytes present. Previous studies have shown a role for tight junction abnormalities in BBB leakage in multiple sclerosis (MS) brain. This marks a key stage in the development of inflammatory demyelination in MS. The aim of this study was to identify aberrantly expressed genes involved in BBB changes in MS lesions. A focused endothelial cell biology microarray, capable of detecting changes in expression of 113 endothelial cell-specific genes, was employed to analyse endothelial cell mRNA extracted from post-mortem control white matter, MS normal appearing white matter (NAWM), chronic active or inactive lesions by laser capture microdissection. Microarray analysis found 52 genes out of 113 analysed, predominantly in the activation functional group, to be differentially expressed in lesions compared to control or NAWM (p < 0.01). The majority of the differentially expressed genes were validated by quantitative real time PCR. In addition, the protein expression profiles of ICAM2, MMP2, and VEGFR1 were examined by immunofluorescent staining of selected tissue blocks. ICAM-2 was expressed at a higher level in chronic inactive lesions than control or NAWM, corresponding with the increased mRNA measured by microarray and real time PCR. The data shown, presenting a number of differentially expressed genes in the microvascular compartment of MS lesions, may shed light on the molecular mechanisms that are involved in the breakdown of the BBB. This moves us a step closer to the identification of potential therapeutic targets for repair of the compromised BBB.


Asunto(s)
Vasos Sanguíneos/metabolismo , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Esclerosis Múltiple/genética , Adulto , Anciano , Anciano de 80 o más Años , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Circulación Cerebrovascular/genética , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Microdisección/métodos , Microscopía Confocal , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/genética , Uniones Estrechas/metabolismo
20.
Dev Biol ; 324(1): 88-98, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18835385

RESUMEN

The mammalian metanephric kidney is derived from the intermediate mesoderm. In this report, we use molecular fate mapping to demonstrate that the majority of cell types within the metanephric kidney arise from an Osr1(+) population of metanephric progenitor cells. These include the ureteric epithelium of the collecting duct network, the cap mesenchyme and its nephron epithelia derivatives, the interstitial mesenchyme, vasculature and smooth muscle. Temporal fate mapping shows a progressive restriction of Osr1(+) cell fates such that at the onset of active nephrogenesis, Osr1 activity is restricted to the Six2(+) cap mesenchyme nephron progenitors. However, low-level labeling of Osr1(+) cells suggests that the specification of interstitial mesenchyme and cap mesenchyme progenitors occurs within the Osr1(+) population prior to the onset of metanephric development. Furthermore, although Osr1(+) progenitors give rise to much of the kidney, Osr1 function is only essential for the development of the nephron progenitor compartment. These studies provide new insights into the cellular origins of metanephric kidney structures and lend support to a model where Osr1 function is limited to establishing the nephron progenitor pool.


Asunto(s)
Riñón/embriología , Mesodermo/embriología , Nefronas/embriología , Células Madre/citología , Factores de Transcripción/biosíntesis , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Proteínas de Homeodominio/metabolismo , Riñón/citología , Riñón/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Mutantes , Nefronas/citología , Nefronas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Urotelio/embriología , Urotelio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA