Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31813624

RESUMEN

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Interacciones Microbiota-Huesped/fisiología , Nociceptores/fisiología , Animales , Epitelio/metabolismo , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/microbiología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Nociceptores/metabolismo , Ganglios Linfáticos Agregados/inervación , Ganglios Linfáticos Agregados/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología
2.
Hum Mol Genet ; 28(5): 718-735, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30371777

RESUMEN

Large-scale 'omic' studies investigating the pathophysiological processes that lead to Alzheimer's disease (AD) dementia have identified an increasing number of susceptibility genes, many of which are poorly characterized and have not previously been implicated in AD. Here, we evaluated the utility of human induced pluripotent stem cell-derived neurons and astrocytes as tools to systematically test AD-relevant cellular phenotypes following perturbation of candidate genes identified by genome-wide studies. Lentiviral-mediated delivery of shRNAs was used to modulate expression of 66 genes in astrocytes and 52 genes in induced neurons. Five genes (CNN2, GBA, GSTP1, MINT2 and FERMT2) in neurons and nine genes (CNN2, ITGB1, MINT2, SORL1, VLDLR, NPC1, NPC2, PSAP and SCARB2) in astrocytes significantly altered extracellular amyloid-ß (Aß) levels. Knockdown of AP3M2, CNN2, GSTP1, NPC1, NPC2, PSAP and SORL1 reduced interleukin-6 levels in astrocytes. Only knockdown of FERMT2 led to a reduction in the proportion of TAU that is phosphorylated. Further, CRISPR-Cas9 targeting of FERMT2 in both familial AD (fAD) and fAD-corrected human neurons validated the findings of reduced extracellular Aß. Interestingly, FERMT2 reduction had no effect on the Aß42:40 ratio in corrected neurons and a reduction of phospho-tau, but resulted in an elevation in Aß42:40 ratio and no reduction in phospho-tau in fAD neurons. Taken together, this study has prioritized 15 genes as being involved in contributing to Aß accumulation, phosphorylation of tau and/or cytokine secretion, and, as illustrated with FERMT2, it sets the stage for further cell-type-specific dissection of the role of these genes in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Neuronas/metabolismo , Proteostasis , Proteínas tau/metabolismo , Biomarcadores , Encéfalo/metabolismo , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Marcación de Gen , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Fenotipo
3.
Mov Disord ; 30(14): 1961-4, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26573701

RESUMEN

BACKGROUND: Biomarkers for Huntington's disease progression could accelerate therapeutic developments and improve patient care. Brain microRNAs relating to clinical features of Huntington's disease may represent a potential Huntington's disease biomarker in blood. OBJECTIVE: This study was undertaken to examine candidate microRNAs in plasma to determine whether changes observed in HD brains are detectable in peripheral samples. METHODS: Four microRNAs from 26 manifest Huntington's disease, four asymptomatic Huntington's disease gene carriers, and eight controls were quantified in plasma using reverse transcription quantitative polymerase chain reaction. Linear regression was used to assess microRNA levels across control, asymptomatic gene carriers, and manifest patients. RESULTS: miR-10b-5p (P = 0.0068) and miR-486-5p (P = 0.044) were elevated in Huntington's disease plasma. miR-10b-5p was decreased in asymptomatic gene carriers as compared with patients with Huntington's disease (P = 0.049), but no difference between asymptomatic gene carriers and healthy controls was observed (P = 0.24). CONCLUSIONS: These findings suggest that microRNA changes observed in Huntington's disease brain may be detectable in plasma and have potential clinical utility.


Asunto(s)
Encéfalo/patología , Enfermedad de Huntington/metabolismo , MicroARNs/sangre , Adolescente , Adulto , Biomarcadores/sangre , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Acta Neuropathol Commun ; 11(1): 39, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36899414

RESUMEN

Despite ongoing debate, the amyloid ß-protein (Aß) remains the prime therapeutic target for the treatment of Alzheimer's disease (AD). However, rational drug design has been hampered by a lack of knowledge about neuroactive Aß. To help address this deficit, we developed live-cell imaging of iPSC-derived human neurons (iNs) to study the effects of the most disease relevant form of Aß-oligomeric assemblies (oAß) extracted from AD brain. Of ten brains studied, extracts from nine caused neuritotoxicity, and in eight cases this was abrogated by Aß immunodepletion. Here we show that activity in this bioassay agrees relatively well with disruption of hippocampal long-term potentiation, a correlate of learning and memory, and that measurement of neurotoxic oAß can be obscured by more abundant non-toxic forms of Aß. These findings indicate that the development of novel Aß targeting therapeutics may benefit from unbiased activity-based discovery. To test this principle, we directly compared 5 clinical antibodies (aducanumab, bapineuzumab,  BAN2401, gantenerumab, and SAR228810) together with an in-house aggregate-preferring antibody (1C22) and established relative EC50s in protecting human neurons from human Aß. The results yielded objective numerical data on the potency of each antibody in neutralizing human oAß neuritotoxicity. Their relative efficacies in this morphological assay were paralleled by their functional ability to rescue oAß-induced inhibition of hippocampal synaptic plasticity. This novel paradigm provides an unbiased, all-human system for selecting candidate antibodies for advancement to human immunotherapy.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Inmunoterapia , Neuronas/metabolismo
5.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34847080

RESUMEN

Functional gastrointestinal disorders (FGIDs) have prominent sex differences in incidence, symptoms, and treatment response that are not well understood. Androgens are steroid hormones present at much higher levels in males than females and could be involved in these differences. In adults with irritable bowel syndrome (IBS), a FGID that affects 5% to 10% of the population worldwide, we found that free testosterone levels were lower than those in healthy controls and inversely correlated with symptom severity. To determine how this diminished androgen signaling could contribute to bowel dysfunction, we depleted gonadal androgens in adult mice and found that this caused a profound deficit in gastrointestinal transit. Restoring a single androgen hormone was sufficient to rescue this deficit, suggesting that circulating androgens are essential for normal bowel motility in vivo. To determine the site of action, we probed androgen receptor expression in the intestine and discovered, unexpectedly, that a large subset of enteric neurons became androgen-responsive upon puberty. Androgen signaling to these neurons was required for normal colonic motility in adult mice. Taken together, these observations establish a role for gonadal androgens in the neural regulation of bowel function and link altered androgen levels with a common digestive disorder.


Asunto(s)
Andrógenos/sangre , Colon/metabolismo , Motilidad Gastrointestinal , Síndrome del Colon Irritable/sangre , Receptores Androgénicos/biosíntesis , Adulto , Animales , Colon/fisiopatología , Femenino , Humanos , Síndrome del Colon Irritable/metabolismo , Síndrome del Colon Irritable/fisiopatología , Masculino , Ratones
6.
Neurobiol Pain ; 9: 100056, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33392418

RESUMEN

Nociceptor sensory neurons innervate barrier tissues that are constantly exposed to microbial stimuli. During infection, pathogenic microorganisms can breach barrier surfaces and produce pain by directly activating nociceptors. Microorganisms that live in symbiotic relationships with their hosts, commensals and mutualists, have also been associated with pain, but the molecular mechanisms of how symbionts act on nociceptor neurons to modulate pain remain largely unknown. In this review, we will discuss the known molecular mechanisms of how microbes directly interact with sensory afferent neurons affecting nociception in the gut, skin and lungs. We will touch on how bacterial, viral and fungal pathogens signal to the host to inflict or suppress pain. We will also discuss recent studies examining how gut symbionts affect pain. Specifically, we will discuss how gut symbionts may interact with sensory afferent neurons either directly, through secretion of metabolites or neurotransmitters, or indirectly,through first signaling to epithelial cells or immune cells, to regulate visceral, neuropathic and inflammatory pain. While this area of research is still in its infancy, more mechanistic studies to examine microbial-sensory neuron crosstalk in nociception may allow us to develop new therapies for the treatment of acute and chronic pain.

7.
Neuron ; 109(21): 3402-3420.e9, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34473944

RESUMEN

We have generated a controlled and manipulable resource that captures genetic risk for Alzheimer's disease: iPSC lines from 53 individuals coupled with RNA and proteomic profiling of both iPSC-derived neurons and brain tissue of the same individuals. Data collected for each person include genome sequencing, longitudinal cognitive scores, and quantitative neuropathology. The utility of this resource is exemplified here by analyses of neurons derived from these lines, revealing significant associations between specific Aß and tau species and the levels of plaque and tangle deposition in the brain and, more importantly, with the trajectory of cognitive decline. Proteins and networks are identified that are associated with AD phenotypes in iPSC neurons, and relevant associations are validated in brain. The data presented establish this iPSC collection as a resource for investigating person-specific processes in the brain that can aid in identifying and validating molecular pathways underlying AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Anciano , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Cognición , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Proteómica , Proteínas tau/genética , Proteínas tau/metabolismo
8.
Brain Commun ; 2(1): fcaa010, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32280944

RESUMEN

Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other 'synucleinopathies'. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.

9.
Transl Psychiatry ; 8(1): 245, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30410030

RESUMEN

The identification of convergent phenotypes in different models of psychiatric illness highlights robust phenotypes that are more likely to be implicated in disease pathophysiology. Here, we utilize human iPSCs harboring distinct mutations in DISC1 that have been found in families with major mental illness. One mutation was engineered to mimic the consequences on DISC1 protein of a balanced translocation linked to mental illness in a Scottish pedigree; the other mutation was identified in an American pedigree with a high incidence of mental illness. Directed differentiation of these iPSCs using NGN2 expression shows rapid conversion to a homogenous population of mature excitatory neurons. Both DISC1 mutations result in reduced DISC1 protein expression, and show subtle effects on certain presynaptic proteins. In addition, RNA sequencing and qPCR showed decreased expression of UNC5D, DPP10, PCDHA6, and ZNF506 in neurons with both DISC1 mutations. Longitudinal analysis of neurite outgrowth revealed decreased neurite outgrowth in neurons with each DISC1 mutation, which was mimicked by UNC5D knockdown and rescued by transient upregulation of endogenous UNC5D. This study shows a narrow range of convergent phenotypes of two mutations found in families with major mental illness, and implicates dysregulated netrin signaling in DISC1 biology.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Receptores de Netrina/metabolismo , Neuritas , Neuronas , Receptores de Superficie Celular/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Linaje , Análisis de Secuencia de ARN , Transcriptoma
10.
Transl Psychiatry ; 8(1): 77, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29643329

RESUMEN

The development of three-dimensional culture methods has allowed for the study of developing cortical morphology in human cells. This provides a new tool to study the neurodevelopmental consequences of disease-associated mutations. Here, we study the effects of isogenic DISC1 mutation in cerebral organoids. DISC1 has been implicated in psychiatric disease based on genetic studies, including its interruption by a balanced translocation that increases the risk of major mental illness. Isogenic wild-type and DISC1-disrupted human-induced pluripotent stem cells were used to generate cerebral organoids, which were then examined for morphology and gene expression. We show that DISC1-mutant cerebral organoids display disorganized structural morphology and impaired proliferation, which is phenocopied by WNT agonism and rescued by WNT antagonism. Furthermore, there are many shared changes in gene expression with DISC1 disruption and WNT agonism, including in neural progenitor and cell fate markers, regulators of neuronal migration, and interneuron markers. These shared gene expression changes suggest mechanisms for the observed morphologic dysregulation with DISC1 disruption and points to new avenues for future studies. The shared changes in three-dimensional cerebral organoid morphology and gene expression with DISC1 interruption and WNT agonism further strengthens the link between DISC1 mutation, abnormalities in WNT signaling, and neuropsychiatric disease.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vía de Señalización Wnt , Apoptosis , Proliferación Celular , Corteza Cerebral/patología , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Proteínas del Tejido Nervioso/genética , Organoides/metabolismo , Organoides/patología , Técnicas de Cultivo de Tejidos
11.
Nat Commun ; 9(1): 2676, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29992960

RESUMEN

Although the amyloid ß-protein (Aß) is believed to play an initiating role in Alzheimer's disease (AD), the molecular characteristics of the key pathogenic Aß forms are not well understood. As a result, it has proved difficult to identify optimal agents that target disease-relevant forms of Aß. Here, we combined the use of Aß-rich aqueous extracts of brain samples from AD patients as a source of human Aß and live-cell imaging of iPSC-derived human neurons to develop a bioassay capable of quantifying the relative protective effects of multiple anti-Aß antibodies. We report the characterization of 1C22, an aggregate-preferring murine anti-Aß antibody, which better protects against forms of Aß oligomers that are toxic to neurites than do the murine precursors of the clinical immunotherapeutics, bapineuzumab and solanezumab. These results suggest further examination of 1C22 is warranted, and that this bioassay maybe useful as a primary screen to identify yet more potent anti-Aß therapeutics.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales/inmunología , Encéfalo/inmunología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo
12.
Stem Cell Reports ; 9(6): 1868-1884, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29153990

RESUMEN

Alzheimer's disease (AD) induces memory and cognitive impairment in the absence of motor and sensory deficits during its early and middle course. A major unresolved question is the basis for this selective neuronal vulnerability. Aß, which plays a central role in AD pathogenesis, is generated throughout the brain, yet some regions outside of the limbic and cerebral cortices are relatively spared from Aß plaque deposition and synapse loss. Here, we examine neurons derived from iPSCs of patients harboring an amyloid precursor protein mutation to quantify AD-relevant phenotypes following directed differentiation to rostral fates of the brain (vulnerable) and caudal fates (relatively spared) in AD. We find that both the generation of Aß and the responsiveness of TAU to Aß are affected by neuronal cell type, with rostral neurons being more sensitive than caudal neurons. Thus, cell-autonomous factors may in part dictate the pattern of selective regional vulnerability in human neurons in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/patología , Ratones , Neuronas/patología , Fenotipo , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA