Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Development ; 140(6): 1342-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23444360

RESUMO

Wnt1-expressing progenitors generate midbrain dopamine (MbDA) and cerebellum (Cb) neurons in distinct temporal windows and from spatially discrete progenitor domains. It has been shown that Wnt1 and Lmx1a participate in a cross-regulatory loop that is utilized during MbDA neuron development. However, Wnt1 expression dynamically changes over time and precedes that of Lmx1a. The spatial and temporal requirements of Wnt1 in development and specifically its requirement for MbDA neurons remain to be determined. To address these issues, we generated a conditional Wnt1 allele and temporally deleted Wnt1 coupled with genetic lineage analysis. Using this approach, we show that patterning of the midbrain (Mb) and Cb by Wnt1 occurs between the one-somite and the six- to eight-somite stages and is solely dependent on Wnt1 function in the Mb, but not in the Cb. Interestingly, an En1-derived domain persists after the early deletion of Wnt1 and mutant cells express OTX2. However, the En1-derived Wnt1-mutant domain does not contain LMX1a-expressing progenitors, and MbDA neurons are depleted. Thus, we demonstrate an early requirement of Wnt1 for all MbDA neurons. Subsequently, we deleted Wnt1 in the ventral Mb and show a continued late requirement for Wnt1 in MbDA neuron development, but not in LMX1a-expressing progenitors. Specifically, Wnt1 deletion disrupts the birthdating of MbDA neurons and causes a depletion of MbDA neurons positioned medially and a concomitant expansion of MbDA neurons positioned laterally during embryogenesis. Collectively, our analyses resolve the spatial and temporal function of Wnt1 in Mb and Cb patterning and in MbDA neuron development in vivo.


Assuntos
Diferenciação Celular/genética , Neurônios Dopaminérgicos/fisiologia , Mesencéfalo/embriologia , Proteína Wnt1/fisiologia , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Cerebelo/embriologia , Cerebelo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cinética , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Especificidade de Órgãos/genética , Gravidez , Fatores de Tempo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
2.
Dev Biol ; 372(2): 249-62, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23041116

RESUMO

Midbrain dopamine (MbDA) neurons are partitioned into medial and lateral cohorts that control complex functions. However, the genetic underpinnings of MbDA neuron heterogeneity are unclear. While it is known that Wnt1-expressing progenitors contribute to MbDA neurons, the role of Wnt1 in MbDA neuron development in vivo is unresolved. We show that mice with a spontaneous point mutation in Wnt1 have a unique phenotype characterized by the loss of medial MbDA neurons concomitant with a severe depletion of Wnt1-expressing progenitors and diminished LMX1a-expressing progenitors. Wnt1 mutant embryos also have alterations in a hierarchical gene regulatory loop suggesting multiple gene involvement in the Wnt1 mutant MbDA neuron phenotype. To investigate this possibility, we conditionally deleted Gbx2, Fgf8, and En1/2 after their early role in patterning and asked whether these genetic manipulations phenocopied the depletion of MbDA neurons in Wnt1 mutants. The conditional deletion of Gbx2 did not result in re-positioning or distribution of MbDA neurons. The temporal deletion of Fgf8 did not result in the loss of either LMX1a-expressing progenitors nor the initial population of differentiated MbDA neurons, but did result in a complete loss of MbDA neurons at later stages. The temporal deletion and species specific manipulation of En1/2 demonstrated a continued and species specific role of Engrailed genes in MbDA neuron development. Notably, our conditional deletion experiments revealed phenotypes dissimilar to Wnt1 mutants indicating the unique role of Wnt1 in MbDA neuron development. By placing Wnt1, Fgf8, and En1/2 in the context of their temporal requirement for MbDA neuron development, we further deciphered the developmental program underpinning MbDA neuron progenitors.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Animais , Mesencéfalo/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
3.
Mol Cell Neurosci ; 49(2): 217-29, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22173107

RESUMO

The cerebellum (Cb) controls movement related physiology using a diverse array of morphologically and biochemically distinct neurons. During development, the Cb is derived from rhombomere 1 (r1), an embryonic compartment patterned by a signaling center referred to as the isthmus organizer. The secreted glycoprotein WNT1 is expressed in the midbrain primordia (mesencephalon, mes) and at the posterior limit of the mes. WNT1 plays a pivotal role in maintaining the isthmus organizer and mutations in Wnt1 produce severe Cb defects that are generally attributed to aberrant organizer activity. Interestingly, Wnt1 is also expressed at the most posterior limit of dorsal r1, in a region known as the upper rhombic lip (URL). However, the distribution and molecular identity of Wnt1 expressing progenitors have not been carefully described in r1. We used Wnt1-Venus transgenic mice to generate a molecular map of Wnt1 expressing progenitors in relation to other well characterized Cb biomarkers such as MATH1 (ATOH1), LMX1a and OTX2. Our analysis validated Wnt1 expression in the URL and revealed molecularly-defined developmental zones in r1. We then used genetic inducible fate mapping (GIFM) to link transient Wnt1 expression in r1 to terminal cell fates in the mature Cb. Wnt1 expressing progenitors primarily contributed to neurons in deep cerebellar nuclei, granule cells, and unipolar brush cells in distinct but overlapping temporal windows and sparsely contributed to inhibitory neurons and Bergmann glia. We further demonstrate that the Wnt1 lineage does not follow a competency model of progressive lineage restriction to generate the Cb or the functionally related precerebellar system. Instead, progenitors initiate Wnt1 expression de novo to give rise to each Cb cell type and precerebellar nuclei. We also used GIFM to determine how the temporal control of Wnt1 expression is related to molecular identity and cell migration in Cb development. Our findings provide new insight into how lineage and timing establish cell diversity within the Cb system.


Assuntos
Cerebelo/citologia , Cerebelo/embriologia , Neurônios/metabolismo , Proteína Wnt1/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Cerebelo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Fatores de Tempo , Proteína Wnt1/fisiologia
4.
J Psychopharmacol ; 37(11): 1051-1057, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37522187

RESUMO

Animal models are important in preclinical psychopharmacology to study mechanisms and potential treatments for psychiatric disorders. A working group of 14 volunteers, comprising an international team of researchers from academia and industry, convened in 2021 to discuss how to improve the translational relevance and interpretation of findings from animal models that are used in preclinical psychopharmacology. The following paper distils the outcomes of the working group's discussions into 10 key considerations for the planning and reporting of behavioural studies in animal models relevant to psychiatric disorders. These form the iTRIPP guidelines (Improving Translational Relevance In Preclinical Psychopharmacology). These guidelines reflect the key considerations that the group thinks will likely have substantial impact in terms of improving the translational relevance of behavioural studies in animal models that are used to study psychiatric disorders and their treatment. They are relevant to the research community when drafting and reviewing manuscripts, presentations and grant applications. The iTRIPP guidelines are intended to complement general recommendations for planning and reporting animal studies that have been published elsewhere, by enabling researchers to fully consider the most appropriate animal model for the research purpose and to interpret their findings appropriately. This in turn will increase the clinical benefit of such research and is therefore important not only for the scientific community but also for patients and the lay public.


Assuntos
Transtornos Mentais , Psicofarmacologia , Animais , Humanos , Transtornos Mentais/tratamento farmacológico , Modelos Animais de Doenças
5.
Mol Cell Neurosci ; 45(2): 132-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600933

RESUMO

Conditional marking and gene inactivation experiments are valuable approaches used to understand developmental and molecular mechanisms. CreER(T) is a fundamental component in recombinase-based conditional strategies and is used to gain temporal control subsequent to tamoxifen administration. We tested the hypothesis that tamoxifen dose linearly correlates with recombination efficiency in vivo. Wnt1-CreER(T) and tamoxifen administration were used to mark progenitors that contributed to the trigeminal ganglia. We executed a dose response study to determine the number of neurons that had undergone recombination in response to tamoxifen administered at doses ranging from 50 to 500 mg/kg. Our findings show a substantial variability in the amount of recombination within and between dose groups with no clear correlation between tamoxifen dose and the number of marked cells. This is the first study that we are aware of in which cell counts, robust quantitative data, and statistical analyses were performed on sections obtained from embryos marked in response to a wide range of tamoxifen dose in vivo. We provide an important quantitative and statistical framework for designing CreER(T)-based experiments and choosing tamoxifen dosing paradigms.


Assuntos
Células-Tronco Neurais/efeitos dos fármacos , Recombinação Genética/efeitos dos fármacos , Tamoxifeno/administração & dosagem , Gânglio Trigeminal/citologia , Proteína Wnt1/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Camundongos , Camundongos Transgênicos
6.
Neuron ; 43(3): 345-57, 2004 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15294143

RESUMO

Brain structures derived from the mesencephalon (mes) and rhombomere 1 (r1) modulate distinct motor and sensory modalities. The precise origin and cellular behaviors underpinning the cytoarchitectural organization of the mes and r1, however, are unknown. Using a novel inducible genetic fate mapping approach in mouse, we determined the fate and lineage relationships of mes/r1 cells with fine temporal and spatial resolution. We demonstrate that the mes and r1 are neuromeres that along with the isthmic organizer are partitioned along the anterior-posterior axis by lineage restriction boundaries established sequentially between E8.5 and E9.5. Furthermore, a small group of cells originating from the most posterior mes exhibit anterior intracompartmental expansion and contribute throughout the inferior colliculus. Finally, we also uncovered transient and differential genetic lineages of ventral midbrain dopaminergic and ventral hindbrain serotonergic neuronal precursors with respect to Wnt1 and Gli1 expression.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Animais , Linhagem da Célula/genética , Feminino , Fatores de Transcrição Kruppel-Like , Masculino , Mesencéfalo/citologia , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Gravidez , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Rombencéfalo/citologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteínas Wnt , Proteína Wnt1 , Proteína GLI1 em Dedos de Zinco
7.
Nat Commun ; 9(1): 3456, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150678

RESUMO

Discovery of coding variants in genes that confer risk of neurodevelopmental disorders is an important step towards understanding the pathophysiology of these disorders. Whole-genome sequencing of 31,463 Icelanders uncovers a frameshift variant (E712KfsTer10) in microtubule-associated protein 1B (MAP1B) that associates with ID/low IQ in a large pedigree (genome-wide corrected P = 0.022). Additional stop-gain variants in MAP1B (E1032Ter and R1664Ter) validate the association with ID and IQ. Carriers have 24% less white matter (WM) volume (ß = -2.1SD, P = 5.1 × 10-8), 47% less corpus callosum (CC) volume (ß = -2.4SD, P = 5.5 × 10-10) and lower brain-wide fractional anisotropy (P = 6.7 × 10-4). In summary, we show that loss of MAP1B function affects general cognitive ability through a profound, brain-wide WM deficit with likely disordered or compromised axons.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Substância Branca/metabolismo , Substância Branca/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cognição/fisiologia , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Linhagem
8.
Front Neuroanat ; 11: 67, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28878630

RESUMO

The auditory system contains a diverse array of interconnected anatomical structures that mediate the perception of sound. The cochlear nucleus of the hindbrain serves as the initial site of convergence for auditory stimuli, while the inferior colliculus of the midbrain serves as an integration and relay station for all ascending auditory information. We used Genetic Inducible Fate Mapping (GIFM) to determine how the timing of Wnt1 expression is related to the competency states of auditory neuron progenitors. We demonstrate that the Wnt1 lineage defines progenitor pools of auditory neurons in the developing midbrain and hindbrain. The timing of Wnt1 expression specifies unique cell types during embryogenesis and follows a mixed model encompassing a brief epoch of de novo expression followed by rapid and progressive lineage restriction to shape the inferior colliculus. In contrast, Wnt1 fate mapping of the embryonic hindbrain revealed de novo induction of Wnt1 in auditory hindbrain progenitors, which is related to the development of biochemically distinct neurons in the cochlear nucleus. Thus, we uncovered two modes of lineage allocation that explain the relationship between the timing of Wnt1 expression and the development of the cochlear nucleus and the inferior colliculus. Finally, our analysis of Wnt1sw/sw mutant mice demonstrated a functional requirement of Wnt1 for the development of auditory midbrain and hindbrain neurons. Collectively, our study provides a deeper understanding of Wnt1 lineage allocation and function in mammalian brain development.

9.
Front Neuroanat ; 11: 50, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28785208

RESUMO

The cerebellum (Cb) is an exquisite structure that controls elaborate motor behaviors and is essential for sensory-motor learning. During development, the Cb is derived from rhombomere 1 (r1). Within this embryonic compartment, precursors in r1 are patterned by signaling cues originating from the isthmus organizer (IsO) and subsequently undergo complex morphogenic movements to establish their final position in the mature Cb. The transcription factor Gbx2 is expressed in the developing Cb and is intimately involved in organizing and patterning the Cb. Nevertheless, how precursors expressing Gbx2 at specific embryonic time points contribute to distinct cell types in the adult Cb is unresolved. In this study, we used Genetic Inducible Fate Mapping (GIFM) to mark Gbx2-expressing precursors with fine temporal resolution and to subsequently track this lineage through embryogenesis. We then determined the terminal neuronal fate of the Gbx2 lineage in the adult Cb. Our analysis demonstrates that the Gbx2 lineage contributes to the Cb with marking over the course of five stages: Embryonic day 7.5 (E7.5) through E11.5. The Gbx2 lineage gives rise to Purkinje cells, granule neurons, and deep cerebellar neurons across these marking stages. Notably, the contribution of the Gbx2 lineage shifts as development proceeds with each marking stage producing a distinct profile of mature neurons in the adult Cb. These findings demonstrate the relationship between the temporal expression of Gbx2 and the terminal cell fate of neurons in the Cb. Based on these results, Gbx2 is critical to Cb development, not only for its well-defined role in positioning and maintaining the IsO, but also for guiding the development of Cb precursors and determining the identity of Cb neurons.

10.
Pediatr Neurol ; 60: 1-12, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27267556

RESUMO

On March 10 to March 12, 2015, the National Institute of Neurological Disorders and Stroke and the Tuberous Sclerosis Alliance sponsored a workshop in Bethesda, Maryland, to assess progress and new opportunities for research in tuberous sclerosis complex with the goal of updating the 2003 Research Plan for Tuberous Sclerosis (http://www.ninds.nih.gov/about_ninds/plans/tscler_research_plan.htm). In addition to the National Institute of Neurological Disorders and Stroke and Tuberous Sclerosis Alliance, participants in the strategic planning effort and workshop included representatives from six other Institutes of the National Institutes of Health, the Department of Defense Tuberous Sclerosis Complex Research Program, and a broad cross-section of basic scientists and clinicians with expertise in tuberous sclerosis complex along with representatives from the pharmaceutical industry. Here we summarize the outcomes from the extensive premeeting deliberations and final workshop recommendations, including (1) progress in the field since publication of the initial 2003 research plan for tuberous sclerosis complex, (2) the key gaps, needs, and challenges that hinder progress in tuberous sclerosis complex research, and (3) a new set of research priorities along with specific recommendations for addressing the major challenges in each priority area. The new research plan is organized around both short-term and long-term goals with the expectation that progress toward specific objectives can be achieved within a five to ten year time frame.


Assuntos
Pesquisa Biomédica , Esclerose Tuberosa/fisiopatologia , Esclerose Tuberosa/terapia , Animais , Modelos Animais de Doenças , Objetivos , Humanos , Planejamento Estratégico , Esclerose Tuberosa/genética , Estados Unidos
11.
Neuron ; 78(5): 895-909, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23664552

RESUMO

Tuberous sclerosis is a developmental genetic disorder caused by mutations in TSC1, which results in epilepsy, autism, and intellectual disability. The cause of these neurological deficits remains unresolved. Imaging studies suggest that the thalamus may be affected in tuberous sclerosis patients, but this has not been experimentally interrogated. We hypothesized that thalamic deletion of Tsc1 at distinct stages of mouse brain development would produce differential phenotypes. We show that mosaic Tsc1 deletion within thalamic precursors at embryonic day (E) 12.5 disrupts thalamic circuitry and alters neuronal physiology. Tsc1 deletion at this early stage is unique in causing both seizures and compulsive grooming in adult mice. In contrast, only a subset of these phenotypes occurs when thalamic Tsc1 is deleted at a later embryonic stage. Our findings demonstrate that abnormalities in a discrete population of neurons can cause global brain dysfunction and that phenotype severity depends on developmental timing and degree of genetic mosaicism.


Assuntos
Comportamento Animal/fisiologia , Córtex Cerebral/fisiologia , Neurônios/fisiologia , Deleção de Sequência/genética , Tálamo , Proteínas Supressoras de Tumor/genética , Animais , Animais Recém-Nascidos , Biofísica , Mapeamento Encefálico , Proteínas de Ligação a DNA/metabolismo , Estimulação Elétrica , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Asseio Animal/fisiologia , Força da Mão/fisiologia , Proteínas de Homeodomínio/genética , Hiperalgesia/genética , Técnicas In Vitro , Modelos Lineares , Masculino , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Proteína Básica da Mielina/metabolismo , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/fisiologia , Proteínas Nucleares/metabolismo , Medição da Dor , Técnicas de Patch-Clamp , Fosfopiruvato Hidratase/metabolismo , Estimulação Física , Gravidez , Proteínas/genética , RNA não Traduzido , Convulsões/genética , Convulsões/fisiopatologia , Tamoxifeno/farmacologia , Tálamo/citologia , Tálamo/crescimento & desenvolvimento , Tálamo/fisiologia , Proteína 1 do Complexo Esclerose Tuberosa , Ubiquitina-Proteína Ligases , Vibrissas/inervação
12.
PLoS One ; 6(6): e20940, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698205

RESUMO

BACKGROUND: Forging a relationship between progenitors with dynamically changing gene expression and their terminal fate is instructive for understanding the logic of how cell-type diversity is established. The mouse spinal cord is an ideal system to study these mechanisms in the context of developmental genetics and nervous system development. Here we focus on the Gastrulation homeobox 2 (Gbx2) transcription factor, which has not been explored in spinal cord development. METHODOLOGY/PRINCIPAL FINDINGS: We determined the molecular identity of Gbx2-expressing spinal cord progenitors. We also utilized genetic inducible fate mapping to mark the Gbx2 lineage at different embryonic stages in vivo in mouse. Collectively, we uncover cell behaviors, cytoarchitectonic organization, and the terminal cell fate of the Gbx2 lineage. Notably, both ventral motor neurons and interneurons are derived from the Gbx2 lineage, but only during a short developmental period. Short-term fate mapping during mouse spinal cord development shows that Gbx2 expression is transient and is extinguished ventrally in a rostral to caudal gradient. Concomitantly, a permanent lineage restriction boundary ensures that spinal cord neurons derived from the Gbx2 lineage are confined to a dorsal compartment that is maintained in the adult and that this lineage generates inhibitory interneurons of the spinal cord. Using lineage tracing and molecular markers to follow Gbx2-mutant cells, we show that the loss of Gbx2 globally affects spinal cord patterning including the organization of interneuron progenitors. Finally, long-term lineage analysis reveals that the presence and timing of Gbx2 expression in interneuron progenitors results in the differential contribution to subtypes of terminally differentiated interneurons in the adult spinal cord. CONCLUSIONS/SIGNIFICANCE: We illustrate the complex cellular nature of Gbx2 expression and lineage contribution to the mouse spinal cord. In a broader context, this study provides a direct link between spinal cord progenitors undergoing dynamic changes in molecular identity and terminal neuronal fate.


Assuntos
Linhagem da Célula , Proteínas de Homeodomínio/fisiologia , Medula Espinal/crescimento & desenvolvimento , Animais , Camundongos , Medula Espinal/citologia
13.
J Comp Neurol ; 519(15): 2978-3000, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21713770

RESUMO

Midbrain dopamine (MbDA) neurons are functionally heterogeneous and modulate complex functions through precisely organized anatomical groups. MbDA neurons are generated from Wnt1-expressing progenitors located in the ventral mesencephalon (vMes) during embryogenesis. However, it is unclear whether the progenitor pool is partitioned into distinct cohorts based on molecular identity and whether the timing of gene expression uniquely identifies subtypes of MbDA neurons. In this study we show that Wnt1-expressing MbDA progenitors from embryonic day (E)8.5-12.5 have dynamic molecular identities that correlate with specific spatial locations in the vMes. We also tested the hypothesis that the timing of Wnt1 expression in progenitors is related to the distribution of anatomically distinct cohorts of adult MbDA neurons using genetic inducible fate mapping (GIFM). We demonstrate that the Wnt1 lineage contributes to specific cohorts of MbDA neurons during a 7-day epoch and that the contribution to MbDA neurons predominates over other ventral Mb domains. In addition, we show that calbindin-, GIRK2-, and calretinin-expressing MbDA neuron subtypes are derived from Wnt1-expressing progenitors marked over a broad temporal window. Through GIFM and quantitative analysis we demonstrate that the Wnt1 lineage does not undergo progressive lineage restriction, which eliminates a restricted competence model of generating MbDA diversity. Interestingly, we uncover that two significant peaks of Wnt1 lineage contribution to MbDA neurons occur at E9.5 and E11.5. Collectively, our findings delineate the temporal window of MbDA neuron generation and show that lineage and timing predicts the terminal distribution pattern of MbDA neurons.


Assuntos
Dopamina/metabolismo , Mesencéfalo/citologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Proteína Wnt1/metabolismo , Animais , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/fisiologia , Antagonistas de Estrogênios/farmacologia , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células-Tronco/citologia , Tamoxifeno/farmacologia , Proteína Wnt1/genética
14.
J Comp Neurol ; 519(15): 3001-18, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21713771

RESUMO

The ventral midbrain (vMb) is organized into distinct anatomical domains and contains cohorts of functionally distinct subtypes of midbrain dopamine (mDA) neurons. We tested the hypothesis that genetic history and timing of gene expression within mDA neuron progenitors impart spatial diversity. Using genetic inducible fate mapping to mark the Sonic hedgehog (Shh) and Gli1 lineages at varying embryonic stages, we performed a quantitative and qualitative comparison of the two lineages' contribution to the mDA neuron domains. Dynamic changes in Shh and Gli1 expression in the vMb primordia delineated their spatial contribution to the embryonic day 12.5 vMb: Both lineages first contributed to the medial domain, but subsequently the Gli1 lineage exclusively contributed to the lateral vMb while the Shh lineage expanded more broadly across the vMb. The contribution of both lineages to the differentiated mDA neuron domain was initially biased anteriorly and became more uniform across the anterior/posterior vMb throughout development. Our findings demonstrate that the early Shh and Gli1 lineages specify mDA neurons of the substantia nigra pars compacta while the late Shh and Gli1 lineages maintain their progenitor state longer in the posterior vMb to extend the production of mDA neurons in the ventral tegmental area. Together, our study demonstrates that the timing of gene expression along with the genetic lineage (Shh or Gli1) within the neural progenitors segregate mDA neurons into distinct spatial domains.


Assuntos
Dopamina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Mesencéfalo/citologia , Neurônios/fisiologia , Animais , Linhagem da Célula , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Feminino , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Masculino , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Proteína GLI1 em Dedos de Zinco
15.
J Vis Exp ; (34)2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-20042997

RESUMO

Fate maps are generated by marking and tracking cells in vivo to determine how progenitors contribute to specific structures and cell types in developing and adult tissue. An advance in this concept is Genetic Inducible Fate Mapping (GIFM), linking gene expression, cell fate, and cell behaviors in vivo, to create fate maps based on genetic lineage. GIFM exploits X-CreER lines where X is a gene or set of gene regulatory elements that confers spatial expression of a modified bacteriophage protein, Cre recombinase (CreER(T)). CreER(T) contains a modified estrogen receptor ligand binding domain which renders CreER(T) sequestered in the cytoplasm in the absence of the drug tamoxifen. The binding of tamoxifen releases CreER(T), which translocates to the nucleus and mediates recombination between DNA sequences flanked by loxP sites. In GIFM, recombination typically occurs between a loxP flanked Stop cassette preceding a reporter gene such as GFP. Mice are bred to contain either a region- or cell type-specific CreER and a conditional reporter allele. Untreated mice will not have marking because the Stop cassette in the reporter prevents further transcription of the reporter gene. We administer tamoxifen by oral gavage to timed-pregnant females, which provides temporal control of CreER(T) release and subsequent translocation to the nucleus removing the Stop cassette from the reporter. Following recombination, the reporter allele is constitutively and heritably expressed. This series of events marks cells such that their genetic history is indelibly recorded. The recombined reporter thus serves as a high fidelity genetic lineage tracer that, once on, is uncoupled from the gene expression initially used to drive CreER(T). We apply GIFM in mouse to study normal development and ascertain the contribution of genetic lineages to adult cell types and tissues. We also use GIFM to follow cells on mutant genetic backgrounds to better understand complex phenotypes that mimic salient features of human genetic disorders. This video article guides researchers through experimental methods to successfully apply GIFM. We demonstrate the method using our well characterized Wnt1-CreER(T);mGFP mice by administering tamoxifen at embryonic day (E)8.5 via oral gavage followed by dissection at E12.5 and analysis by epifluorescence stereomicroscopy. We also demonstrate how to micro-dissect fate mapped domains for explant preparation or FACS analysis and dissect adult fate-mapped brains for whole mount fluorescent imaging. Collectively, these procedures allow researchers to address critical questions in developmental biology and disease models.


Assuntos
Linhagem da Célula/genética , Técnicas Genéticas , Integrases/genética , Animais , Camundongos
16.
Gene Expr Patterns ; 9(7): 475-89, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19616131

RESUMO

A long-standing problem in development is understanding how progenitor cells transiently expressing genes contribute to complex anatomical and functional structures. In the developing nervous system an additional level of complexity arises when considering how cells of distinct lineages relate to newly established neural circuits. To address these problems, we used both cumulative marking with Cre/loxP and Genetic Inducible Fate Mapping (GIFM), which permanently and heritably marks small populations of progenitors and their descendants with fine temporal control using CreER/loxP. A key component used in both approaches is a conditional phenotyping allele that has the potential to be expressed in all cell types, but is quiescent because of a loxP flanked Stop sequence, which precedes a reporter allele. Upon recombination, the resulting phenotyping allele is 'turned on' and then constitutively expressed. Thus, the reporter functions as a high fidelity genetic lineage tracer in vivo. Currently there is an array of reporter alleles that can be used in marking strategies, but their recombination efficiency and applicability to a wide array of tissues has not been thoroughly described. To assess the recombination/marking potential of the reporters, we utilized CreER(T) under the control of a Wnt1 transgene (Wnt1-CreER(T)) as well as a cumulative, non-inducible En1(Cre) knock-in line in combination with three different reporters: R26R (LacZ reporter), Z/EG (EGFP reporter), and Tau-Lox-STOP-Lox-mGFP-IRES-NLS-LacZ (membrane-targeted GFP/nuclear LacZ reporter). We marked the Wnt1 lineage using each of the three reporters at embryonic day (E) 8.5 followed by analysis at E10.0, E12.5, and in the adult. We also compared cumulative marking of cells with a history of En1 expression at the same stages. We evaluated the reporters by whole-mount and section analysis and ascertained the strengths and weaknesses of each of the reporters. Comparative analysis with the reporters elucidated complexities of how the Wnt1 and En1 lineages contribute to developing embryos and to axonal projection patterns of neurons derived from these lineages.


Assuntos
Alelos , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sistema Nervoso/embriologia , Animais , Linhagem da Célula , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Genes Reporter , Integrases/genética , Integrases/metabolismo , Óperon Lac/genética , Camundongos , Camundongos Transgênicos , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , Células-Tronco/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
17.
Dev Dyn ; 235(9): 2376-85, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16871622

RESUMO

A fascinating aspect of developmental biology is how organs are assembled in three dimensions over time. Fundamental to understanding organogenesis is the ability to determine when and where specific cell types are generated, the lineage of each cell, and how cells move to reside in their final position. Numerous methods have been developed to mark and follow the fate of cells in various model organisms used by developmental biologists, but most are not readily applicable to mouse embryos in utero because they involve physical marking of cells through injection of tracers. The advent of sophisticated transgenic and gene targeting techniques, combined with the use of site-specific recombinases, has revolutionized fate mapping studies in mouse. Furthermore, using genetic fate mapping to mark cells has opened up the possibility of addressing fundamental questions that cannot be studied with traditional methods of fate mapping in other organisms. Specifically, genetic fate mapping allows both the relationship between embryonic gene expression and cell fate (genetic lineage) to be determined, as well as the link between gene expression domains and anatomy (genetic anatomy) to be established. In this review, we present the ever-evolving development of genetic fate mapping techniques in mouse, especially the recent advance of Genetic Inducible Fate Mapping. We then review recent studies in the nervous system (focusing on the anterior hindbrain) as well as in the limb and with adult stem cells to highlight fundamental developmental processes that can be discovered using genetic fate mapping approaches. We end with a look toward the future at a powerful new approach that combines genetic fate mapping with cellular phenotyping alleles to study cell morphology, physiology, and function using examples from the nervous system.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sistema Nervoso/embriologia , Animais , Padronização Corporal/genética , Feminino , Técnicas Genéticas , Camundongos , Sistema Nervoso/metabolismo , Neurônios/citologia , Gravidez , Rombencéfalo/citologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo
18.
Curr Top Dev Biol ; 69: 101-38, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16243598

RESUMO

The brain is a remarkably complex anatomical structure that contains a diverse array of subdivisions, cell types, and synaptic connections. It is equally extraordinary in its physiological properties, as it constantly evaluates and integrates external stimuli as well as controls a complicated internal environment. The brain can be divided into three primary broad regions: the forebrain, midbrain (Mb), and hindbrain (Hb), each of which contain further subdivisions. The regions considered in this chapter are the Mb and most-anterior Hb (Mb/aHb), which are derived from the mesencephalon (mes) and rhombomere 1 (r1), respectively. The dorsal Mb consists of the laminated superior colliculus and the globular inferior colliculus (Fig. 1A and B), which modulate visual and auditory stimuli, respectively. The dorsal component of the aHb is the highly foliated cerebellum (Cb), which is primarily attributed to controlling motor skills (Fig. 1A and B). In contrast, the ventral Mb/aHb (Fig. 1B) consists of distinct clusters of neurons that together comprise a network of nuclei and projections-notably, the Mb dopaminergic and Hb serotonergic and Mb/aHb cholinergic neurons (Fig. 1G and H), which modulate a collection of behaviors, including movement, arousal, feeding, wakefulness, and emotion. Historically, the dorsal Mb and Cb have been studied using the chick as a model system because of the ease of performing both cell labeling and tissue transplants in the embryo in ovo; currently DNA electroporation techniques are also used. More recently the mouse has emerged as a powerful genetic system with numerous advantages to study events underpinning Mb/aHb development. There is a diverse array of spontaneous mutants with both Mb- and Cb-related phenotypes. In addition, numerous gene functions have been enumerated in mouse, gene expression is similar across vertebrates, and powerful genetic tools have been developed. Finally, additional insight into Mb/aHb function has been gained from studies of genetic diseases, such as Parkinson's disease, schizophrenia, cancer, and Dandy Walker syndrome, that afflict the Mb/aHb in humans and have genetic counterparts in mouse. Accordingly, this chapter discusses a spectrum of experiments, including classic embryology, in vitro assays, sophisticated genetic methods, and human diseases. We begin with an overview of Mb and aHb anatomy and physiology and mes/r1 gene expression patterns. We then provide a summary of fate-mapping studies that collectively demonstrate the complex cell behaviors that occur while the Mb and aHb primordia are established during embryogenesis and discuss the integration of both anterior-posterior (A-P) and dorsal-ventral (D-V) patterning. Finally, we describe some aspects of postnatal development and some of the insights gained from human diseases.


Assuntos
Encefalopatias/genética , Cerebelo/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Mesencéfalo/embriologia , Recombinação Genética , Animais , Cerebelo/metabolismo , Humanos , Mesencéfalo/metabolismo
19.
J Neurosci Res ; 82(1): 83-92, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16118800

RESUMO

Microtubule-associated protein (MAP)1B-heterozygous (MAP1B+/-) mice are deficient in the expression of MAP1B in the hippocampus, cerebellum, and olfactory cortex. Although MAP1B+/- mice showed half the normal levels of MAP1B protein, they had no measurable amounts of phosphorylated MAP1B. High-frequency theta burst stimulation of Schaffer collateral-CA1 axons in hippocampal slices from MAP1B+/- mice elicited long-term potentiation (LTP) that decayed rapidly to baseline, in contrast to the non-decremental LTP exhibited by age-matched wild-type slices. A separate group of MAP1B+/- and wild-type slices was examined for a longer time course of 3 hr post-tetanus in response to multiple high-frequency stimulus trains that induced saturated LTP. MAP1B+/- slices showed marked reductions in both immediate post-tetanic potentiation and LTP that decayed much more rapidly than that in wild-type slices. The induction of LTP was associated with a rapid dephosphorylation of MAP1B within 5-15 min post-tetanus, suggesting that the normal expression of MAP1B and conversion to a dephosphorylated state may be a cellular mediator of cytoskeletal alterations necessary for long-term activity-dependent synaptic plasticity.


Assuntos
Hipocampo/fisiopatologia , Potenciação de Longa Duração/genética , Proteínas Associadas aos Microtúbulos/deficiência , Animais , Western Blotting/métodos , Relação Dose-Resposta à Radiação , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Regulação da Expressão Gênica/genética , Hipocampo/lesões , Hipocampo/metabolismo , Hipocampo/patologia , Imuno-Histoquímica/métodos , Técnicas In Vitro , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/fisiologia , Fosforilação , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA