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










Base de datos
Intervalo de año de publicación
1.
STAR Protoc ; 4(4): 102739, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38043059

RESUMEN

Zebrafish (Danio rerio) are increasingly used to investigate spine development, growth, and for studying the etiology of spinal deformity, such as scoliosis. Here, we present a micro-computed tomography-based pipeline for visualizing the zebrafish skeleton. We describe steps for sample preparation, imaging, data management, and processing. We then detail analysis of vertebral and spine morphology using open-source software. This protocol will be useful for scientists using zebrafish to understand spine development and disease. For complete details on the use and execution of this protocol, please refer to Bearce et al. (2022).1.


Asunto(s)
Escoliosis , Animales , Escoliosis/diagnóstico por imagen , Pez Cebra , Microtomografía por Rayos X , Columna Vertebral/diagnóstico por imagen
2.
Elife ; 112022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36453722

RESUMEN

The spine provides structure and support to the body, yet how it develops its characteristic morphology as the organism grows is little understood. This is underscored by the commonality of conditions in which the spine curves abnormally such as scoliosis, kyphosis, and lordosis. Understanding the origin of these spinal curves has been challenging in part due to the lack of appropriate animal models. Recently, zebrafish have emerged as promising tools with which to understand the origin of spinal curves. Using zebrafish, we demonstrate that the urotensin II-related peptides (URPs), Urp1 and Urp2, are essential for maintaining spine morphology. Urp1 and Urp2 are 10-amino acid cyclic peptides expressed by neurons lining the central canal of the spinal cord. Upon combined genetic loss of Urp1 and Urp2, adolescent-onset planar curves manifested in the caudal region of the spine. Highly similar curves were caused by mutation of Uts2r3, an URP receptor. Quantitative comparisons revealed that urotensin-associated curves were distinct from other zebrafish spinal curve mutants in curve position and direction. Last, we found that the Reissner fiber, a proteinaceous thread that sits in the central canal and has been implicated in the control of spine morphology, breaks down prior to curve formation in mutants with perturbed cilia motility but was unaffected by loss of Uts2r3. This suggests a Reissner fiber-independent mechanism of curvature in urotensin-deficient mutants. Overall, our results show that Urp1 and Urp2 control zebrafish spine morphology and establish new animal models of spine deformity.


The backbone, or spine, is an integral part of the human body, providing support to our torsos so that we can sit, stand, bend and twist. If this structure does not form correctly, it can lead to pain, neurologic problems, and mobility issues. The spine normally has curves, but these can become deformed for many reasons, including genetic and muscular factors. There are also cases in which the cause of a spine distortion is unknown, such as in scoliosis (where the spine twists to the side), lordosis (where the lower part of the spine curves excessively), and kyphosis (where the upper part of the spine shows extreme curvature). The structure of the spine is laid out during embryonic development and maintained throughout life. Experiments in zebrafish have shown that a crucial element in preserving the shape of the spine is the flow of cerebrospinal fluid or CSF. Propelled by the movement of little 'hairs' at the surface of specialized cells, this liquid runs through our central nervous system along a cavity lined with neurons. These nerve cells produce Urp1 and Urp2, two short molecules (or peptides) built from the same components as proteins. In zebrafish embryos, lowering the levels of these peptides had previously been shown to cause early body deformities. But what role, if any, do Urp1 and Urp2 play in maintaining the shape of the spine in adult zebrafish? Bearce et al. set out to answer this question. First, they generated mutant zebrafish which did not carry either Urp1, Urp2 or both peptides. Contrary to previous findings, all three of these mutants developed normally as embryos. Once they were adults, zebrafish lacking Urp1 exhibited normal spines, while those lacking Urp2 had slightly deformed curves. However, zebrafish lacking both peptides had prominent curves in the tail-region of their spines, somewhat akin to lordosis in humans. This indicates that both peptides are necessary for adult spine structure, but work in a semi-redundant manner. Interestingly, the defects observed first appeared in adolescent fish and gradually worsened as they grew; many forms of human spinal abnormalities follow a similar trajectory. Bearce et al. also tested the role of the protein Uts2r3, a receptor for peptides which belong to the urotensin family (such as Urp1 and Urp2). Fish lacking this protein showed normal spine structure as embryos, but distorted spinal curves as adults, suggesting that Urp1 and Urp2 might control spine morphology by signaling via the Uts2r3 receptor. Together, Bearce et al.'s observations show that disturbing urotensin signaling leads to a lordosis-like condition in adult zebrafish, with evident deformities in the tail-region of the spine. Considering the broad similarities in structures between the zebrafish and the human spine, these results point to a possible involvement of urotensin signaling in spine distortion in humans. More studies using zebrafish will likely provide further insights into the principles that control the shape of the spine and what goes wrong when it breaks down.


Asunto(s)
Escoliosis , Urotensinas , Animales , Urotensinas/genética , Pez Cebra/genética , Columna Vertebral
3.
Genet Med ; 24(11): 2249-2261, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36074124

RESUMEN

PURPOSE: The clinical spectrum of motile ciliopathies includes laterality defects, hydrocephalus, and infertility as well as primary ciliary dyskinesia when impaired mucociliary clearance results in otosinopulmonary disease. Importantly, approximately 30% of patients with primary ciliary dyskinesia lack a genetic diagnosis. METHODS: Clinical, genomic, biochemical, and functional studies were performed alongside in vivo modeling of DAW1 variants. RESULTS: In this study, we identified biallelic DAW1 variants associated with laterality defects and respiratory symptoms compatible with motile cilia dysfunction. In early mouse embryos, we showed that Daw1 expression is limited to distal, motile ciliated cells of the node, consistent with a role in left-right patterning. daw1 mutant zebrafish exhibited reduced cilia motility and left-right patterning defects, including cardiac looping abnormalities. Importantly, these defects were rescued by wild-type, but not mutant daw1, gene expression. In addition, pathogenic DAW1 missense variants displayed reduced protein stability, whereas DAW1 loss-of-function was associated with distal type 2 outer dynein arm assembly defects involving axonemal respiratory cilia proteins, explaining the reduced cilia-induced fluid flow in particle tracking velocimetry experiments. CONCLUSION: Our data define biallelic DAW1 variants as a cause of human motile ciliopathy and determine that the disease mechanism involves motile cilia dysfunction, explaining the ciliary beating defects observed in affected individuals.


Asunto(s)
Trastornos de la Motilidad Ciliar , Ciliopatías , Proteínas del Citoesqueleto , Animales , Humanos , Ratones , Axonema/genética , Cilios/metabolismo , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Proteínas del Citoesqueleto/genética , Mutación , Proteínas/genética , Pez Cebra/genética
4.
Development ; 149(12)2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35708608

RESUMEN

Motile cilia generate cell propulsion and extracellular fluid flows that are crucial for airway clearance, fertility and left-right patterning. Motility is powered by dynein arm complexes that are assembled in the cytoplasm then imported into the cilium. Studies in Chlamydomonas reinhardtii showed that ODA16 is a cofactor which promotes dynein arm import. Here, we demonstrate that the zebrafish homolog of ODA16, Daw1, facilitates the onset of robust cilia motility during development. Without Daw1, cilia showed markedly reduced motility during early development; however, motility subsequently increased to attain close to wild-type levels. Delayed motility onset led to differential effects on early and late cilia-dependent processes. Remarkably, abnormal body axis curves, which formed during the first day of development due to reduced cilia motility, self-corrected when motility later reached wild-type levels. Zebrafish larva therefore possess the ability to survey and correct body shape abnormalities. This work defines Daw1 as a factor which promotes the onset of timely cilia motility and can explain why human patients harboring DAW1 mutations exhibit significant laterality perturbations but mild airway and fertility complications.


Asunto(s)
Cilios , Dineínas , Animales , Movimiento Celular , Cilios/metabolismo , Dineínas/metabolismo , Humanos , Mutación/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Semin Cell Dev Biol ; 110: 104-112, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32693941

RESUMEN

How developing and growing organisms attain their proper shape is a central problem of developmental biology. In this review, we investigate this question with respect to how the body axis and spine form in their characteristic linear head-to-tail fashion in vertebrates. Recent work in the zebrafish has implicated motile cilia and cerebrospinal fluid flow in axial morphogenesis and spinal straightness. We begin by introducing motile cilia, the fluid flows they generate and their roles in zebrafish development and growth. We then describe how cilia control body and spine shape through sensory cells in the spinal canal, a thread-like extracellular structure called the Reissner fiber, and expression of neuropeptide signals. Last, we discuss zebrafish mutants in which spinal straightness breaks down and three-dimensional curves form. These curves resemble the common but little-understood human disease Idiopathic Scoliosis. Zebrafish research is therefore poised to make progress in our understanding of this condition and, more generally, how body and spine shape is acquired and maintained through development and growth.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Cilios/metabolismo , Proteínas del Citoesqueleto/genética , Morfogénesis/genética , Escoliosis/genética , Columna Vertebral/metabolismo , Proteínas de Pez Cebra/genética , Animales , Axonema/metabolismo , Axonema/ultraestructura , Moléculas de Adhesión Celular Neuronal/deficiencia , Líquido Cefalorraquídeo/química , Cilios/patología , Cilios/ultraestructura , Proteínas del Citoesqueleto/deficiencia , Modelos Animales de Enfermedad , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Humanos , Mutación , Escoliosis/metabolismo , Escoliosis/patología , Transducción de Señal , Columna Vertebral/anomalías , Columna Vertebral/crecimiento & desarrollo , Urotensinas/genética , Pez Cebra , Proteínas de Pez Cebra/deficiencia
6.
Dev Biol ; 459(2): 79-86, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31758943

RESUMEN

Building a left-right (L-R) asymmetric organ requires asymmetric information. This comes from various sources, including asymmetries in embryo-scale genetic cascades (including the left-sided Nodal cascade), organ-intrinsic mechanical forces, and cell-level chirality, but the relative influence of these sources and how they collaborate to drive asymmetric morphogenesis is not understood. During zebrafish heart development, the linear heart tube extends to the left of the midline in a process known as jogging. The jogged heart then undergoes dextral (i.e. rightward) looping to correctly position the heart chambers relative to one another. Left lateralized jogging is governed by the left-sided expression of Nodal in mesoderm tissue, while looping laterality is mainly controlled by heart-intrinsic cell-level asymmetries in the actomyosin cytoskeleton. The purpose of lateralized jogging is not known. Moreover, after jogging, the heart tube returns to an almost midline position and so it is not clear whether or how jogging may impact the dextral loop. Here, we characterize a novel loss-of-function mutant in the zebrafish Nodal homolog southpaw (spaw) that appears to be a true null. We then assess the relationship between jogging and looping laterality in embryos lacking asymmetric Spaw signals. We found that the probability of a dextral loop occurring, does not depend on asymmetric Spaw signals per se, but does depend on the laterality of jogging. Thus, we conclude that the role of leftward jogging is to spatially position the heart tube in a manner that promotes robust dextral looping. When jogging laterality is abnormal, the robustness of dextral looping decreases. This establishes a cooperation between embryo-scale Nodal-dependent L-R asymmetries and organ-intrinsic cellular chirality in the control of asymmetric heart morphogenesis and shows that the transient laterality of the early heart tube has consequences for later heart morphogenetic events.


Asunto(s)
Tipificación del Cuerpo/genética , Desarrollo Embrionario/genética , Corazón/embriología , Organogénesis/genética , Pez Cebra/embriología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Mutación con Pérdida de Función , Masculino , Mesodermo/metabolismo , Miocardio/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Nat Commun ; 10(1): 4072, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492868

RESUMEN

The human PKD2 locus encodes Polycystin-2 (PC2), a TRPP channel that localises to several distinct cellular compartments, including the cilium. PKD2 mutations cause Autosomal Dominant Polycystic Kidney Disease (ADPKD) and affect many cellular pathways. Data underlining the importance of ciliary PC2 localisation in preventing PKD are limited because PC2 function is ablated throughout the cell in existing model systems. Here, we dissect the ciliary role of PC2 by analysing mice carrying a non-ciliary localising, yet channel-functional, PC2 mutation. Mutants develop embryonic renal cysts that appear indistinguishable from mice completely lacking PC2. Despite not entering the cilium in mutant cells, mutant PC2 accumulates at the ciliary base, forming a ring pattern consistent with distal appendage localisation. This suggests a two-step model of ciliary entry; PC2 first traffics to the cilium base before TOP domain dependent entry. Our results suggest that PC2 localisation to the cilium is necessary to prevent PKD.


Asunto(s)
Cilios/metabolismo , Riñón/patología , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/metabolismo , Glicosilación , Humanos , Riñón/embriología , Masculino , Ratones Endogámicos C57BL , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPP/genética
8.
Development ; 146(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31416929

RESUMEN

Consistent asymmetries between the left and right sides of animal bodies are common. For example, the internal organs of vertebrates are left-right (L-R) asymmetric in a stereotyped fashion. Other structures, such as the skeleton and muscles, are largely symmetric. This Review considers how symmetries and asymmetries form alongside each other within the embryo, and how they are then maintained during growth. I describe how asymmetric signals are generated in the embryo. Using the limbs and somites as major examples, I then address mechanisms for protecting symmetrically forming tissues from asymmetrically acting signals. These examples reveal that symmetry should not be considered as an inherent background state, but instead must be actively maintained throughout multiple phases of embryonic patterning and organismal growth.


Asunto(s)
Tipificación del Cuerpo , Desarrollo Embrionario , Animales , Enfermedad , Extremidades/embriología , Humanos , Ratones , Somitos/embriología
9.
Curr Biol ; 29(3): R101-R103, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30721676

RESUMEN

It has long been noticed that zebrafish defective in ciliary beating develop abnormal body curvatures. Recently, insights into how cilia keep the body straight have emerged, with implications for understanding human scoliosis.


Asunto(s)
Cilios , Proteínas de Pez Cebra , Animales , Biología Evolutiva , Humanos , Morfogénesis , Pez Cebra
10.
Proc Natl Acad Sci U S A ; 115(21): 5474-5479, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735715

RESUMEN

Mammalian sex determination is controlled by the antagonistic interactions of two genetic pathways: The SRY-SOX9-FGF9 network promotes testis determination partly by opposing proovarian pathways, while RSPO1/WNT-ß-catenin/FOXL2 signals control ovary development by inhibiting SRY-SOX9-FGF9. The molecular basis of this mutual antagonism is unclear. Here we show that ZNRF3, a WNT signaling antagonist and direct target of RSPO1-mediated inhibition, is required for sex determination in mice. XY mice lacking ZNRF3 exhibit complete or partial gonadal sex reversal, or related defects. These abnormalities are associated with ectopic WNT/ß-catenin activity and reduced Sox9 expression during fetal sex determination. Using exome sequencing of individuals with 46,XY disorders of sex development, we identified three human ZNRF3 variants in very rare cases of XY female presentation. We tested two missense variants and show that these disrupt ZNRF3 activity in both human cell lines and zebrafish embryo assays. Our data identify a testis-determining function for ZNRF3 and indicate a mechanism of direct molecular interaction between two mutually antagonistic organogenetic pathways.


Asunto(s)
Trastornos del Desarrollo Sexual/genética , Diferenciación Sexual , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Adolescente , Adulto , Animales , Células Cultivadas , Trastornos del Desarrollo Sexual/patología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Gónadas/patología , Humanos , Masculino , Ratones , Mutación Missense , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Testículo/metabolismo , Testículo/patología , Trombospondinas/genética , Trombospondinas/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Adulto Joven , Pez Cebra , beta Catenina/genética , beta Catenina/metabolismo
11.
Trends Genet ; 33(9): 616-628, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28720483

RESUMEN

Vertebrates exhibit striking left-right (L-R) asymmetries in the structure and position of the internal organs. Symmetry is broken by motile cilia-generated asymmetric fluid flow, resulting in a signaling cascade - the Nodal-Pitx2 pathway - being robustly established within mesodermal tissue on the left side only. This pathway impinges upon various organ primordia to instruct their side-specific development. Recently, progress has been made in understanding both the breaking of embryonic L-R symmetry and how the Nodal-Pitx2 pathway controls lateralized cell differentiation, migration, and other aspects of cell behavior, as well as tissue-level mechanisms, that drive asymmetries in organ formation. Proper execution of asymmetric organogenesis is critical to health, making furthering our understanding of L-R development an important concern.


Asunto(s)
Tipificación del Cuerpo , Animales , Morfogénesis
12.
Curr Top Dev Biol ; 124: 1-40, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28335857

RESUMEN

Cardiac development is a dynamic process regulated by spatial and temporal cues that are integrated to effect molecular, cellular, and tissue-level events that form the adult heart. Disruption of these highly orchestrated events can be devastating for cardiac form and function. Aberrations in heart development result in congenital heart defects (CHDs), which affect 1 in 100 infants in the United States each year. Zebrafish have proven informative as a model organism to understand both heart development and the mechanisms associated with CHDs due to the similarities in heart morphogenesis among vertebrates, as well as their genetic tractability and amenability to live imaging. In this review, we discuss the mechanisms of zebrafish heart development and the utility of zebrafish for understanding syndromic CHDs, those cardiac abnormalities that occur in the context of multisystem disorders. We conclude with avenues of zebrafish research that will potentially inform future therapeutic approaches for the treatment of CHDs.


Asunto(s)
Modelos Animales de Enfermedad , Cardiopatías Congénitas/patología , Pez Cebra/fisiología , Animales , Corazón/embriología , Humanos , Modelos Biológicos , Síndrome
13.
Artículo en Inglés | MEDLINE | ID: mdl-27821532

RESUMEN

Left-right (L-R) asymmetry of the internal organs of vertebrates is presaged by domains of asymmetric gene expression in the lateral plate mesoderm (LPM) during somitogenesis. Ciliated L-R coordinators (LRCs) are critical for biasing the initiation of asymmetrically expressed genes, such as nodal and pitx2, to the left LPM. Other midline structures, including the notochord and floorplate, are then required to maintain these asymmetries. Here we report an unexpected role for the zebrafish EGF-CFC gene one-eyed pinhead (oep) in the midline to promote pitx2 expression in the LPM. Late zygotic oep (LZoep) mutants have strongly reduced or absent pitx2 expression in the LPM, but this expression can be rescued to strong levels by restoring oep in midline structures only. Furthermore, removing midline structures from LZoep embryos can rescue pitx2 expression in the LPM, suggesting the midline is a source of an LPM pitx2 repressor that is itself inhibited by oep Reducing lefty1 activity in LZoep embryos mimics removal of the midline, implicating lefty1 in the midline-derived repression. Together, this suggests a model where Oep in the midline functions to overcome a midline-derived repressor, involving lefty1, to allow for the expression of left side-specific genes in the LPM.This article is part of the themed issue 'Provocative questions in left-right asymmetry'.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Mesodermo/embriología , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo
14.
PLoS Genet ; 12(6): e1006070, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27272319

RESUMEN

During mammalian development, left-right (L-R) asymmetry is established by a cilia-driven leftward fluid flow within a midline embryonic cavity called the node. This 'nodal flow' is detected by peripherally-located crown cells that each assemble a primary cilium which contain the putative Ca2+ channel PKD2. The interaction of flow and crown cell cilia promotes left side-specific expression of Nodal in the lateral plate mesoderm (LPM). Whilst the PKD2-interacting protein PKD1L1 has also been implicated in L-R patterning, the underlying mechanism by which flow is detected and the genetic relationship between Polycystin function and asymmetric gene expression remains unknown. Here, we characterize a Pkd1l1 mutant line in which Nodal is activated bilaterally, suggesting that PKD1L1 is not required for LPM Nodal pathway activation per se, but rather to restrict Nodal to the left side downstream of nodal flow. Epistasis analysis shows that Pkd1l1 acts as an upstream genetic repressor of Pkd2. This study therefore provides a genetic pathway for the early stages of L-R determination. Moreover, using a system in which cultured cells are supplied artificial flow, we demonstrate that PKD1L1 is sufficient to mediate a Ca2+ signaling response after flow stimulation. Finally, we show that an extracellular PKD domain within PKD1L1 is crucial for PKD1L1 function; as such, destabilizing the domain causes L-R defects in the mouse. Our demonstration that PKD1L1 protein can mediate a response to flow coheres with a mechanosensation model of flow sensation in which the force of fluid flow drives asymmetric gene expression in the embryo.


Asunto(s)
Tipificación del Cuerpo/genética , Cilios/genética , Proteínas de la Membrana/genética , Mesodermo/metabolismo , Proteína Nodal/genética , Canales Catiónicos TRPP/genética , Animales , Embrión de Mamíferos/citología , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Mesodermo/embriología , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Proteína Nodal/biosíntesis , Estructura Terciaria de Proteína , Canales Catiónicos TRPP/antagonistas & inhibidores
15.
Cell Rep ; 14(8): 1841-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26904945

RESUMEN

Cilia are microtubule-based projections that function in the movement of extracellular fluid. This requires cilia to be: (1) motile and driven by dynein complexes and (2) correctly polarized on the surface of cells, which requires planar cell polarity (PCP). Few factors that regulate both processes have been discovered. We reveal that C21orf59/Kurly (Kur), a cytoplasmic protein with some enrichment at the base of cilia, is needed for motility; zebrafish mutants exhibit characteristic developmental abnormalities and dynein arm defects. kur was also required for proper cilia polarization in the zebrafish kidney and the larval skin of Xenopus laevis. CRISPR/Cas9 coupled with homologous recombination to disrupt the endogenous kur locus in Xenopus resulted in the asymmetric localization of the PCP protein Prickle2 being lost in mutant multiciliated cells. Kur also makes interactions with other PCP components, including Disheveled. This supports a model wherein Kur plays a dual role in cilia motility and polarization.


Asunto(s)
Proteínas con Dominio LIM/genética , Microtúbulos/metabolismo , Xenopus laevis/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Sitios de Unión , Sistemas CRISPR-Cas , Movimiento Celular , Polaridad Celular , Cilios/metabolismo , Proteínas Dishevelled/genética , Proteínas Dishevelled/metabolismo , Embrión no Mamífero , Expresión Génica , Sitios Genéticos , Recombinación Homóloga , Riñón/citología , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Proteínas con Dominio LIM/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Proteínas de la Membrana , Microtúbulos/ultraestructura , Mutación , Unión Proteica , Transducción de Señal , Piel/citología , Piel/crecimiento & desarrollo , Piel/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
16.
Development ; 141(20): 3966-77, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25294941

RESUMEN

Initially identified in DNA damage repair, ATM-interactor (ATMIN) further functions as a transcriptional regulator of lung morphogenesis. Here we analyse three mouse mutants, Atmin(gpg6/gpg6), Atmin(H210Q/H210Q) and Dynll1(GT/GT), revealing how ATMIN and its transcriptional target dynein light chain LC8-type 1 (DYNLL1) are required for normal lung morphogenesis and ciliogenesis. Expression screening of ciliogenic genes confirmed Dynll1 to be controlled by ATMIN and further revealed moderately altered expression of known intraflagellar transport (IFT) protein-encoding loci in Atmin mutant embryos. Significantly, Dynll1(GT/GT) embryonic cilia exhibited shortening and bulging, highly similar to the characterised retrograde IFT phenotype of Dync2h1. Depletion of ATMIN or DYNLL1 in cultured cells recapitulated the in vivo ciliogenesis phenotypes and expression of DYNLL1 or the related DYNLL2 rescued the effects of loss of ATMIN, demonstrating that ATMIN primarily promotes ciliogenesis by regulating Dynll1 expression. Furthermore, DYNLL1 as well as DYNLL2 localised to cilia in puncta, consistent with IFT particles, and physically interacted with WDR34, a mammalian homologue of the Chlamydomonas cytoplasmic dynein 2 intermediate chain that also localised to the cilium. This study extends the established Atmin-Dynll1 relationship into a developmental and a ciliary context, uncovering a novel series of interactions between DYNLL1, WDR34 and ATMIN. This identifies potential novel components of cytoplasmic dynein 2 and furthermore provides fresh insights into the molecular pathogenesis of human skeletal ciliopathies.


Asunto(s)
Cilios/fisiología , Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , Factores de Transcripción/fisiología , Animales , Chlamydomonas/metabolismo , Cilios/metabolismo , Dineínas Citoplasmáticas , Daño del ADN , Dineínas/metabolismo , Marcadores Genéticos , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Mutación , Fenotipo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
18.
Dis Model Mech ; 5(3): 299-312, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22566558

RESUMEN

The ciliopathies are an apparently disparate group of human diseases that all result from defects in the formation and/or function of cilia. They include disorders such as Meckel-Grüber syndrome (MKS), Joubert syndrome (JBTS), Bardet-Biedl syndrome (BBS) and Alström syndrome (ALS). Reflecting the manifold requirements for cilia in signalling, sensation and motility, different ciliopathies exhibit common elements. The mouse has been used widely as a model organism for the study of ciliopathies. Although many mutant alleles have proved lethal, continued investigations have led to the development of better models. Here, we review current mouse models of a core set of ciliopathies, their utility and future prospects.


Asunto(s)
Anomalías Múltiples/patología , Cilios/patología , Modelos Animales de Enfermedad , Anomalías Múltiples/metabolismo , Animales , Cilios/metabolismo , Humanos , Ratones , Órganos de los Sentidos/metabolismo , Órganos de los Sentidos/patología , Transducción de Señal , Investigación Biomédica Traslacional
19.
Development ; 138(6): 1131-42, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21307093

RESUMEN

In mammals, left-right (L-R) asymmetry is established by posteriorly oriented cilia driving a leftwards laminar flow in the embryonic node, thereby activating asymmetric gene expression. The two-cilia hypothesis argues that immotile cilia detect and respond to this flow through a Pkd2-mediated mechanism; a putative sensory partner protein has, however, remained unidentified. We have identified the Pkd1-related locus Pkd1l1 as a crucial component of L-R patterning in mouse. Systematic comparison of Pkd1l1 and Pkd2 point mutants reveals strong phenocopying, evidenced by both morphological and molecular markers of sidedness; both mutants fail to activate asymmetric gene expression at the node or in the lateral plate and exhibit right isomerism of the lungs. Node and cilia morphology were normal in mutants and cilia demonstrated typical motility, consistent with Pkd1l1 and Pkd2 activity downstream of nodal flow. Cell biological analysis reveals that Pkd1l1 and Pkd2 localise to the cilium and biochemical experiments demonstrate that they can physically interact. Together with co-expression in the node, these data argue that Pkd1l1 is the elusive Pkd2 binding partner required for L-R patterning and support the two-cilia hypothesis.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de la Membrana/fisiología , Canales Catiónicos TRPP/metabolismo , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo/fisiología , Células Cultivadas , Cilios/genética , Cilios/metabolismo , Cilios/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Polimorfismo de Nucleótido Simple/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Homología de Secuencia de Aminoácido , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/fisiología
20.
Hum Mol Genet ; 18(10): 1719-39, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19223390

RESUMEN

The mammalian Sonic hedgehog (Shh) signalling pathway is essential for embryonic development and the patterning of multiple organs. Disruption or activation of Shh signalling leads to multiple birth defects, including holoprosencephaly, neural tube defects and polydactyly, and in adults results in tumours of the skin or central nervous system. Genetic approaches with model organisms continue to identify novel components of the pathway, including key molecules that function as positive or negative regulators of Shh signalling. Data presented here define Tulp3 as a novel negative regulator of the Shh pathway. We have identified a new mouse mutant that is a strongly hypomorphic allele of Tulp3 and which exhibits expansion of ventral markers in the caudal spinal cord, as well as neural tube defects and preaxial polydactyly, consistent with increased Shh signalling. We demonstrate that Tulp3 acts genetically downstream of Shh and Smoothened (Smo) in neural tube patterning and exhibits a genetic interaction with Gli3 in limb development. We show that Tulp3 does not appear to alter expression or processing of Gli3, and we demonstrate that transcriptional regulation of other negative regulators (Rab23, Fkbp8, Thm1, Sufu and PKA) is not affected. We discuss the possible mechanism of action of Tulp3 in Shh-mediated signalling in light of these new data.


Asunto(s)
Tipificación del Cuerpo , Regulación hacia Abajo , Proteínas Hedgehog/metabolismo , Polidactilia/metabolismo , Proteínas/metabolismo , Transducción de Señal , Disrafia Espinal/metabolismo , Animales , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutación , Tubo Neural/embriología , Tubo Neural/metabolismo , Polidactilia/embriología , Polidactilia/genética , Proteínas/genética , Médula Espinal/embriología , Médula Espinal/metabolismo , Disrafia Espinal/embriología , Disrafia Espinal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...