Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Proc Natl Acad Sci U S A ; 120(16): e2214997120, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37043537

RESUMO

While somatic variants of TRAF7 (Tumor necrosis factor receptor-associated factor 7) underlie anterior skull-base meningiomas, here we report the inherited mutations of TRAF7 that cause congenital heart defects. We show that TRAF7 mutants operate in a dominant manner, inhibiting protein function via heterodimerization with wild-type protein. Further, the shared genetics of the two disparate pathologies can be traced to the common origin of forebrain meninges and cardiac outflow tract from the TRAF7-expressing neural crest. Somatic and inherited mutations disrupt TRAF7-IFT57 interactions leading to cilia degradation. TRAF7-mutant meningioma primary cultures lack cilia, and TRAF7 knockdown causes cardiac, craniofacial, and ciliary defects in Xenopus and zebrafish, suggesting a mechanistic convergence for TRAF7-driven meningiomas and developmental heart defects.


Assuntos
Cardiopatias Congênitas , Neoplasias Meníngeas , Meningioma , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cardiopatias Congênitas/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Meningioma/patologia , Mutação , Crânio/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Humanos , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral
3.
Science ; 379(6627): 71-78, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36603098

RESUMO

The breaking of bilateral symmetry in most vertebrates is critically dependent upon the motile cilia of the embryonic left-right organizer (LRO), which generate a directional fluid flow; however, it remains unclear how this flow is sensed. Here, we demonstrated that immotile LRO cilia are mechanosensors for shear force using a methodological pipeline that combines optical tweezers, light sheet microscopy, and deep learning to permit in vivo analyses in zebrafish. Mechanical manipulation of immotile LRO cilia activated intraciliary calcium transients that required the cation channel Polycystin-2. Furthermore, mechanical force applied to LRO cilia was sufficient to rescue and reverse cardiac situs in zebrafish that lack motile cilia. Thus, LRO cilia are mechanosensitive cellular levers that convert biomechanical forces into calcium signals to instruct left-right asymmetry.


Assuntos
Padronização Corporal , Sinalização do Cálcio , Cálcio , Cílios , Peixe-Zebra , Animais , Cálcio/metabolismo , Cílios/fisiologia , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/metabolismo , Canais de Cátion TRPP/metabolismo
4.
Front Cardiovasc Med ; 9: 1037500, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36451924

RESUMO

Molecular phenotyping by imaging of intact tissues has been used to reveal 3D molecular and structural coherence in tissue samples using tissue clearing techniques. However, clearing and imaging of cardiac tissue remains challenging for large-scale (>100 mm3) specimens due to sample distortion. Thus, directly assessing tissue microstructural geometric properties confounded by distortion such as cardiac helicity has been limited. To combat sample distortion, we developed a passive CLARITY technique (Pocket CLARITY) that utilizes a permeable cotton mesh pocket to encapsulate the sample to clear large-scale cardiac swine samples with minimal tissue deformation and protein loss. Combined with light sheet auto-fluorescent and scattering microscopy, Pocket CLARITY enabled the characterization of myocardial microstructural helicity of cardiac tissue from control, heart failure, and myocardial infarction in swine. Pocket CLARITY revealed with high fidelity that transmural microstructural helicity of the heart is significantly depressed in cardiovascular disease (CVD), thereby revealing new insights at the tissue level associated with impaired cardiac function.

5.
Circ Res ; 131(12): 980-1000, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36367103

RESUMO

BACKGROUND: RBPs (RNA-binding proteins) perform indispensable functions in the post-transcriptional regulation of gene expression. Numerous RBPs have been implicated in cardiac development or physiology based on gene knockout studies and the identification of pathogenic RBP gene mutations in monogenic heart disorders. The discovery and characterization of additional RBPs performing indispensable functions in the heart will advance basic and translational cardiovascular research. METHODS: We performed a differential expression screen in zebrafish embryos to identify genes enriched in nkx2.5-positive cardiomyocytes or cardiopharyngeal progenitors compared to nkx2.5-negative cells from the same embryos. We investigated the myocardial-enriched gene RNA-binding protein with multiple splicing (variants) 2 [RBPMS2)] by generating and characterizing rbpms2 knockout zebrafish and human cardiomyocytes derived from RBPMS2-deficient induced pluripotent stem cells. RESULTS: We identified 1848 genes enriched in the nkx2.5-positive population. Among the most highly enriched genes, most with well-established functions in the heart, we discovered the ohnologs rbpms2a and rbpms2b, which encode an evolutionarily conserved RBP. Rbpms2 localizes selectively to cardiomyocytes during zebrafish heart development and strong cardiomyocyte expression persists into adulthood. Rbpms2-deficient embryos suffer from early cardiac dysfunction characterized by reduced ejection fraction. The functional deficit is accompanied by myofibril disarray, altered calcium handling, and differential alternative splicing events in mutant cardiomyocytes. These phenotypes are also observed in RBPMS2-deficient human cardiomyocytes, indicative of conserved molecular and cellular function. RNA-sequencing and comparative analysis of genes mis-spliced in RBPMS2-deficient zebrafish and human cardiomyocytes uncovered a conserved network of 29 ortholog pairs that require RBPMS2 for alternative splicing regulation, including RBFOX2, SLC8A1, and MYBPC3. CONCLUSIONS: Our study identifies RBPMS2 as a conserved regulator of alternative splicing, myofibrillar organization, and calcium handling in zebrafish and human cardiomyocytes.


Assuntos
Cálcio , Miocárdio , Proteínas de Ligação a RNA , Proteínas de Peixe-Zebra , Animais , Humanos , Cálcio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Processamento de RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
6.
Nat Rev Cardiol ; 19(4): 211-227, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34862511

RESUMO

Although cardiac abnormalities have been observed in a growing class of human disorders caused by defective primary cilia, the function of cilia in the heart remains an underexplored area. The primary function of cilia in the heart was long thought to be restricted to left-right axis patterning during embryogenesis. However, new findings have revealed broad roles for cilia in congenital heart disease, valvulogenesis, myocardial fibrosis and regeneration, and mechanosensation. In this Review, we describe advances in our understanding of the mechanisms by which cilia function contributes to cardiac left-right axis development and discuss the latest findings that highlight a broader role for cilia in cardiac development. Specifically, we examine the growing line of evidence connecting cilia function to the pathogenesis of congenital heart disease. Furthermore, we also highlight research from the past 10 years demonstrating the role of cilia function in common cardiac valve disorders, including mitral valve prolapse and aortic valve disease, and describe findings that implicate cardiac cilia in mechanosensation potentially linking haemodynamic and contractile forces with genetic regulation of cardiac development and function. Finally, given the presence of cilia on cardiac fibroblasts, we also explore the potential role of cilia in fibrotic growth and summarize the evidence implicating cardiac cilia in heart regeneration.


Assuntos
Cardiomiopatias , Cardiopatias Congênitas , Doenças das Valvas Cardíacas , Cardiomiopatias/patologia , Cílios/genética , Cílios/patologia , Fibrose , Coração , Cardiopatias Congênitas/patologia , Humanos
7.
Curr Biol ; 28(9): R567-R569, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29738734

RESUMO

While a ciliated organizer generates vertebrate left-right asymmetry, most invertebrates lack an organizer and instead utilize a myosin-based mechanism. A recent study now reveals that this myosin mechanism is conserved in the vertebrate organizer and functions to regulate cilia.


Assuntos
Cílios , Miosinas , Animais , Vertebrados
8.
Development ; 144(24): 4684-4693, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29113992

RESUMO

Pontin (Ruvbl1) and Reptin (Ruvbl2) are closely related AAA ATPases. They are components of the Ruvbl1-Ruvbl2-Tah1-Pih1 (R2TP) complexes that function as co-chaperones for the assembly of multiple macromolecular protein complexes. Here, we show that Pontin is essential for cilia motility in both zebrafish and mouse and that Pontin and Reptin function cooperatively in this process. Zebrafish pontin mutants display phenotypes tightly associated with cilia defects, and cilia motility is lost in a number of ciliated tissues along with a reduction in the number of outer and inner dynein arms. Pontin protein is enriched in cytosolic puncta in ciliated cells in zebrafish embryos. In mouse testis, Pontin is essential for the stabilization of axonemal dynein intermediate chain 1 (DNAI1) and DNAI2, the first appreciated step in axonemal dynein arm assembly. Strikingly, multiple dynein arm assembly factors show structural similarities to either Tah1 or Pih1, the other two components of the R2TP complex. Based on these results, we propose that Pontin and Reptin function to facilitate dynein arm assembly in cytosolic foci enriched with R2TP-like complexes.


Assuntos
Axonema/metabolismo , DNA Helicases/genética , Proteínas Nucleares/genética , Motilidade dos Espermatozoides/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Animais , Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo , Cílios/patologia , Cílios/fisiologia , Proteínas de Choque Térmico HSP90/metabolismo , Masculino , Camundongos , Camundongos Knockout , Movimento
9.
J Am Soc Nephrol ; 28(1): 118-129, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27401686

RESUMO

Phosphoinositides, a family of phosphorylated derivatives of phosphatidylinositol (PtdIns), are tightly regulated both temporally and spatially by PtdIns phosphatases and kinases. Mutations in inositol polyphosphate 5-phosphatase E (INPP5E) cause Joubert syndrome, a human disorder associated with numerous ciliopathic defects, including renal cyst formation, linking phosphoinositides to ciliopathies. However, the molecular mechanism by which INPP5E-mediated PtdIns signaling regulates ciliogenesis and cystogenesis is unclear. Here, we utilized an in vivo vertebrate model of renal cystogenesis to show that Inpp5e enzymatic activity at the apical membrane directs apical docking of basal bodies in renal epithelia. Knockdown or knockout of inpp5e led to ciliogenesis defects and cystic kidneys in zebrafish. Furthermore, knockdown of inpp5e in embryos led to defects in cell polarity, cortical organization of F-actin, and apical segregation of PtdIns(4,5)P2 and PtdIns(3,4,5)P3 Knockdown of the ezrin gene, which encodes an ezrin/radixin/moesin (ERM) protein that crosslinks PtdIns(4,5)P2 and F-actin, phenocopied inpp5e knockdowns. Notably, overexpression of the ezrin gene rescued inpp5e morphants. Finally, treatment with the PI 3-kinase inhibitor LY294002, which decreases PtdIns(3,4,5)P3 levels, rescued the cellular, phenotypic, and renal functional defects in inpp5e-knockdown embryos. Together, our data indicate that Inpp5e functions as a key regulator of cell polarity in the renal epithelia, by inhibiting PtdIns(3,4,5)P3 and subsequently stabilizing PtdIns(4,5)P2 and recruiting Ezrin, F-actin, and basal bodies to the apical membrane, and suggest a possible novel approach for treating human ciliopathies.


Assuntos
Membrana Celular/fisiologia , Fosfatidilinositóis/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Anormalidades Múltiplas/genética , Animais , Cerebelo/anormalidades , Cílios/fisiologia , Anormalidades do Olho/genética , Humanos , Doenças Renais Císticas/genética , Monoéster Fosfórico Hidrolases/genética , Retina/anormalidades , Peixe-Zebra
10.
Methods Mol Biol ; 1454: 123-47, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27514920

RESUMO

Cilia play a key role in the determination of the left-right axis in vertebrates by generating and sensing flow of extraembryonic fluid at the left-right organizer (LRO). Perception of cilia-directed flow triggers a calcium signaling cascade which originates within the cilium itself and then is relayed into the surrounding mesendodermal tissue, thereby directing organ situs via the nodal pathway. Two types of cilia, motile and immotile, function simultaneously to coordinate and direct asymmetric intraciliary calcium signaling cues in the LRO. Here, we describe tools, reagents, and methodologies for the visualization and manipulation of both cilia types as well as intraciliary calcium signaling in the LRO of zebrafish.


Assuntos
Cálcio/metabolismo , Cílios/metabolismo , Peixe-Zebra/metabolismo , Animais , Transporte Biológico , Sinalização do Cálcio , Embrião não Mamífero , Microscopia , Microscopia de Fluorescência , Fenótipo
11.
Curr Biol ; 25(5): 556-67, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25660539

RESUMO

BACKGROUND: Bilateral symmetry during vertebrate development is broken at the left-right organizer (LRO) by ciliary motility and the resultant directional flow of extracellular fluid. However, how ciliary motility is perceived and transduced into asymmetrical intracellular signaling at the LRO remains controversial. Previous work has indicated that sensory cilia and polycystin-2 (Pkd2), a cation channel, are required for sensing ciliary motility, yet their function and the molecular mechanism linking both to left-right signaling cascades are unknown. RESULTS: Here we report novel intraciliary calcium oscillations (ICOs) at the LRO that connect ciliary sensation of ciliary motility to downstream left-right signaling. Utilizing cilia-targeted genetically encoded calcium indicators in live zebrafish embryos, we show that ICOs depend on Pkd2 and are left-biased at the LRO in response to ciliary motility. Asymmetric ICOs occur with onset of LRO ciliary motility, thus representing the earliest known LR asymmetric molecular signal. Suppression of ICOs using a cilia-targeted calcium sink reveals that they are essential for LR development. CONCLUSIONS: These findings demonstrate that intraciliary calcium initiates LR development and identify cilia as a functional ion signaling compartment connecting ciliary motility and flow to molecular LR signaling.


Assuntos
Padronização Corporal/fisiologia , Sinalização do Cálcio/fisiologia , Cílios/metabolismo , Síndrome de Heterotaxia/embriologia , Transdução de Sinais/fisiologia , Peixe-Zebra/embriologia , Fatores de Ribosilação do ADP/metabolismo , Análise de Variância , Animais , Líquido Extracelular/metabolismo , Técnicas Histológicas , Humanos , Modelos Biológicos , Ligantes da Sinalização Nodal/metabolismo , Canais de Cátion TRPP/metabolismo , Proteínas de Peixe-Zebra/metabolismo
12.
Nature ; 504(7480): 456-9, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24226769

RESUMO

Heterotaxy is a disorder of left-right body patterning, or laterality, that is associated with major congenital heart disease. The aetiology and mechanisms underlying most cases of human heterotaxy are poorly understood. In vertebrates, laterality is initiated at the embryonic left-right organizer, where motile cilia generate leftward flow that is detected by immotile sensory cilia, which transduce flow into downstream asymmetric signals. The mechanism that specifies these two cilia types remains unknown. Here we show that the N-acetylgalactosamine-type O-glycosylation enzyme GALNT11 is crucial to such determination. We previously identified GALNT11 as a candidate disease gene in a patient with heterotaxy, and now demonstrate, in Xenopus tropicalis, that galnt11 activates Notch signalling. GALNT11 O-glycosylates human NOTCH1 peptides in vitro, thereby supporting a mechanism of Notch activation either by increasing ADAM17-mediated ectodomain shedding of the Notch receptor or by modification of specific EGF repeats. We further developed a quantitative live imaging technique for Xenopus left-right organizer cilia and show that Galnt11-mediated Notch1 signalling modulates the spatial distribution and ratio of motile and immotile cilia at the left-right organizer. galnt11 or notch1 depletion increases the ratio of motile cilia at the expense of immotile cilia and produces a laterality defect reminiscent of loss of the ciliary sensor Pkd2. By contrast, Notch overexpression decreases this ratio, mimicking the ciliopathy primary ciliary dyskinesia. Together our data demonstrate that Galnt11 modifies Notch, establishing an essential balance between motile and immotile cilia at the left-right organizer to determine laterality, and reveal a novel mechanism for human heterotaxy.


Assuntos
Padronização Corporal , Cílios/fisiologia , Síndrome de Heterotaxia/genética , N-Acetilgalactosaminiltransferases/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Proteínas de Xenopus/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Sequência de Aminoácidos , Animais , Cílios/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Glicosilação , Humanos , Camundongos , Dados de Sequência Molecular , N-Acetilgalactosaminiltransferases/deficiência , N-Acetilgalactosaminiltransferases/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Receptor Notch1/química , Receptor Notch1/deficiência , Receptor Notch1/genética , Xenopus/embriologia , Xenopus/genética , Proteínas de Xenopus/deficiência , Proteínas de Xenopus/genética
13.
Annu Rev Genet ; 47: 353-76, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24016188

RESUMO

Once obscure, the cilium has come into the spotlight during the past decade. It is now clear that aside from generating locomotion by motile cilia, both motile and immotile cilia serve as signaling platforms for the cell. Through both motility and sensory functions, cilia play critical roles in development, homeostasis, and disease. To date, the cilium proteome contains more than 1,000 different proteins, and human genetics is identifying new ciliopathy genes at an increasing pace. Although assigning a function to immotile cilia was a challenge not so long ago, the myriad of signaling pathways, proteins, and biological processes associated with the cilium have now created a new obstacle: how to distill all these interactions into specific themes and mechanisms that may explain how the organelle serves to maintain organism homeostasis. Here, we review the basics of cilia biology, novel functions associated with cilia, and recent advances in cilia genetics, and on the basis of this framework, we further discuss the meaning and significance of ciliary connections.


Assuntos
Cílios/fisiologia , Anormalidades Múltiplas , Animais , Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/fisiopatologia , Movimento Celular , Doenças Cerebelares/genética , Doenças Cerebelares/fisiopatologia , Cerebelo/anormalidades , Cílios/ultraestrutura , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/fisiopatologia , Dano ao DNA , Reparo do DNA , Modelos Animais de Doenças , Encefalocele/genética , Encefalocele/fisiopatologia , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Flagelos/fisiologia , Flagelos/ultraestrutura , Síndrome de Heterotaxia/genética , Síndrome de Heterotaxia/fisiopatologia , Homeostase , Humanos , Doenças Renais Císticas/congênito , Doenças Renais Císticas/genética , Doenças Renais Císticas/fisiopatologia , Proteínas Motores Moleculares/fisiologia , Sistema Nervoso/citologia , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/fisiopatologia , Retina/anormalidades , Retina/fisiopatologia , Retinite Pigmentosa
14.
Proc Natl Acad Sci U S A ; 110(31): 12697-702, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858445

RESUMO

Primary ciliary dyskinesia (PCD) is an autosomal recessive disease caused by defective cilia motility. The identified PCD genes account for about half of PCD incidences and the underlying mechanisms remain poorly understood. We demonstrate that Reptin/Ruvbl2, a protein known to be involved in epigenetic and transcriptional regulation, is essential for cilia motility in zebrafish. We further show that Reptin directly interacts with the PCD protein Lrrc6/Seahorse and this interaction is critical for the in vivo function of Lrrc6/Seahorse in zebrafish. Moreover, whereas the expression levels of multiple dynein arm components remain unchanged or become elevated, the density of axonemal dynein arms is reduced in reptin(hi2394) mutants. Furthermore, Reptin is highly enriched in the cytosol and colocalizes with Lrrc6/Seahorse. Combined, these results suggest that the Reptin-Lrrc6/Seahorse complex is involved in dynein arm formation. We also show that although the DNA damage response is induced in reptin(hi2394) mutants, it remains unchanged in cilia biogenesis mutants and lrrc6/seahorse mutants, suggesting that increased DNA damage response is not intrinsic to ciliary defects and that in vertebrate development, Reptin functions in multiple processes, both cilia specific and cilia independent.


Assuntos
Dineínas do Axonema/metabolismo , Axonema/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Dineínas do Axonema/genética , Axonema/genética , Cílios/genética , Cílios/metabolismo , Dano ao DNA , Mutação , Proteínas Nucleares/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
Curr Opin Genet Dev ; 23(3): 352-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23790954

RESUMO

Although congenital heart disease (CHD) is the most common survivable birth defect, the etiology of most CHD remains unclear. Several lines of evidence from humans and vertebrate models have supported a genetic component for CHD, yet the extreme locus heterogeneity and lack of a distinct genotype-phenotype correlation have limited causative gene discovery. However, recent advances in genomic technologies are permitting detailed evaluation of the genetic abnormalities in large cohorts of CHD patients. This has led to the identification of copy-number variation and de novo mutations together accounting for up to 15% of CHD. Further, new strategies coupling human genetics with model organisms have provided mechanistic insights into the molecular and developmental pathways underlying CHD pathogenesis, notably chromatin remodeling and ciliary signaling.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Variações do Número de Cópias de DNA/genética , Cardiopatias Congênitas/genética , Mutação/genética , Estudos de Associação Genética , Genoma Humano , Cardiopatias Congênitas/etiologia , Cardiopatias Congênitas/patologia , Humanos , Transdução de Sinais , Fatores de Transcrição/genética
16.
Methods Enzymol ; 525: 159-89, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23522470

RESUMO

The target-of-rapamycin (TOR) pathway is a major regulator of cellular growth, division, and metabolism by coupling growth and nutrient cues to fundamental processes such as translation and transcription. In vertebrate models, disruptions affecting components of the TOR pathway result in cilia size abnormalities and ciliopathic phenotypes such as renal cysts. However, the mechanism by which the TOR pathway influenced cilia length and function was unknown. Recent work from zebrafish, in conjunction with the green alga Chlamydomonas, demonstrates that TOR complex 1 (TORC1)-mediated cilia length control is achieved through translational regulation of ciliary precursors. Further, in zebrafish, aberrant TORC1 signaling leads to left-right asymmetric patterning defects due to inefficient ciliary motility and fluid flow generation in the Kupffer's vesicle, a conserved embryonic organ that establishes laterality. Together, such findings show the utility of a combinatorial zebrafish and Chlamydomonas approach to dissecting the mechanistic relationship between the cilium and signaling molecules such as TOR. Here, we describe tools, reagents, and methodologies for the manipulation of the TOR pathway and the analysis of cilia morphology and function in zebrafish and Chlamydomonas.


Assuntos
Cílios/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Cílios/fisiologia , Embrião não Mamífero/metabolismo , Flagelos/metabolismo , Flagelos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo
17.
J Mol Cell Cardiol ; 58: 199-208, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23376035

RESUMO

Mutations in PKD1 and PKD2, the genes encoding the proteins polycystin-1 (PC1) and polycystin-2 (PC2), cause autosomal dominant polycystic kidney disease (ADPKD). Although the leading cause of mortality in ADPKD is cardiovascular disease, the relationship between these conditions remains poorly understood. PC2 is an intracellular calcium channel expressed in renal epithelial cells and in cardiomyocytes, and is thus hypothesized to modulate intracellular calcium signaling and affect cardiac function. Our first aim was to study cardiac function in a zebrafish model lacking PC2 (pkd2 mutants). Next, we aimed to explore the relevance of this zebrafish model to human ADPKD by examining the Mayo Clinic's ADPKD database for an association between ADPKD and idiopathic dilated cardiomyopathy (IDCM). Pkd2 mutant zebrafish showed low cardiac output and atrioventricular block. Isolated pkd2 mutant hearts displayed impaired intracellular calcium cycling and calcium alternans. These results indicate heart failure in the pkd2 mutants. In human ADPKD patients, we found IDCM to coexist frequently with ADPKD. This association was strongest in patients with PKD2 mutations. Our results demonstrate that PC2 modulates intracellular calcium cycling, contributing to the development of heart failure. In human subjects we found an association between ADPKD and IDCM and suggest that PKD mutations contribute to the development of heart failure.


Assuntos
Cardiomiopatia Dilatada/genética , Proteínas de Transporte/genética , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Proteínas de Peixe-Zebra/genética , Animais , Cálcio/metabolismo , Sinalização do Cálcio/genética , Cardiomiopatia Dilatada/fisiopatologia , Predisposição Genética para Doença , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Podócitos/metabolismo , Podócitos/patologia , Rim Policístico Autossômico Dominante/fisiopatologia , Canais de Cátion TRPP/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
19.
Proc Natl Acad Sci U S A ; 109(6): 2021-6, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22308353

RESUMO

The cilium serves as a cellular antenna by coordinating upstream environmental cues with numerous downstream signaling processes that are indispensable for the function of the cell. This role is supported by the revelation that defects of the cilium underlie an emerging class of human disorders, termed "ciliopathies." Although mounting interest in the cilium has demonstrated the essential role that the organelle plays in vertebrate development, homeostasis, and disease pathogenesis, the mechanisms regulating cilia morphology and function remain unclear. Here, we show that the target-of-rapamycin (TOR) growth pathway modulates cilia size and function during zebrafish development. Knockdown of tuberous sclerosis complex 1a (tsc1a), which encodes an upstream inhibitor of TOR complex 1 (Torc1), increases cilia length. In contrast, treatment of embryos with rapamycin, an inhibitor of Torc1, shortens cilia length. Overexpression of ribosomal protein S6 kinase 1 (S6k1), which encodes a downstream substrate of Torc1, lengthens cilia. Furthermore, we provide evidence that TOR-mediated cilia assembly is evolutionarily conserved and that protein synthesis is essential for this regulation. Finally, we demonstrate that TOR signaling and cilia length are pivotal for a variety of downstream ciliary functions, such as cilia motility, fluid flow generation, and the establishment of left-right body asymmetry. Our findings reveal a unique role for the TOR pathway in regulating cilia size through protein synthesis and suggest that appropriate and defined lengths are necessary for proper function of the cilium.


Assuntos
Cílios/metabolismo , Biossíntese de Proteínas , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Padronização Corporal , Cílios/enzimologia , Evolução Molecular , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Movimento , Tamanho do Órgão , Reologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo , Peixe-Zebra
20.
Methods Cell Biol ; 101: 39-74, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21550439

RESUMO

The cilium, a previously little studied cell surface protrusion, has emerged as an important organelle in vertebrate cells. This tiny structure is essential for normal embryonic development, including the formation of left-right asymmetry, limb morphogenesis, and the differentiation of sensory cells. In the adult, cilia also function in a variety of processes, such as the survival of photoreceptor cells, and the homeostasis in several tissues, including the epithelia of nephric ducts. Human ciliary malfunction is associated with situs inversus, kidney cysts, polydactyly, blindness, mental retardation, obesity, and many other abnormalities. The genetic accessibility and optical transparency of the zebrafish make it an excellent vertebrate model system to study cilia biology. In this chapter, we describe the morphology and distribution of cilia in zebrafish embryonic and larval organs. We also provide essential protocols to analyze cilia formation and function.


Assuntos
Cílios/fisiologia , Peixe-Zebra/embriologia , Animais , Humanos , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...