Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 120(31): e2307898120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487087

ABSTRACT

Cells of vertebrate and invertebrate organisms express proteins specialized in membrane channel-based cell-cell communication that are absent in unicellular organisms. We recently described the prediction of some members of the large-pore channel family in kinetoplastids, consisting of proteins called unnexins, which share several structural features with innexin and pannexin proteins. Here, we demonstrated that the unnexin1 protein (Unx1) is delivered to the cell membrane, displaying a topology consisting of four transmembrane domains with C and N termini on the cytoplasmic side and form large-pore channels that are permeable to small molecules. Low extracellular Ca2+/Mg2+ levels or extracellular alkalinization, but not mechanical stretching, increases channel activity. The Unx1 channel mediates the influx of Ca2+ and does not form intercellular dye coupling between HeLa Unx1 transfected cells. Unx1 channel function was further evidenced by its ability to mediate ionic currents when expressed in Xenopus oocytes. Downregulation of Unx1 mRNA with morpholine contains Trypanosoma cruzi invasion. Phylogenetic analysis revealed the presence of Unx1 homologs in other protozoan parasites, suggesting a conserved function for these channel parasites in other protists. Our data demonstrate that Unx1 forms large-pore membrane channels, which may serve as a diffusional pathway for ions and small molecules that are likely to be metabolic substrates or waste products, and signaling autocrine and paracrine molecules that could be involved in cell invasion. As morpholinos-induced downregulation of Unx1 reduces the infectivity of trypomastigotes, the Unx1 channels might be an attractive target for developing trypanocide drugs.


Subject(s)
Protein Subunits , Phylogeny , Cell Membrane , Cytoplasm , Morpholinos
2.
Brasília; CONITEC; 2022.
Non-conventional in Portuguese | BRISA/RedTESA | ID: biblio-1443379

ABSTRACT

CONDIÇÃO CLÍNICA: A distrofia muscular de Duchenne (DMD) faz parte do espectro das distrofinopatias. As distrofinopatias são um grupo de doenças neuromusculares com herança ligada ao cromossomo X, causadas por mutações no gene que codifica a distrofina (DMD), uma proteína estrutural das células musculares. Em quadros leves das distrofinopatias pode-se notar o aumento assintomático da concentração sérica de creatina fosfoquinase (CK), cãibras musculares e mioglobinúria. Enquanto nos casos graves estão as doenças musculares progressivas, especialmente DMD. A distrofia muscular de Becker, apesar de também ser progressiva e grave, é uma forma de distrofinopatia mais branda comparada à DMD. Ela é caracterizada por fraqueza muscular esquelética de início tardio com gravidade e/ou curso variáveis. TRATAMENTO: Não há cura para a DMD, e as intervenções são baseadas na prevenção e tratamento das complicações. Atualmente, a terapia padrão geralmente é baseada em corticosteróides. Os corticosteróides são capazes de retardar o declínio da força e da função muscular, proporcionando uma redução do risco de escoliose e estabilização da função pulmonar. Pode haver benefícios para a função cardíaca, embora as evidências sejam limitadas. De acordo com uma revisão sistemática Cochrane não há consenso sobre a idade ou estágio funcional para o início do tratamento com corticosteroides sendo necessários mais estudos para determinar as condições ideias para iniciá-lo, ainda que haja autores que defendam que deveria ocorrer antes do declínio físico substancial, normalmente aos 5 anos de idade. .ESTRATÉGIA DE BUSCA: Uma busca foi realizada no banco de dados eletrônico ClinicalTrials.gov, em 14 de julho de 2021. O termo empregado foi: "Duchenne Muscular Dystrophy". Foram considerados os estudos dos medicamentos a partir da fase 1/2 de pesquisa clínica, com a DMD como alvo e sem registro para essa indicação terapêutica no Brasil. Dos 314 cadastros de estudos, 157 atendiam ao critério de elegibilidade referente à fase da pesquisa. Destes, apenas os estudos em andamento ou concluídos nos últimos 5 anos foram selecionados, restando 79 protocolos. A fim de mapear os resultados publicados para esses ensaios clínicos, utilizaram-se os códigos de registro do ClinicalTrials.gov nas bases de dados MEDLINE (via PubMed), EMBASE e Google Acadêmico. Na ausência de resultados, buscas complementares foram feitas com o nome do medicamento associado à indicação. Pipelines de desenvolvimento de medicamentos para DMD também foram revisados. Por fim, os sítios eletrônicos da Agência Nacional de Vigilância Sanitária (Anvisa, Food and Drug Administration (FDA, EUA) e European Medicines Agency (EMA) foram consultadas com o intuito de verificar os registros. O levantamento de resultados publicados e do registro dos medicamentos aconteceu entre 14 de junho e 17 de julho de 2021. MEDICAMENTOS: Ainda assim, os avanços na tecnologia devem ser reconhecidos. De 1998 a 2021, treze oligonucleotídeos antisense foram aprovados pela FDA, sendo quatro deles para DMD. Dos treze, dez receberam aprovação a partir de 2016. Esses resultados, somados aos ensaios clínicos em andamento e aos esforços para amadurecimento das tecnologias, consolidam a perspectiva desta abordagem terapêutica no tratamento da DMD. Por fim, é importante destacar que o pequeno número de pacientes incluídos nos estudos de doenças raras é agravado no caso das estratégias terapêuticas baseadas no genótipo. No caso da DMD, cada uma das tecnologias incluídas neste informe aplica-se a subgrupos com 8-15% do total de pacientes com a doença. INFORMAÇÕES ADICIONAIS: Uma série de tratamentos emergentes para DMD foram identificados durante o mapeamento de tecnologias para este informe, incluindo terapias com oligonucleotídeos antisense, substituição de genes mediada por vírus adeno-associado recombinante (AAVr) e diversos medicamentos para o tratamento sintomático da doença. No entanto, a maior parte encontra-se nas primeiras fases de desenvolvimento, não apresenta resultados publicados ou apenas resultados preliminares foram divulgados. Diante deste cenário, este informe abordou exclusivamente os medicamentos voltados à causa-base da doença e que foram recém-registrados nas agências FDA e/ou EMA. CONSIDERAÇÕES FINAIS: Os quatro oligômeros fosforodiamidato morfolino que receberam aprovação acelerada da FDA para o tratamento de DMD apresentaram aumento da produção de distrofina e parecem ser bem tolerados. No entanto, conforme previsto no Programa de Aprovação Acelerada da FDA, a continuação das aprovações depende da verificação de um benefício clínico em ensaios de confirmação. O eteplirsen e o golodirsen são os únicos oligômeros fosforodiamidato morfolino que apresentam avaliações publicadas acerca do seu efeito funcional. Até a última atualização deste informe, os benefícios clínicos desses medicamentos não estão bem estabelecidos, uma vez que houve declínio na deambulação e função pulmonar dos pacientes tratados, ainda que a maior parte dos valores tenham sido inferiores àqueles encontrados nos controles e na história natural publicada, especialmente para TC6. Para que ocorra a oferta desses medicamentos no SUS, é necessária a análise pela Conitec, conforme disposto na Lei nº 12.401/2011, que alterou a Lei nº 8.080/1990. Os relatórios de recomendação da Conitec levam em consideração as evidências científicas sobre eficácia, a acurácia, a efetividade e a segurança do medicamento, e, também, a avaliação econômica comparativa dos benefícios e dos custos em relação às tecnologias já incorporadas e o impacto da incorporação da tecnologia no SUS.


Subject(s)
Humans , Muscular Dystrophy, Duchenne/drug therapy , Morpholinos/therapeutic use , Brazil , Efficacy , Cost-Benefit Analysis/economics , Technological Development and Innovation Projects
3.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525659

ABSTRACT

Embryonic lipids are crucial for the formation of cellular membranes and dynamically participate in metabolic pathways. Cells can synthesize simple fatty acids, and the elongation of fatty acids facilitates the formation of complex lipids. The aim of this work was to investigate the involvement of the elongation of very long chain fatty acid enzyme 5 (ELOVL5) in embryonic development and lipid determination. Bovine embryos were produced in vitro using a standard protocol and randomly divided to receive one of three treatments at Day 4: morpholino (Mo) gene expression knockdown assay for ELOVL5 (ELOVL5-Mo), Mo antisense oligonucleotides for the thalassemic ß-globulin human mRNA (technical control Mo), and placebo (biological control). The phenotypes of embryonic development, cell number, ELOVL5 protein abundance, lipid droplet deposits, and lipid fingerprint were investigated. No detrimental effects (p > 0.05) were observed on embryo development in terms of cleavage (59.4 ± 3.5%, 63.6 ± 4.1%, and 65.4 ± 2.2%), blastocyst production (31.3 ± 4.2%, 28.1 ± 4.9%, and 36.1 ± 2.1%), and blastocyst cell number (99.6 ± 7.7, 100.2 ± 6.2, 86.8 ± 5.6), respectively, for biological control, technical control Mo, and ELOVL5-Mo. ELOVL5 protein abundance and cytoplasmic lipid droplet deposition were increased (p < 0.05) in ELOVL5-Mo-derived blastocysts compared with the controls. However, seven lipid species, including phosphatidylcholines, phosphatidylethanolamines, and triacylglycerol, were downregulated in the ELOVL5-Mo-derived blastocysts compared with the biological control. Therefore, ELOVL5 is involved in the determination of embryonic lipid content and composition. Transient translational blockage of ELOVL5 reduced the expression of specific lipid species and promoted increased cytoplasmic lipid droplet deposition, but with no apparent deleterious effect on embryonic development and blastocyst cell number.


Subject(s)
Blastocyst/metabolism , Cell Membrane/chemistry , Cytoplasm/chemistry , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Animals , Blastocyst/chemistry , Cattle , Embryonic Development , Fatty Acid Elongases/antagonists & inhibitors , Female , Gene Knockdown Techniques , Humans , Lipid Metabolism , Morpholinos/pharmacology , Pregnancy , beta-Globins/antagonists & inhibitors , beta-Globins/genetics
4.
J Vis Exp ; (146)2019 04 22.
Article in English | MEDLINE | ID: mdl-31058898

ABSTRACT

Cave animals provide a compelling system for investigating the evolutionary mechanisms and genetic bases underlying changes in numerous complex traits, including eye degeneration, albinism, sleep loss, hyperphagia, and sensory processing. Species of cavefish from around the world display a convergent evolution of morphological and behavioral traits due to shared environmental pressures between different cave systems. Diverse cave species have been studied in the laboratory setting. The Mexican tetra, Astyanax mexicanus, with sighted and blind forms, has provided unique insights into biological and molecular processes underlying the evolution of complex traits and is well-poised as an emerging model system. While candidate genes regulating the evolution of diverse biological processes have been identified in A. mexicanus, the ability to validate a role for individual genes has been limited. The application of transgenesis and gene-editing technology has the potential to overcome this significant impediment and to investigate the mechanisms underlying the evolution of complex traits. Here, we describe a different methodology for manipulating gene expression in A. mexicanus. Approaches include the use of morpholinos, Tol2 transgenesis, and gene-editing systems, commonly used in zebrafish and other fish models, to manipulate gene function in A. mexicanus. These protocols include detailed descriptions of timed breeding procedures, the collection of fertilized eggs, injections, and the selection of genetically modified animals. These methodological approaches will allow for the investigation of the genetic and neural mechanisms underlying the evolution of diverse traits in A. mexicanus.


Subject(s)
Characidae/genetics , Gene Editing , Animals , Biological Evolution , Caves , Female , Male , Morpholinos , Phenotype
5.
Sci Rep ; 9(1): 7131, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31073223

ABSTRACT

The Reprimo gene family comprises a group of single-exon genes for which their physiological function remains poorly understood. Heretofore, mammalian Reprimo (RPRM) has been described as a putative p53-dependent tumor suppressor gene that functions at the G2/M cell cycle checkpoint. Another family member, Reprimo-like (RPRML), has not yet an established role in physiology or pathology. Importantly, RPRML expression pattern is conserved between zebrafish and human species. Here, using CRISPR-Cas9 and antisense morpholino oligonucleotides, we disrupt the expression of rprml in zebrafish and demonstrate that its loss leads to impaired definitive hematopoiesis. The formation of hemangioblasts and the primitive wave of hematopoiesis occur normally in absence of rprml. Later in development there is a significant reduction in erythroid-myeloid precursors (EMP) at the posterior blood island (PBI) and a significant decline of definitive hematopoietic stem/progenitor cells (HSPCs). Furthermore, loss of rprml also increases the activity of caspase-3 in endothelial cells within the caudal hematopoietic tissue (CHT), the first perivascular niche where HSPCs reside during zebrafish embryonic development. Herein, we report an essential role for rprml during hematovascular development in zebrafish embryos, specifically during the definitive waves of hematopoiesis, indicating for the first time a physiological role for the rprml gene.


Subject(s)
Hemangioblasts/metabolism , Membrane Proteins/genetics , Zebrafish/embryology , Animals , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Embryonic Development , Hematopoiesis , Morpholinos/pharmacology , Multigene Family , Zebrafish/blood , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Biochem Pharmacol ; 163: 362-370, 2019 05.
Article in English | MEDLINE | ID: mdl-30849304

ABSTRACT

Treacher Collins Syndrome (TCS) is a congenital disease characterized by defects in the craniofacial skeleton and absence of mental alterations. Recently we modelled TCS in zebrafish (Danio rerio) embryos through the microinjection of Morpholino® oligonucleotides blocking the translation of the ortholog of the main causative gene (TCOF1). We showed that Cnbp, a key cytoprotective protein involved in normal rostral head development, was detected in lower levels (without changes in its mRNA expression) in TCS-like embryos. As previous reports suggested that Cnbp is degraded through the proteasomal pathway, we tested whether proteasome inhibitors (MG132 and Bortezomib (Velcade®, Millennium laboratories)) were able to ameliorate cranial skeleton malformations in TCS. Here we show that treatment with both proteasome inhibitors produced a robust craniofacial cartilage phenotype recovery. This recovery seems to be consequence of a decreased degradation of Cnbp in TCS-like embryos. Critical TCS manifestations, such as neuroepithelial cell death and cell redox imbalance were attenuated. Thus, proteasome inhibitors may offer an opportunity for TCS molecular and phenotypic manifestation's prevention. Although further development of new safe inhibitors compatible with administration during pregnancy is required, our results encourage this therapeutic approach.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Mandibulofacial Dysostosis/metabolism , Morpholinos/adverse effects , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Gene Knockdown Techniques , Mandibulofacial Dysostosis/pathology , Phosphoproteins/genetics , Zebrafish , Zebrafish Proteins/genetics
7.
Development ; 145(22)2018 11 21.
Article in English | MEDLINE | ID: mdl-30297374

ABSTRACT

Ric-8A is a pleiotropic guanine nucleotide exchange factor involved in the activation of various heterotrimeric G-protein pathways during adulthood and early development. Here, we sought to determine the downstream effectors of Ric-8A during the migration of the vertebrate cranial neural crest (NC) cells. We show that the Gα13 knockdown phenocopies the Ric-8A morphant condition, causing actin cytoskeleton alteration, protrusion instability, and a strong reduction in the number and dynamics of focal adhesions. In addition, the overexpression of Gα13 is sufficient to rescue Ric-8A-depleted cells. Ric-8A and Gα13 physically interact and colocalize in protrusions of the cells leading edge. The focal adhesion kinase FAK colocalizes and interacts with the endogenous Gα13, and a constitutively active form of Src efficiently rescues the Gα13 morphant phenotype in NC cells. We propose that Ric-8A-mediated Gα13 signalling is required for proper cranial NC cell migration by regulating focal adhesion dynamics and protrusion formation.


Subject(s)
Cell Movement , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neural Crest/cytology , Signal Transduction , Xenopus Proteins/metabolism , Xenopus/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Down-Regulation/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Focal Adhesions/drug effects , Models, Biological , Morpholinos/pharmacology , Neural Crest/metabolism , Phenotype , Signal Transduction/drug effects , Xenopus/embryology , src-Family Kinases/metabolism
8.
Neurology ; 90(24): e2146-e2154, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29752304

ABSTRACT

OBJECTIVE: To describe the quantification of novel dystrophin production in patients with Duchenne muscular dystrophy (DMD) after long-term treatment with eteplirsen. METHODS: Clinical study 202 was an observational, open-label extension of the randomized, controlled study 201 assessing the safety and efficacy of eteplirsen in patients with DMD with a confirmed mutation in the DMD gene amenable to correction by skipping of exon 51. Patients received once-weekly IV doses of eteplirsen 30 or 50 mg/kg. Upper extremity muscle biopsy samples were collected at combined study week 180, blinded, and assessed for dystrophin-related content by Western blot, Bioquant software measurement of dystrophin-associated immunofluorescence intensity, and percent dystrophin-positive fibers (PDPF). Results were contrasted with matched untreated biopsies from patients with DMD. Reverse transcription PCR followed by Sanger sequencing of newly formed slice junctions was used to confirm the mechanism of action of eteplirsen. RESULTS: Reverse transcription PCR analysis and sequencing of the newly formed splice junction confirmed that 100% of treated patients displayed the expected skipped exon 51 sequence. In treated patients vs untreated controls, Western blot analysis of dystrophin content demonstrated an 11.6-fold increase (p = 0.007), and PDPF analysis demonstrated a 7.4-fold increase (p < 0.001). The PDPF findings were confirmed in a re-examination of the sample (15.5-fold increase, p < 0.001). Dystrophin immunofluorescence intensity was 2.4-fold greater in treated patients than in untreated controls (p < 0.001). CONCLUSION: Taken together, the 4 assays, each based on unique evaluation mechanisms, provided evidence of eteplirsen muscle cell penetration, exon skipping, and induction of novel dystrophin expression. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence of the muscle cell penetration, exon skipping, and induction of novel dystrophin expression by eteplirsen, as confirmed by 4 assays.


Subject(s)
Dystrophin/biosynthesis , Exons/genetics , Morpholinos/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Biopsy , Child , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Treatment Outcome
9.
Mol Neurobiol ; 55(8): 6547-6557, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29327202

ABSTRACT

Adenosinergic signaling has important effects on brain function, anatomy, and physiology in both late and early stages of development. Exposure to caffeine, a non-specific blocker of adenosine receptor, has been indicated as a developmental risk factor. Disruption of adenosinergic signaling during early stages of development can change the normal neural network formation and possibly lead to an increase in susceptibility to seizures. In this work, morpholinos (MO) temporarily blocked the translation of adenosine receptor transcripts, adora1, adora2aa, and adora2ab, during the embryonic phase of zebrafish. It was observed that the block of adora2aa and adora2aa + adora2ab transcripts increased the mortality rate and caused high rate of malformations. To test the susceptibility of MO adora1, MO adora2aa, MO adora2ab, and MO adora2aa + adora2ab animals to seizure, pentylenetetrazole (10 mM) was used as a convulsant agent in larval and adult stages of zebrafish development. Although no MO promoted significant differences in latency time to reach the seizures stages in 7-day-old larvae, during the adult stage, all MO animals showed a decrease in the latency time to reach stages III, IV, and V of seizure. These results indicated that transient interventions in the adenosinergic signaling through high affinity adenosine receptors during embryonic development promote strong outcomes on survival and morphology. Additionally, long-term effects on neural development can lead to permanent impairment on neural signaling resulting in increased susceptibility to seizure.


Subject(s)
Adenosine/metabolism , Embryonic Development , Epilepsy/embryology , Epilepsy/pathology , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism , Animals , Disease Susceptibility , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Female , Fertilization , Larva/drug effects , Male , Morpholinos/pharmacology , Motor Activity/drug effects , Phenotype
11.
Neurochem Res ; 42(10): 2959-2967, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28623607

ABSTRACT

Presenilins are transmembrane proteases required for the proteolytic cleavage of Notch and also act as the catalytic core of the γ-secretase complex, which is responsible for the final cleavage of the amyloid precursor protein into Amyloid-ß (Aß) peptides of varying lengths. Presenilin-1 gene (psen1) mutations are the main cause of early-onset autosomal-dominant Familial Alzheimer Disease. Elucidating the roles of Presenilin-1 and other hallmark proteins involved in Alzheimer's disease is crucial for understanding the disease etiology and underlying molecular mechanisms. In our study, we used a morpholino antisense nucleotide that targets exon 8 splicing site of psen1 resulting in a dominant negative protein previously validated to investigate behavioral and molecular effects in 5 days post fertilization (dpf) zebrafish larvae. Morphants showed specific cognitive deficits in two optomotor tasks and morphological phenotypes similar to those induced by suppression of Notch signaling pathway. They also had increased mRNA levels of neurog1 at 5 dpf, confirming the potential interaction of Presenilin-1 and Notch in our model. We also evaluated levels of apoptotic markers including p53, PAR-4, Caspase-8 and bax-alpha and found only bax-a decreased at 5dpf. Western Blot analysis showed an increase in Aß1-42 and a decrease in the selective post-synaptic marker PSD-95 at 5 dpf. Our data demonstrates that psen1 splicing interference induces phenotypes that resemble early-stage AD, including cognitive deficit, Aß1-42 accumulation and synaptic reduction, reinforcing the potential contribution of zebrafish larvae to studies of human brain diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/metabolism , Disks Large Homolog 4 Protein/metabolism , Morpholinos/metabolism , Peptide Fragments/metabolism , Presenilin-1/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cognition/physiology , Cognitive Dysfunction/genetics , Larva , Mutation/genetics , Peptide Fragments/genetics , Synapses/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Dev Biol ; 403(1): 89-100, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25912688

ABSTRACT

The Zmiz2 (Zimp7) protein and its homolog Zmiz1 (Zimp10) were initially identified in humans as androgen receptor co-activators. Sequence analysis revealed the presence of an SP-RING/Miz domain, which is highly conserved in members of the PIAS family and confers SUMO-conjugating activity. Zimp7 has been shown to interact with components of the Wnt/ß-Catenin signaling pathway and with Brg1 and BAF57, components of the ATP-dependent mammalian SWI/SNF-like BAF chromatin-remodeling complexes. In this work, we analyze the role of zygotic Zimp7 in zebrafish development. We describe evidence indicating that Zimp7 is required for mesoderm development and dorsoventral patterning. Morpholino-mediated reduction of zygotic Zimp7 produced axial mesodermal defects that were preceded by up-regulation of organizer genes such as bozozok, goosecoid and floating head at the onset of gastrulation and by down-regulation of the ventral markers vox, vent and eve1 indicating loss of the ventrolateral mesoderm. Consistently, embryos overexpressing zimp7 RNA exhibited midline defects such as loss of forebrain and cyclopia accompanied by transcriptional changes directly opposite of those found in the morphants. In addition, the patterning of ventralized embryos produced by the overexpression of vox and vent was restored by a reduction of Zimp7 activity. Altogether, our findings indicate that Zimp7 is involved in transcriptional regulation of factors that are essential for patterning in the dorsoventral axis.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Organizers, Embryonic/embryology , Protein Inhibitors of Activated STAT/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zinc Fingers/genetics , Animals , Blastula/metabolism , Gastrulation/genetics , Gene Knockdown Techniques , Goosecoid Protein/biosynthesis , Homeodomain Proteins/biosynthesis , Mesoderm/embryology , Morpholinos/genetics , Protein Inhibitors of Activated STAT/genetics , RNA, Messenger/biosynthesis , Repressor Proteins/biosynthesis , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription, Genetic/genetics , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
13.
Development ; 141(23): 4598-609, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25377554

ABSTRACT

Egg activation at fertilization is an excellent process for studying calcium regulation. Nicotinic acid adenine dinucleotide-phosphate (NAADP), a potent calcium messenger, is able to trigger calcium release, likely through two-pore channels (TPCs). Concomitantly, a family of ectocellular enzymes, the ADP-ribosyl cyclases (ARCs), has emerged as being able to change their enzymatic mode from one of nucleotide cyclization in formation of cADPR to a base-exchange reaction in the generation of NAADP. Using sea star oocytes we gain insights into the functions of endogenously expressed TPCs and ARCs in the context of the global calcium signals at fertilization. Three TPCs and one ARC were found in the sea star (Patiria miniata) that were localized in the cortex of the oocytes and eggs. PmTPCs were localized in specialized secretory organelles called cortical granules, and PmARCs accumulated in a different, unknown, set of vesicles, closely apposed to the cortical granules in the egg cortex. Using morpholino knockdown of PmTPCs and PmARC in the oocytes, we found that both calcium regulators are essential for early embryo development, and that knockdown of PmTPCs leads to aberrant construction of the fertilization envelope at fertilization and changes in cortical granule pH. The calcium signals at fertilization are not significantly altered when individual PmTPCs are silenced, but the timing and shape of the cortical flash and calcium wave are slightly changed when the expression of all three PmTPCs is perturbed concomitantly, suggesting a cooperative activity among TPC isoforms in eliciting calcium signals that may influence localized physiological activities.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Embryo, Nonmammalian/metabolism , Fertilization/physiology , Oocytes/metabolism , Starfish/physiology , ADP-ribosyl Cyclase/genetics , Animals , Calcium Channels/genetics , Immunoblotting , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mass Spectrometry , Microscopy, Electron, Transmission , Morpholinos/genetics , NADP/analogs & derivatives , NADP/metabolism , Starfish/metabolism
14.
PLoS One ; 9(10): e110559, 2014.
Article in English | MEDLINE | ID: mdl-25343614

ABSTRACT

In vertebrates, the embryonic dorsal midline is a crucial signalling centre that patterns the surrounding tissues during development. Members of the FoxA subfamily of transcription factors are expressed in the structures that compose this centre. Foxa2 is essential for dorsal midline development in mammals, since knock-out mouse embryos lack a definitive node, notochord and floor plate. The related gene foxA4 is only present in amphibians. Expression begins in the blastula -chordin and -noggin expressing centre (BCNE) and is later restricted to the dorsal midline derivatives of the Spemann's organiser. It was suggested that the early functions of mammalian foxa2 are carried out by foxA4 in frogs, but functional experiments were needed to test this hypothesis. Here, we show that some important dorsal midline functions of mammalian foxa2 are exerted by foxA4 in Xenopus. We provide new evidence that the latter prevents the respecification of dorsal midline precursors towards contiguous fates, inhibiting prechordal and paraxial mesoderm development in favour of the notochord. In addition, we show that foxA4 is required for the correct regionalisation and maintenance of the central nervous system. FoxA4 participates in constraining the prospective rostral forebrain territory during neural specification and is necessary for the correct segregation of the most anterior ectodermal derivatives, such as the cement gland and the pituitary anlagen. Moreover, the early expression of foxA4 in the BCNE (which contains precursors of the whole forebrain and most of the midbrain and hindbrain) is directly required to restrict anterior neural development.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Embryo, Nonmammalian/metabolism , Forkhead Transcription Factors/metabolism , Mesoderm/embryology , Notochord/embryology , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Blastula/drug effects , Blastula/metabolism , Body Patterning/drug effects , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Gene Knockdown Techniques , Glycoproteins/metabolism , Head/abnormalities , Head/embryology , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Models, Biological , Morphogenesis/drug effects , Morpholinos/pharmacology , Neural Plate/embryology , Neural Plate/metabolism , Neurogenesis/drug effects , Notochord/drug effects , Notochord/metabolism , Phenotype , Xenopus/metabolism
15.
Dev Biol ; 370(1): 42-51, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22841627

ABSTRACT

The capacity for tissue and organ regeneration in humans is dwarfed by comparison to that of salamanders. Emerging evidence suggests that mechanisms learned from the early phase of salamander limb regeneration-wound healing, cellular dedifferentiation and blastemal formation-will reveal therapeutic approaches for tissue regeneration in humans. Here we describe a unique transcriptional fingerprint of regenerating limb tissue in the Mexican axolotl (Ambystoma mexicanum) that is indicative of cellular reprogramming of differentiated cells to a germline-like state. Two genes that are required for self-renewal of germ cells in mice and flies, Piwi-like 1 (PL1) and Piwi-like 2 (PL2), are expressed in limb blastemal cells, the basal layer keratinocytes and the thickened apical epithelial cap in the wound epidermis in the regenerating limb. Depletion of PL1 and PL2 by morpholino oligonucleotides decreased cell proliferation and increased cell death in the blastema leading to a significant retardation of regeneration. Examination of key molecules that are known to be required for limb development or regeneration further revealed that FGF8 is transcriptionally downregulated in the presence of the morpholino oligos, indicating PL1 and PL2 might participate in FGF signaling during limb regeneration. Given the requirement for FGF signaling in limb development and regeneration, the results suggest that PL1 and PL2 function to establish a unique germline-like state that is associated with successful regeneration.


Subject(s)
Ambystoma mexicanum/physiology , Extremities/physiology , Gene Expression Regulation, Developmental/physiology , Germ Cells/metabolism , Regeneration/physiology , Ambystoma mexicanum/genetics , Amino Acid Sequence , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Molecular Sequence Data , Morpholinos/genetics , Regeneration/genetics , Wound Healing/physiology
16.
Dev Biol ; 364(2): 99-113, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22309705

ABSTRACT

Neural crest induction is the result of the combined action at the neural plate border of FGF, BMP, and Wnt signals from the neural plate, mesoderm and nonneural ectoderm. In this work we show that the expression of Indian hedgehog (Ihh, formerly named Banded hedgehog) and members of the Hedgehog pathway occurs at the prospective neural fold, in the premigratory and migratory neural crest. We performed a functional analysis that revealed the requirement of Ihh signaling in neural crest development. During the early steps of neural crest induction loss of function experiments with antisense morpholino or locally grafted cyclopamine-loaded beads suppressed the expression of early neural crest markers concomitant with the increase in neural and epidermal markers. We showed that changes in Ihh activity produced no alterations in either cell proliferation or apoptosis, suggesting that this signal involves cell fate decisions. A temporal analysis showed that Hedgehog is continuously required not only in the early and late specification but also during the migration of the neural crest. We also established that the mesodermal source of Ihh is important to maintain specification and also to support the migratory process. By a combination of embryological and molecular approaches our results demonstrated that Ihh signaling drives in the migration of neural crest cells by autocrine or paracrine mechanisms. Finally, the abrogation of Ihh signaling strongly affected only the formation of cartilages derived from the neural crest, while no effects were observed on melanocytes. Taken together, our results provide insights into the role of the Ihh cell signaling pathway during the early steps of neural crest development.


Subject(s)
Cell Movement , Hedgehog Proteins/physiology , Neural Crest/growth & development , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biomarkers/analysis , Cell Proliferation/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental/drug effects , Melanocytes/drug effects , Melanocytes/physiology , Morpholinos/pharmacology , Neural Crest/drug effects , Signal Transduction , Veratrum Alkaloids/pharmacology , Xenopus laevis/metabolism
17.
Dev Growth Differ ; 53(8): 934-47, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21999883

ABSTRACT

Cellular nucleic acid binding protein (Cnbp) is a highly conserved single-stranded nucleic acid binding protein required for rostral head development. The use of a morpholino that inhibits Cnbp mRNA translation previously revealed a role of Cnbp in balancing neural crest cell apoptosis and proliferation in the developing zebrafish. Here, we report the use of another morpholino that specifically modifies the splicing of Cnbp pre-mRNA resulting in a reduction of full-length mRNA levels along with the generation of a novel transcript coding for an isoform that may act as dominant negative proteins. The use of this morpholino resulted in more severe phenotypes that enabled us to demonstrate that Cnbp loss-of-function adversely affects the formation and survival of craniofacial cartilaginous structures not only controlling the ratio of cell proliferation and apoptosis but also defining skeletogenic neural crest cell fate.


Subject(s)
Neural Crest/embryology , RNA-Binding Proteins/physiology , Skull/embryology , Zebrafish Proteins/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Models, Biological , Molecular Sequence Data , Morpholinos/pharmacology , Neural Crest/drug effects , Neural Crest/metabolism , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sequence Homology , Skull/drug effects , Skull/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL