Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.102
Filtrar
1.
Development ; 151(18)2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39253748

RESUMO

Caenorhabditis elegans males undergo sex-specific tail tip morphogenesis (TTM) under the control of the DM-domain transcription factor DMD-3. To find genes regulated by DMD-3, we performed RNA-seq of laser-dissected tail tips. We identified 564 genes differentially expressed (DE) in wild-type males versus dmd-3(-) males and hermaphrodites. The transcription profile of dmd-3(-) tail tips is similar to that in hermaphrodites. For validation, we analyzed transcriptional reporters for 49 genes and found male-specific or male-biased expression for 26 genes. Only 11 DE genes overlapped with genes found in a previous RNAi screen for defective TTM. GO enrichment analysis of DE genes finds upregulation of genes within the unfolded protein response pathway and downregulation of genes involved in cuticle maintenance. Of the DE genes, 40 are transcription factors, indicating that the gene network downstream of DMD-3 is complex and potentially modular. We propose modules of genes that act together in TTM and are co-regulated by DMD-3, among them the chondroitin synthesis pathway and the hypertonic stress response.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese , RNA-Seq , Cauda , Animais , Caenorhabditis elegans/genética , Morfogênese/genética , Masculino , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Redes Reguladoras de Genes , Especificidade de Órgãos/genética
2.
Development ; 151(19)2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39258889

RESUMO

Pharyngeal endoderm cells undergo convergence and extension (C&E), which is essential for endoderm pouch formation and craniofacial development. Our previous work implicates Gα13/RhoA-mediated signaling in regulating this process, but the underlying mechanisms remain unclear. Here, we have used endoderm-specific transgenic and Gα13 mutant zebrafish to demonstrate that Gα13 plays a crucial role in pharyngeal endoderm C&E by regulating RhoA activation and E-cadherin expression. We showed that during C&E, endodermal cells gradually establish stable cell-cell contacts, acquire apical-basal polarity and undergo actomyosin-driven apical constriction, which are processes that require Gα13. Additionally, we found that Gα13-deficient embryos exhibit reduced E-cadherin expression, partially contributing to endoderm C&E defects. Notably, interfering with RhoA function disrupts spatial actomyosin activation without affecting E-cadherin expression. Collectively, our findings identify crucial cellular processes for pharyngeal endoderm C&E and reveal that Gα13 controls this through two independent pathways - modulating RhoA activation and regulating E-cadherin expression - thus unveiling intricate mechanisms governing pharyngeal endoderm morphogenesis.


Assuntos
Caderinas , Endoderma , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP , Regulação da Expressão Gênica no Desenvolvimento , Faringe , Proteínas de Peixe-Zebra , Peixe-Zebra , Proteína rhoA de Ligação ao GTP , Animais , Endoderma/metabolismo , Endoderma/embriologia , Endoderma/citologia , Caderinas/metabolismo , Caderinas/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Faringe/embriologia , Faringe/metabolismo , Actomiosina/metabolismo , Transdução de Sinais , Morfogênese/genética , Polaridade Celular , Animais Geneticamente Modificados , Embrião não Mamífero/metabolismo
3.
PLoS Biol ; 22(8): e3002762, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39173068

RESUMO

During embryonic development, tissues and organs are gradually shaped into their functional morphologies through a series of spatiotemporally tightly orchestrated cell behaviors. A highly conserved organ shape across metazoans is the epithelial tube. Tube morphogenesis is a complex multistep process of carefully choreographed cell behaviors such as convergent extension, cell elongation, and lumen formation. The identity of the signaling molecules that coordinate these intricate morphogenetic steps remains elusive. The notochord is an essential tubular organ present in the embryonic midline region of all members of the chordate phylum. Here, using genome editing, pharmacology and quantitative imaging in the early chordate Ciona intestinalis we show that Ano10/Tmem16k, a member of the evolutionarily ancient family of transmembrane proteins called Anoctamin/TMEM16 is essential for convergent extension, lumen expansion, and connection during notochord morphogenesis. We find that Ano10/Tmem16k works in concert with the plasma membrane (PM) localized Na+/Ca2+ exchanger (NCX) and the endoplasmic reticulum (ER) residing SERCA, RyR, and IP3R proteins to establish developmental stage specific Ca2+ signaling molecular modules that regulate notochord morphogenesis and Ca2+ dynamics. In addition, we find that the highly conserved Ca2+ sensors calmodulin (CaM) and Ca2+/calmodulin-dependent protein kinase (CaMK) show an Ano10/Tmem16k-dependent subcellular localization. Their pharmacological inhibition leads to convergent extension, tubulogenesis defects, and deranged Ca2+ dynamics, suggesting that Ano10/Tmem16k is involved in both the "encoding" and "decoding" of developmental Ca2+ signals. Furthermore, Ano10/Tmem16k mediates cytoskeletal reorganization during notochord morphogenesis, likely by altering the localization of 2 important cytoskeletal regulators, the small GTPase Ras homolog family member A (RhoA) and the actin binding protein Cofilin. Finally, we use electrophysiological recordings and a scramblase assay in tissue culture to demonstrate that Ano10/Tmem16k likely acts as an ion channel but not as a phospholipid scramblase. Our results establish Ano10/Tmem16k as a novel player in the prevertebrate molecular toolkit that controls organ morphogenesis across scales.


Assuntos
Anoctaminas , Ciona intestinalis , Morfogênese , Notocorda , Animais , Notocorda/metabolismo , Notocorda/embriologia , Anoctaminas/metabolismo , Anoctaminas/genética , Ciona intestinalis/metabolismo , Ciona intestinalis/embriologia , Ciona intestinalis/genética , Morfogênese/genética , Sinalização do Cálcio , Regulação da Expressão Gênica no Desenvolvimento , Retículo Endoplasmático/metabolismo , Cálcio/metabolismo
4.
JCI Insight ; 9(16)2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39171526

RESUMO

Craniofacial dysmorphisms are among the most common birth defects. Proteasome mutations frequently result in craniofacial dysmorphisms, including lower jaw malformations; however, the underlying mechanisms are unknown. Here, we used a zebrafish proteasome subunit ß 1 (psmb1) mutant to define the cellular mechanisms underlying proteasome mutation-induced craniofacial dysmorphisms. psmb1 mutants exhibited a flattened ceratohyal and smaller Meckel's and palatoquadrate cartilages. Ceratohyal flattening was a result of failed chondrocyte convergent extension, accompanied by reduced numbers of chondrocytes in the lower jaw due to defects in chondrocyte differentiation. Morphogenesis of craniofacial muscles and tendons was similarly perturbed. psmb1 mutants lacked the hyohyal muscles, and craniofacial tendons were shortened and disorganized. We additionally identified a critical period for proteasome function in craniofacial development, specifically during chondrocyte and muscle differentiation. psmb1 overexpression in sox10+ cells of mutant embryos rescued both cartilage and tendon phenotypes but induced only a partial rescue of the muscle phenotype, indicating that psmb1 was required in both tissue-autonomous and nonautonomous fashions during craniofacial development. Overall, our work demonstrates that psmb1 is required for craniofacial cartilage, tendon, and muscle differentiation and morphogenesis.


Assuntos
Cartilagem , Condrócitos , Morfogênese , Complexo de Endopeptidases do Proteassoma , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Peixe-Zebra/genética , Morfogênese/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Condrócitos/metabolismo , Cartilagem/metabolismo , Cartilagem/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/patologia , Tendões/embriologia , Tendões/metabolismo , Tendões/anormalidades , Tendões/patologia , Diferenciação Celular/genética , Mutação , Regulação da Expressão Gênica no Desenvolvimento , Condrogênese/genética
5.
RNA Biol ; 21(1): 11-22, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39190491

RESUMO

Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability and is caused by mutations in the gene encoding the Fragile X messenger ribonucleoprotein (FMRP). FMRP is an evolutionarily conserved and neuronally enriched RNA-binding protein (RBP) with functions in RNA editing, RNA transport, and protein translation. Specific target RNAs play critical roles in neurodevelopment, including the regulation of neurite morphogenesis, synaptic plasticity, and cognitive function. The different biological functions of FMRP are modulated by its cooperative interaction with distinct sets of neuronal RNA and protein-binding partners. Here, we focus on interactions between FMRP and components of the microRNA (miRNA) pathway. Using the Drosophila S2 cell model system, we show that the Drosophila ortholog of FMRP (dFMRP) can repress translation when directly tethered to a reporter mRNA. This repression requires the activity of AGO1, GW182, and MOV10/Armitage, conserved proteins associated with the miRNA-containing RNA-induced silencing complex (miRISC). Additionally, we find that untagged dFMRP can interact with a short stem-loop sequence in the translational reporter, a prerequisite for repression by exogenous miR-958. Finally, we demonstrate that dFmr1 interacts genetically with GW182 to control neurite morphogenesis. These data suggest that dFMRP may recruit the miRISC to nearby miRNA binding sites and repress translation via its cooperative interactions with evolutionarily conserved components of the miRNA pathway.


Assuntos
Proteínas de Drosophila , Proteína do X Frágil da Deficiência Intelectual , MicroRNAs , Neuritos , Biossíntese de Proteínas , Animais , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Neuritos/metabolismo , Morfogênese/genética , Complexo de Inativação Induzido por RNA/metabolismo , Complexo de Inativação Induzido por RNA/genética , Drosophila/metabolismo , Drosophila/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Ligação Proteica
6.
Plant Physiol ; 196(2): 1595-1607, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39054117

RESUMO

Root hair growth has been studied to understand the principles underlying the regulation of directional growth. Arabidopsis (Arabidopsis thaliana) [Ca2+]cyt-ASSOCIATED PROTEIN KINASE 1 (CAP1) maintains normal vesicle trafficking and cytoskeleton arrangement during root hair growth in response to ammonium signaling. In the current study, we identified CAP1 SUPPRESSOR 1 (CAPS1) as a genetic suppressor of the cap1-1 mutation. The CAPS1 mutation largely rescued the short root hair phenotype of cap1-1. Loss of CAPS1 function resulted in significantly longer root hairs in cap1-1. MutMap analysis revealed that CAPS1 is identical to NIMA (NEVER IN MITOSIS A)-RELATED KINASE 2 (NEK2). In addition, our studies showed that NEK2 is expressed in root and root hairs. Its distribution was associated with the pattern of microtubule (MT) arrangement and partially colocalized with CAP1. Further biochemical studies revealed that CAP1 physically interacts with NEK2 and may enhance its phosphorylation. Our study suggests that NEK2 acts as a potential phosphorylation target of CAP1 in maintaining the stability of root hair MTs to regulate root hair elongation.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Quinases Relacionadas a NIMA , Raízes de Plantas , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/genética , Raízes de Plantas/metabolismo , Arabidopsis/genética , Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Quinases Relacionadas a NIMA/metabolismo , Quinases Relacionadas a NIMA/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Microtúbulos/metabolismo , Morfogênese/genética , Mutação/genética , Fosforilação , Regulação da Expressão Gênica de Plantas
7.
Dev Biol ; 515: 92-101, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39029571

RESUMO

Congenital lung malformations are fatal at birth in their severe forms. Prevention and early intervention of these birth defects require a comprehensive understanding of the molecular mechanisms of lung development. We find that the loss of inturned (Intu), a cilia and planar polarity effector gene, severely disrupts growth and branching morphogenesis of the mouse embryonic lungs. Consistent with our previous results indicating an important role for Intu in ciliogenesis and hedgehog (Hh) signaling, we find greatly reduced number of primary cilia in both the epithelial and mesenchymal tissues of the lungs. We also find significantly reduced expression of Gli1 and Ptch1, direct targets of Hh signaling, suggesting disruption of cilia-dependent Hh signaling in Intu mutant lungs. An agonist of the Hh pathway activator, smoothened, increases Hh target gene expression and tubulogenesis in explanted wild type, but not Intu mutant, lungs, suggesting impaired Hh signaling response underlying lung morphogenetic defects in Intu mutants. Furthermore, removing both Gli2 and Intu completely abolishes branching morphogenesis of the lung, strongly supporting a mechanism by which Intu regulates lung growth and patterning through cilia-dependent Hh signaling. Moreover, a transcriptomics analysis identifies around 200 differentially expressed genes (DEGs) in Intu mutant lungs, including known Hh target genes Gli1, Ptch1/2 and Hhip. Genes involved in muscle differentiation and function are highly enriched among the DEGs, consistent with an important role of Hh signaling in airway smooth muscle differentiation. In addition, we find that the difference in gene expression between the left and right lungs diminishes in Intu mutants, suggesting an important role of Intu in asymmetrical growth and patterning of the mouse lungs.


Assuntos
Cílios , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog , Pulmão , Transdução de Sinais , Animais , Camundongos , Padronização Corporal/genética , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Pulmão/embriologia , Pulmão/metabolismo , Morfogênese/genética , Receptor Patched-1/metabolismo , Receptor Patched-1/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/genética
8.
mBio ; 15(8): e0153524, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38980041

RESUMO

At human body temperature, the fungal pathogen Candida albicans can transition from yeast to filamentous morphologies in response to host-relevant cues. Additionally, elevated temperatures encountered during febrile episodes can independently induce C. albicans filamentation. However, the underlying genetic pathways governing this developmental transition in response to elevated temperatures remain largely unexplored. Here, we conducted a functional genomic screen to unravel the genetic mechanisms orchestrating C. albicans filamentation specifically in response to elevated temperature, implicating 45% of genes associated with the spliceosome or pre-mRNA splicing in this process. Employing RNA-Seq to elucidate the relationship between mRNA splicing and filamentation, we identified greater levels of intron retention in filaments compared to yeast, which correlated with reduced expression of the affected genes. Intriguingly, homozygous deletion of a gene encoding a spliceosome component important for filamentation (PRP19) caused even greater levels of intron retention compared with wild type and displayed globally dysregulated gene expression. This suggests that intron retention is a mechanism for fine-tuning gene expression during filamentation, with perturbations of the spliceosome exacerbating this process and blocking filamentation. Overall, this study unveils a novel biological process governing C. albicans filamentation, providing new insights into the complex regulation of this key virulence trait.IMPORTANCEFungal pathogens such as Candida albicans can cause serious infections with high mortality rates in immunocompromised individuals. When C. albicans is grown at temperatures encountered during human febrile episodes, yeast cells undergo a transition to filamentous cells, and this process is key to its virulence. Here, we expanded our understanding of how C. albicans undergoes filamentation in response to elevated temperature and identified many genes involved in mRNA splicing that positively regulate filamentation. Through transcriptome analyses, we found that intron retention is a mechanism for fine-tuning gene expression in filaments, and perturbation of the spliceosome exacerbates intron retention and alters gene expression substantially, causing a block in filamentation. This work adds to the growing body of knowledge on the role of introns in fungi and provides new insights into the cellular processes that regulate a key virulence trait in C. albicans.


Assuntos
Candida albicans , Proteínas Fúngicas , Regulação Fúngica da Expressão Gênica , Spliceossomos , Candida albicans/genética , Candida albicans/patogenicidade , Candida albicans/crescimento & desenvolvimento , Candida albicans/fisiologia , Candida albicans/metabolismo , Spliceossomos/genética , Spliceossomos/metabolismo , Humanos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Morfogênese/genética , Splicing de RNA , Virulência , Hifas/crescimento & desenvolvimento , Hifas/genética , Íntrons/genética
9.
Funct Integr Genomics ; 24(4): 120, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38960936

RESUMO

The Drosophila egg chamber (EC) starts as a spherical tissue at the beginning. With maturation, the outer follicle cells of EC collectively migrate in a direction perpendicular to the anterior-posterior axis, to shape EC from spherical to ellipsoidal. Filamentous actin (F-actin) plays a significant role in shaping individual migratory cells to the overall EC shape, like in every cell migration. The primary focus of this article is to unveil the function of different Actin Binding Proteins (ABPs) in regulating mature Drosophila egg shape. We have screened 66 ABPs, and the genetic screening data revealed that individual knockdown of Arp2/3 complex genes and the "capping protein ß" (cpb) gene have severely altered the egg phenotype. Arpc1 and cpb RNAi mediated knockdown resulted in the formation of spherical eggs which are devoid of dorsal appendages. Studies also showed the role of Arpc1 and cpb on the number of laid eggs and follicle cell morphology. Furthermore, the depletion of Arpc1 and cpb resulted in a change in F-actin quantity. Together, the data indicate that Arpc1 and cpb regulate Drosophila egg shape, F-actin management, egg-laying characteristics and dorsal appendages formation.


Assuntos
Actinas , Proteínas de Drosophila , Morfogênese , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Actinas/metabolismo , Actinas/genética , Feminino , Morfogênese/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Proteínas de Capeamento de Actina/metabolismo , Proteínas de Capeamento de Actina/genética , Óvulo/metabolismo , Óvulo/crescimento & desenvolvimento
10.
PLoS Genet ; 20(7): e1011330, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39083711

RESUMO

Coordinated activation and inhibition of F-actin supports the movements of morphogenesis. Understanding the proteins that regulate F-actin is important, since these proteins are mis-regulated in diseases like cancer. Our studies of C. elegans embryonic epidermal morphogenesis identified the GTPase CED-10/Rac1 as an essential activator of F-actin. However, we need to identify the GEF, or Guanine-nucleotide Exchange Factor, that activates CED-10/Rac1 during embryonic cell migrations. The two-component GEF, CED-5/CED-12, is known to activate CED-10/Rac1 to promote cell movements that result in the engulfment of dying cells during embryogenesis, and a later cell migration of the larval Distal Tip Cell. It is believed that CED-5/CED-12 powers cellular movements of corpse engulfment and DTC migration by promoting F-actin formation. Therefore, we tested if CED-5/CED-12 was involved in embryonic migrations, and got a contradictory result. CED-5/CED-12 definitely support embryonic migrations, since their loss led to embryos that died due to failed epidermal cell migrations. However, CED-5/CED-12 inhibited F-actin in the migrating epidermis, the opposite of what was expected for a CED-10 GEF. To address how CED-12/CED-5 could have two opposing effects on F-actin, during corpse engulfment and cell migration, we investigated if CED-12 harbors GAP (GTPase Activating Protein) functions. A candidate GAP region in CED-12 faces away from the CED-5 GEF catalytic region. Mutating a candidate catalytic Arginine in the CED-12 GAP region (R537A) altered the epidermal cell migration function, and not the corpse engulfment function. We interfered with GEF function by interfering with CED-5's ability to bind Rac1/CED-10. Mutating Serine-Arginine in CED-5/DOCK predicted to bind and stabilize Rac1 for catalysis, resulted in loss of both ventral enclosure and corpse engulfment. Genetic and expression studies strongly support that the GAP function likely acts on different GTPases. Thus, we propose CED-5/CED-12 support the cycling of multiple GTPases, by using distinct domains, to both promote and inhibit F-actin nucleation.


Assuntos
Actinas , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Movimento Celular , Animais , Actinas/metabolismo , Motivos de Aminoácidos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Epiderme/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Morfogênese/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética
11.
Int J Mol Sci ; 25(14)2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39063093

RESUMO

Higher plants have developed complex mechanisms to adapt to fluctuating environmental conditions with light playing a vital role in photosynthesis and influencing various developmental processes, including photomorphogenesis. Exposure to ultraviolet (UV) radiation can cause cellular damage, necessitating effective DNA repair mechanisms. Histone acetyltransferases (HATs) play a crucial role in regulating chromatin structure and gene expression, thereby contributing to the repair mechanisms. HATs facilitate chromatin relaxation, enabling transcriptional activation necessary for plant development and stress responses. The intricate relationship between HATs, light signaling pathways and chromatin dynamics has been increasingly understood, providing valuable insights into plant adaptability. This review explores the role of HATs in plant photomorphogenesis, chromatin remodeling and gene regulation, highlighting the importance of chromatin modifications in plant responses to light and various stressors. It emphasizes the need for further research on individual HAT family members and their interactions with other epigenetic factors. Advanced genomic approaches and genome-editing technologies offer promising avenues for enhancing crop resilience and productivity through targeted manipulation of HAT activities. Understanding these mechanisms is essential for developing strategies to improve plant growth and stress tolerance, contributing to sustainable agriculture in the face of a changing climate.


Assuntos
Regulação da Expressão Gênica de Plantas , Histona Acetiltransferases , Desenvolvimento Vegetal , Raios Ultravioleta , Histona Acetiltransferases/metabolismo , Histona Acetiltransferases/genética , Desenvolvimento Vegetal/genética , Desenvolvimento Vegetal/efeitos da radiação , Plantas/genética , Plantas/efeitos da radiação , Plantas/metabolismo , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Cromatina/genética , Morfogênese/efeitos da radiação , Morfogênese/genética
12.
Adv Sci (Weinh) ; 11(32): e2400238, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38923264

RESUMO

The placenta links feto-maternal circulation for exchanges of nutrients, gases, and metabolic wastes between the fetus and mother, being essential for pregnancy process and maintenance. The allantois and mesodermal components of amnion, chorion, and yolk sac are derived from extraembryonic mesoderm (Ex-Mes), however, the mechanisms contributing to distinct components of the placenta and regulation the interactions between allantois and epithelium during chorioallantoic fusion and labyrinth formation remains unclear. Isl1 is expressed in progenitors of the Ex-Mes and allantois the Isl1 mut mouse line is analyzed to investigate contribution of Isl1+ Ex-Mes / allantoic progenitors to cells of the allantois and placenta. This study shows that Isl1 identifies the Ex-Mes progenitors for endothelial and vascular smooth muscle cells, and most of the mesenchymal cells of the placenta and umbilical cord. Deletion of Isl1 causes defects in allantois growth, chorioallantoic fusion, and placenta vessel morphogenesis. RNA-seq and CUT&Tag analyses revealed that Isl1 promotes allantoic endothelial, inhibits mesenchymal cell differentiation, and allantoic signals regulated by Isl1 mediating the inductive interactions between the allantois and chorion critical for chorionic epithelium differentiation, villous formation, and labyrinth angiogenesis. This study above reveals that Isl1 plays roles in regulating multiple genetic and epigenetic pathways of vascular morphogenesis, provides the insight into the mechanisms for placental formation, highlighting the necessity of Isl1 for placenta formation/pregnant maintenance.


Assuntos
Alantoide , Proteínas com Homeodomínio LIM , Mesoderma , Placenta , Fatores de Transcrição , Animais , Gravidez , Camundongos , Feminino , Proteínas com Homeodomínio LIM/metabolismo , Proteínas com Homeodomínio LIM/genética , Alantoide/metabolismo , Placenta/metabolismo , Placenta/irrigação sanguínea , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Mesoderma/metabolismo , Mesoderma/embriologia , Morfogênese/genética , Morfogênese/fisiologia , Placentação/genética , Placentação/fisiologia
13.
Cells Dev ; 179: 203932, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38852677

RESUMO

Tooth morphogenesis is a critically ordered process manipulated by a range of signaling factors. Particularly, the involvement of fine-tuned signaling mediated by non-coding RNAs has been of longstanding interest. Here, we revealed a double-negative feedback loop acted by a long non-coding RNA (LOC102159588) and a microRNA (miR-133b) that modulated tooth morphogenesis of miniature swine. Mechanistically, miR-133b repressed the transcription of LOC102159588 through downstream target Sp1. Conversely, LOC102159588 not only inhibited the transport of pre-miR-133b from the nucleus to the cytoplasm by regulating exportin-5 but also served as a sponge in the cytoplasm, suppressing functional miR-133b. Together, the double-negative feedback loop maintained normal tooth morphogenesis by modulating endogenous apoptosis. Related disruptions would lead to an arrest of tooth development and may result in tooth malformations.


Assuntos
Retroalimentação Fisiológica , MicroRNAs , Morfogênese , Dente , Animais , Morfogênese/genética , Dente/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Suínos , Apoptose/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Odontogênese/genética , Porco Miniatura
14.
EMBO Rep ; 25(7): 2861-2877, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38839944

RESUMO

In developing olfactory bulb (OB), mitral cells (MCs) remodel their dendrites to establish the precise olfactory circuit, and these circuits are critical for individuals to sense odors and elicit behaviors for survival. However, how microtubules (MTs) participate in the process of dendritic remodeling remains elusive. Here, we reveal that calmodulin-regulated spectrin-associated proteins (CAMSAPs), a family of proteins that bind to the minus-end of the noncentrosomal MTs, play a crucial part in the development of MC dendrites. We observed that Camsap2 knockout (KO) males are infertile while the reproductive tract is normal. Further study showed that the infertility was due to the severe defects of mating behavior in male mice. Besides, mice with loss-of-function displayed defects in the sense of smell. Furthermore, we found that the deficiency of CAMSAP2 impairs the classical morphology of MCs, and the CAMSAP2-dependent dendritic remodeling process is responsible for this defect. Thus, our findings demonstrate that CAMSAP2 plays a vital role in regulating the development of MCs.


Assuntos
Dendritos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos , Bulbo Olfatório , Olfato , Animais , Feminino , Masculino , Camundongos , Dendritos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Morfogênese/genética , Bulbo Olfatório/metabolismo , Olfato/fisiologia
15.
PLoS Genet ; 20(6): e1011337, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38935810

RESUMO

Sperm heads contain not only the nucleus but also the acrosome which is a distinctive cap-like structure located anterior to the nucleus and is derived from the Golgi apparatus. The Golgi Associated RAB2 Interactors (GARINs; also known as FAM71) protein family shows predominant expression in the testis and all possess a RAB2-binding domain which confers binding affinity to RAB2, a small GTPase that is responsible for membrane transport and vesicle trafficking. Our previous study showed that GARIN1A and GARIN1B are important for acrosome biogenesis and that GARIN1B is indispensable for male fertility in mice. Here, we generated KO mice of other Garins, namely Garin2, Garin3, Garin4, Garin5a, and Garin5b (Garin2-5b). Using computer-assisted morphological analysis, we found that the loss of each Garin2-5b resulted in aberrant sperm head morphogenesis. While the fertilities of Garin2-/- and Garin4-/- males are normal, Garin5a-/- and Garin5b-/- males are subfertile, and Garin3-/- males are infertile. Further analysis revealed that Garin3-/- males exhibited abnormal acrosomal morphology, but not as severely as Garin1b-/- males; instead, the amounts of membrane proteins, particularly ADAM family proteins, decreased in Garin3 KO spermatozoa. Moreover, only Garin4 KO mice exhibit vacuoles in the sperm head. These results indicate that GARINs assure correct head morphogenesis and some members of the GARIN family function distinctively in male fertility.


Assuntos
Fertilidade , Infertilidade Masculina , Camundongos Knockout , Cabeça do Espermatozoide , Animais , Masculino , Camundongos , Acrossomo/metabolismo , Fertilidade/genética , Complexo de Golgi/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Morfogênese/genética , Proteína rab2 de Ligação ao GTP/metabolismo , Proteína rab2 de Ligação ao GTP/genética , Cabeça do Espermatozoide/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/crescimento & desenvolvimento
16.
Adv Exp Med Biol ; 1441: 645-659, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884739

RESUMO

Tetralogy of Fallot and double-outlet right ventricle are outflow tract (OFT) alignment defects situated on a continuous disease spectrum. A myriad of upstream causes can impact on ventriculoarterial alignment that can be summarized as defects in either i) OFT elongation during looping morphogenesis or ii) OFT remodeling during cardiac septation. Embryological processes underlying these two developmental steps include deployment of second heart field cardiac progenitor cells, establishment and transmission of embryonic left/right information driving OFT rotation and OFT cushion and valve morphogenesis. The formation and remodeling of pulmonary trunk infundibular myocardium is a critical component of both steps. Defects in myocardial, endocardial, or neural crest cell lineages can result in alignment defects, reflecting the complex intercellular signaling events that coordinate arterial pole development. Importantly, however, OFT alignment is mechanistically distinct from neural crest-driven OFT septation, although neural crest cells impact indirectly on alignment through their role in modulating signaling during SHF development. As yet poorly understood nongenetic causes of alignment defects that impact the above processes include hemodynamic changes, maternal exposure to environmental teratogens, and stochastic events. The heterogeneity of causes converging on alignment defects characterizes the OFT as a hotspot of congenital heart defects.


Assuntos
Modelos Animais de Doenças , Dupla Via de Saída do Ventrículo Direito , Transdução de Sinais , Tetralogia de Fallot , Tetralogia de Fallot/genética , Tetralogia de Fallot/patologia , Tetralogia de Fallot/fisiopatologia , Tetralogia de Fallot/embriologia , Animais , Dupla Via de Saída do Ventrículo Direito/genética , Dupla Via de Saída do Ventrículo Direito/patologia , Dupla Via de Saída do Ventrículo Direito/fisiopatologia , Humanos , Crista Neural/metabolismo , Crista Neural/patologia , Crista Neural/embriologia , Morfogênese/genética
17.
PLoS Genet ; 20(6): e1011326, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38857279

RESUMO

The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.


Assuntos
Adesão Celular , Movimento Celular , Proteínas Hedgehog , Miosina Tipo II , Transdução de Sinais , Animais , Camundongos , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Adesão Celular/genética , Miosina Tipo II/metabolismo , Miosina Tipo II/genética , Movimento Celular/genética , Epitélio/metabolismo , Morfogênese/genética , Dente/metabolismo , Dente/crescimento & desenvolvimento , Células Epiteliais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Regulação da Expressão Gênica no Desenvolvimento
18.
J Biosci ; 492024.
Artigo em Inglês | MEDLINE | ID: mdl-38864237

RESUMO

We have extensively described that the neoplastic process (NP) has deep evolutionary roots and we have made specific predictions about the connection between cancer and the formation of the first embryo, which allowed for the evolutionary radiation of metazoans. My main hypothesis is that the NP is at the heart of cellular mechanisms responsible for animal morphogenesis, and given its embryological basis, also at the center of cell differentiation-one of the most interesting and relevant aspects of embryogenesis. In this article, I take forward the idea of the role of physics in the modeling of the neoplastic functional module (NFM) and its contribution to morphogenesis to reveal the totipotency of the zygote. In my consideration of these arguments, I examine mechanical and biophysical clues and their intimate connection with cellular differentiation. I expound on how cancer biology is perfectly intertwined with embryonic differentiation and why it is considered a disease of cell differentiation. The neoplasia is controlled by textural gradients that lead to cell differentiation within the embryo. Thus, the embryo would be a benign tumor. Finally, inspired by evolutionary history and by what the nervous system represents for current biology and based on the impressive nervous system of ctenophores as seen in fossil records, I propose a hypothesis with physical foundations (mechanical morphogenesis) for the formation of a preneural pattern of the nervous system of the first animal embryo.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Morfogênese , Neoplasias , Filogenia , Animais , Morfogênese/genética , Neoplasias/patologia , Neoplasias/genética , Desenvolvimento Embrionário/genética , Humanos , Evolução Biológica , Zigoto/crescimento & desenvolvimento
19.
Genetics ; 227(2)2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38785371

RESUMO

Since the days of Ramón y Cajal, the vast diversity of neuronal and particularly dendrite morphology has been used to catalog neurons into different classes. Dendrite morphology varies greatly and reflects the different functions performed by different types of neurons. Significant progress has been made in our understanding of how dendrites form and the molecular factors and forces that shape these often elaborately sculpted structures. Here, we review work in the nematode Caenorhabditis elegans that has shed light on the developmental mechanisms that mediate dendrite morphogenesis with a focus on studies investigating ciliated sensory neurons and the highly elaborated dendritic trees of somatosensory neurons. These studies, which combine time-lapse imaging, genetics, and biochemistry, reveal an intricate network of factors that function both intrinsically in dendrites and extrinsically from surrounding tissues. Therefore, dendrite morphogenesis is the result of multiple tissue interactions, which ultimately determine the shape of dendritic arbors.


Assuntos
Caenorhabditis elegans , Dendritos , Morfogênese , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/citologia , Dendritos/metabolismo , Morfogênese/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/citologia
20.
Hum Mol Genet ; 33(16): 1420-1428, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38743908

RESUMO

Generalized lymphatic anomaly (GLA) and kaposiform lymphangiomatosis (KLA) are rare congenital disorders that arise through anomalous embryogenesis of the lymphatic system. A somatic activating NRAS p.Q61R variant has been recently detected in GLA and KLA tissues, suggesting that the NRAS p.Q61R variant plays an important role in the development of these diseases. To address this role, we studied the effect of the NRAS p.Q61R variant in lymphatic endothelial cells (LECs) on the structure of the lymphatics during embryonic and postnatal lymphangiogenesis applying inducible, LEC-specific NRAS p.Q61R variant in mice. Lox-stop-Lox NrasQ61R mice were crossed with Prox1-CreERT2 mice expressing tamoxifen-inducible Cre recombinase specifically in LECs. Whole-mount immunostaining of embryonic back skin using an antibody against the LEC surface marker VEGFR3 showed considerably greater lymphatic vessel width in LEC-specific NRAS p.Q61R mutant embryos than in littermate controls. These mutant embryos also showed a significant reduction in the number of lymphatic vessel branches. Furthermore, immunofluorescence staining of whole-mount embryonic back skin using an antibody against the LEC-specific nuclear marker Prox1 showed a large increase in the number of LECs in LEC-specific NRAS p.Q61R mutants. In contrast, postnatal induction of the NRAS p.Q61R variant in LECs did not cause abnormal lymphatic vessel morphogenesis. These results suggest that the NRAS p.Q61R variant in LECs plays a role in development of lymphatic anomalies. While this model does not directly reflect the human pathology of GLA and KLA, there are overlapping features, suggesting that further study of this model may help in studying GLA and KLA mechanisms.


Assuntos
Células Endoteliais , Linfangiogênese , Vasos Linfáticos , Animais , Camundongos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Vasos Linfáticos/embriologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Linfangiogênese/genética , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Mutação , Morfogênese/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Humanos , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Embrião de Mamíferos/metabolismo , Proteínas de Homeodomínio , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA