Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 289
Filtrar
1.
Cell Rep ; 43(10): 114814, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39378153

RESUMEN

Fate determination of neural stem cells (NSCs) is crucial for cortex development and is closely linked to neurodevelopmental disorders when gene expression networks are disrupted. The transcriptional corepressor chromodomain Y-like (CDYL) is widely expressed across diverse cell populations within the human embryonic cortex. However, its precise role in cortical development remains unclear. Here, we show that CDYL is critical for human cortical neurogenesis and that its deficiency leads to a substantial increase in gamma-aminobutyric acid (GABA)-ergic neurons in cortical organoids. Subsequently, neuronatin (NNAT) is identified as a significant target of CDYL, and its abnormal expression obviously influences the fate commitment of cortical NSCs. Cross-species comparisons of CDYL targets unravel a distinct developmental trajectory between human cortical organoids and the mouse cortex at an analogous stage. Collectively, our data provide insight into the evolutionary roles of CDYL in human cortex development, emphasizing its critical function in maintaining the fate of human cortical NSCs.

2.
Cell Rep ; 43(8): 114508, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39018244

RESUMEN

Lissencephaly is a rare brain malformation for which our understanding remains limited due to the absence of suitable animal models that accurately represent human phenotypes. Here, we establish doublecortin (DCX) knockout ferrets as a model that faithfully replicates key features of the disorder. We reveal the critical roles of DCX in neural progenitor cell proliferation and radial glial fiber extension, processes essential for normal cortical development. Utilizing single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics, we provide a detailed atlas of the lissencephalic cortex, illustrating disrupted neuronal lamination and the specific interactions between inhibitory and excitatory neurons. These findings enhance our understanding of the cellular and molecular mechanisms underlying lissencephaly and highlight the potential of DCX knockout ferrets as a valuable tool for neurodevelopmental research, offering insights into both the pathology of lissencephaly and the general principles of brain development.


Asunto(s)
Proteínas de Dominio Doblecortina , Proteína Doblecortina , Hurones , Neurogénesis , Animales , Neurogénesis/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/metabolismo , Neuropéptidos/genética , Lisencefalia/genética , Lisencefalia/patología , Corteza Cerebral/patología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Células-Madre Neurales/metabolismo , Proliferación Celular , Neuronas/metabolismo , Técnicas de Inactivación de Genes
3.
Cell Rep Methods ; 4(7): 100819, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38986613

RESUMEN

Cell reprogramming, which guides the conversion between cell states, is a promising technology for tissue repair and regeneration, with the ultimate goal of accelerating recovery from diseases or injuries. To accomplish this, regulators must be identified and manipulated to control cell fate. We propose Fatecode, a computational method that predicts cell fate regulators based only on single-cell RNA sequencing (scRNA-seq) data. Fatecode learns a latent representation of the scRNA-seq data using a deep learning-based classification-supervised autoencoder and then performs in silico perturbation experiments on the latent representation to predict genes that, when perturbed, would alter the original cell type distribution to increase or decrease the population size of a cell type of interest. We assessed Fatecode's performance using simulations from a mechanistic gene-regulatory network model and scRNA-seq data mapping blood and brain development of different organisms. Our results suggest that Fatecode can detect known cell fate regulators from single-cell transcriptomics datasets.


Asunto(s)
Análisis de la Célula Individual , Análisis de la Célula Individual/métodos , Humanos , Animales , Redes Reguladoras de Genes , Biología Computacional/métodos , Diferenciación Celular/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma , Aprendizaje Profundo , Linaje de la Célula/genética , Ratones , Reprogramación Celular/genética , RNA-Seq/métodos
4.
Cell Rep ; 43(4): 114109, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38613782

RESUMEN

The gut must perform a dual role of protecting the host against toxins and pathogens while harboring mutualistic microbiota. Previous studies suggested that the NADPH oxidase Duox contributes to intestinal homeostasis in Drosophila by producing reactive oxygen species (ROS) in the gut that stimulate epithelial renewal. We find instead that the ROS generated by Duox in the Malpighian tubules leads to the production of Upd3, which enters the gut and stimulates stem cell proliferation. We describe in Drosophila the existence of a countercurrent flow system, which pushes tubule-derived Upd3 to the anterior part of the gut and stimulates epithelial renewal at a distance. Thus, our paper clarifies the role of Duox in gut homeostasis and describes the existence of retrograde fluid flow in the gut, collectively revealing a fascinating example of inter-organ communication.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Mucosa Intestinal , Túbulos de Malpighi , Especies Reactivas de Oxígeno , Animales , Túbulos de Malpighi/metabolismo , Proteínas de Drosophila/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mucosa Intestinal/metabolismo , Drosophila melanogaster/metabolismo , NADPH Oxidasas/metabolismo , Oxidasas Duales/metabolismo , Oxidasas Duales/genética , Proliferación Celular , Homeostasis , Drosophila/metabolismo
5.
Cell Rep ; 43(3): 113873, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38427557

RESUMEN

Craniofacial microsomia (CFM) is a congenital defect that usually results from aberrant development of embryonic pharyngeal arches. However, the molecular basis of CFM pathogenesis is largely unknown. Here, we employ the zebrafish model to investigate mechanisms of CFM pathogenesis. In early embryos, tet2 and tet3 are essential for pharyngeal cartilage development. Single-cell RNA sequencing reveals that loss of Tet2/3 impairs chondrocyte differentiation due to insufficient BMP signaling. Moreover, biochemical and genetic evidence reveals that the sequence-specific 5mC/5hmC-binding protein, Sall4, binds the promoter of bmp4 to activate bmp4 expression and control pharyngeal cartilage development. Mechanistically, Sall4 directs co-phase separation of Tet2/3 with Sall4 to form condensates that mediate 5mC oxidation on the bmp4 promoter, thereby promoting bmp4 expression and enabling sufficient BMP signaling. These findings suggest the TET-BMP-Sall4 regulatory axis is critical for pharyngeal cartilage development. Collectively, our study provides insights into understanding craniofacial development and CFM pathogenesis.


Asunto(s)
Cartílago , Pez Cebra , Animales , Pez Cebra/metabolismo , Cartílago/metabolismo , Diferenciación Celular/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Condrogénesis/genética
6.
Cell Rep ; 43(3): 113926, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38457338

RESUMEN

The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.


Asunto(s)
Vía de Señalización Hippo , Proteínas Serina-Treonina Quinasas , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Diferenciación Celular , Mamíferos/metabolismo
7.
Cell Rep ; 43(4): 114113, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38625792

RESUMEN

The continuous regeneration of spermatogonial stem cells (SSCs) underpins spermatogenesis and lifelong male fertility, but the developmental origins of the SSC pool remain unclear. Here, we document that hnRNPU is essential for establishing the SSC pool. In male mice, conditional loss of hnRNPU in prospermatogonia (ProSG) arrests spermatogenesis and results in sterility. hnRNPU-deficient ProSG fails to differentiate and migrate to the basement membrane to establish SSC pool in infancy. Moreover, hnRNPU deletion leads to the accumulation of ProSG and disrupts the process of T1-ProSG to T2-ProSG transition. Single-cell transcriptional analyses reveal that germ cells are in a mitotically quiescent state and lose their unique identity upon hnRNPU depletion. We further show that hnRNPU could bind to Vrk1, Slx4, and Dazl transcripts that have been identified to suffer aberrant alternative splicing in hnRNPU-deficient testes. These observations offer important insights into SSC pool establishment and may have translational implications for male fertility.


Asunto(s)
Espermatogénesis , Espermatogonias , Animales , Masculino , Ratones , Células Madre Germinales Adultas/metabolismo , Empalme Alternativo/genética , Diferenciación Celular , Espermatogénesis/genética , Espermatogonias/metabolismo , Espermatogonias/citología , Células Madre/metabolismo , Células Madre/citología , Testículo/metabolismo , Testículo/citología , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo
8.
Cell Rep ; 43(4): 114077, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38592974

RESUMEN

Enhancer-derived RNAs (eRNAs) play critical roles in diverse biological processes by facilitating their target gene expression. However, the abundance and function of eRNAs in early embryos are not clear. Here, we present a comprehensive eRNA atlas by systematically integrating publicly available datasets of mouse early embryos. We characterize the transcriptional and regulatory network of eRNAs and show that different embryo developmental stages have distinct eRNA expression and regulatory profiles. Paternal eRNAs are activated asymmetrically during zygotic genome activation (ZGA). Moreover, we identify an eRNA, MZGAe1, which plays an important function in regulating mouse ZGA and early embryo development. MZGAe1 knockdown leads to a developmental block from 2-cell embryo to blastocyst. We create an online data portal, M2ED2, to query and visualize eRNA expression and regulation. Our study thus provides a systematic landscape of eRNA and reveals the important role of eRNAs in regulating mouse early embryo development.


Asunto(s)
Desarrollo Embrionario , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Animales , Desarrollo Embrionario/genética , Ratones , Elementos de Facilitación Genéticos/genética , ARN/metabolismo , ARN/genética , Femenino , Embrión de Mamíferos/metabolismo , Cigoto/metabolismo , Redes Reguladoras de Genes , Masculino
9.
Cell Rep ; 43(10): 114755, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39302835

RESUMEN

Cellular crosstalk is an essential process influenced by numerous factors, including secreted vesicles that transfer nucleic acids, lipids, and proteins between cells. Extracellular vesicles (EVs) have been the center of many studies focusing on neurodegenerative disorders, but whether EVs display cell-type-specific features for cellular crosstalk during neurodevelopment is unknown. Here, using human-induced pluripotent stem cell-derived cerebral organoids, neural progenitors, neurons, and astrocytes, we identify heterogeneity in EV protein content and dynamics in a cell-type-specific and time-dependent manner. Our results support the trafficking of key molecules via EVs in neurodevelopment, such as the transcription factor YAP1, and their localization to differing cell compartments depending on the EV recipient cell type. This study sheds new light on the biology of EVs during human brain development.

10.
Cell Rep ; 43(9): 114683, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39196778

RESUMEN

Developmental patterns of behavior are variably organized in time and among different individuals. However, long-term behavioral diversity was previously studied using pre-defined behavioral parameters, representing a limited fraction of the full individuality structure. Here, we continuously extract ∼1.2 billion body postures of ∼2,200 single C. elegans individuals throughout their full development time to create a complete developmental atlas of stereotyped and individual-unique behavioral spaces. Unsupervised inference of low-dimensional movement modes of each single individual identifies a dynamic developmental trajectory of stereotyped behavioral spaces and exposes unique behavioral trajectories of individuals that deviate from the stereotyped patterns. Moreover, classification of behavioral spaces within tens of neuromodulatory and environmentally perturbed populations shows plasticity in the temporal structures of stereotyped behavior and individuality. These results present a comprehensive atlas of continuous behavioral dynamics across development time and a general framework for unsupervised dissection of shared and unique developmental signatures of behavior.


Asunto(s)
Conducta Animal , Caenorhabditis elegans , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Conducta Estereotipada
11.
Cell Rep ; 43(9): 114760, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39299236

RESUMEN

The composition and cellular heterogeneity of the corpus cavernosum (CC) microenvironment have been characterized, but the spatial heterogeneity at the molecular level remains unexplored. In this study, we integrate single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing to comprehensively chart the spatial cellular landscape of the human and rat CC under normal and disease conditions. We observe differences in the proportions of cell subtypes and marker genes between humans and rats. Based on the analysis of the fibroblast (FB) niche, we also find that the enrichment scores of mechanical force signaling vary across different regions and correlate with the spatial distribution of FB subtypes. In vitro, the soft and hard extracellular matrix (ECM) induces the differentiation of FBs into apolipoprotein (APO)+ FBs and cartilage oligomeric matrix protein (COMP)+ FBs, respectively. In summary, our study provides a cross-species and physiopathological transcriptomic atlas of the CC, contributing to a further understanding of the molecular anatomy and regulation of penile erection.


Asunto(s)
Pene , Análisis de la Célula Individual , Animales , Humanos , Masculino , Pene/metabolismo , Ratas , Matriz Extracelular/metabolismo , Transcriptoma/genética , Fibroblastos/metabolismo , Diferenciación Celular , Ratas Sprague-Dawley
12.
Cell Rep ; 43(6): 114362, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38870008

RESUMEN

Wnt/Wingless (Wg) signaling is critical in development and disease, including cancer. Canonical Wnt signaling is mediated by ß-catenin/Armadillo (Arm in Drosophila) transducing signals to the nucleus, with IFT-A/Kinesin 2 complexes promoting nuclear translocation of ß-catenin/Arm. Here, we demonstrate that a conserved small N-terminal Arm34-87/ß-catenin peptide binds to IFT140, acting as a dominant interference tool to attenuate Wg/Wnt signaling in vivo. Arm34-87 expression antagonizes endogenous Wnt/Wg signaling, resulting in the reduction of its target expression. Arm34-87 inhibits Wg/Wnt signaling by interfering with nuclear translocation of endogenous Arm/ß-catenin, and this can be modulated by levels of wild-type ß-catenin or IFT140, with the Arm34-87 effect being enhanced or suppressed. Importantly, this mechanism is conserved in mammals with the equivalent ß-catenin24-79 peptide blocking nuclear translocation and pathway activation, including in cancer cells. Our work indicates that Wnt signaling can be regulated by a defined N-terminal ß-catenin peptide and thus might serve as an entry point for therapeutic applications to attenuate Wnt/ß-catenin signaling.


Asunto(s)
Proteínas del Dominio Armadillo , Núcleo Celular , Proteínas de Drosophila , Vía de Señalización Wnt , beta Catenina , beta Catenina/metabolismo , Animales , Proteínas de Drosophila/metabolismo , Núcleo Celular/metabolismo , Humanos , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Dominio Armadillo/genética , Proteína Wnt1/metabolismo , Proteína Wnt1/genética , Transporte Activo de Núcleo Celular , Drosophila melanogaster/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Unión Proteica , Secuencia de Aminoácidos , Factores de Transcripción
13.
Cell Rep ; 43(5): 114123, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38635399

RESUMEN

Although oligodendrocytes (OLs) synthesize laminin-γ1, the most widely used γ subunit, its functional significance in the CNS remains unknown. To answer this important question, we generated a conditional knockout mouse line with laminin-γ1 deficiency in OL lineage cells (γ1-OKO). γ1-OKO mice exhibit weakness/paralysis and die by post-natal day 33. Additionally, they develop blood-brain barrier (BBB) disruption in the cortex and striatum. Subsequent studies reveal decreased major facilitator superfamily domain containing 2a expression and increased endothelial caveolae vesicles, but unaltered tight junction protein expression and tight junction ultrastructure, indicating a transcellular, rather than a paracellular, mechanism of BBB breakdown. Furthermore, significantly reduced OL lineage cells, OL precursor cells (OPCs), proliferating OPCs, and mature OLs are observed in γ1-OKO brains in a region-specific manner. Consistent with this finding, various defects in myelination are detected in γ1-OKO brains at biochemical and ultrastructural levels. Overall, these results highlight important roles of OL-derived laminin-γ1 in BBB maintenance and OL biology (proliferation, differentiation, and myelination).


Asunto(s)
Barrera Hematoencefálica , Laminina , Ratones Noqueados , Vaina de Mielina , Oligodendroglía , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Laminina/metabolismo , Oligodendroglía/metabolismo , Ratones , Vaina de Mielina/metabolismo , Diferenciación Celular , Proliferación Celular , Sistema Nervioso Central/metabolismo , Linaje de la Célula
14.
Cell Rep ; 43(5): 114168, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38700981

RESUMEN

The first 1,000 days of human life lay the foundation for brain development and later cognitive growth. However, the developmental rules of the functional connectome during this critical period remain unclear. Using high-resolution, longitudinal, task-free functional magnetic resonance imaging data from 930 scans of 665 infants aged 28 postmenstrual weeks to 3 years, we report the early maturational process of connectome segregation and integration. We show the dominant development of local connections alongside a few global connections, the shift of brain hubs from primary regions to high-order association cortices, the developmental divergence of network segregation and integration along the anterior-posterior axis, the prediction of neurocognitive outcomes, and their associations with gene expression signatures of microstructural development and neuronal metabolic pathways. These findings advance our understanding of the principles of connectome remodeling during early life and its neurobiological underpinnings and have implications for studying typical and atypical development.


Asunto(s)
Encéfalo , Conectoma , Imagen por Resonancia Magnética , Humanos , Lactante , Masculino , Femenino , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Preescolar , Red Nerviosa/fisiología , Recién Nacido
15.
Cell Rep ; 43(7): 114398, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38935502

RESUMEN

Mechanosensitive Piezo channels regulate cell division, cell extrusion, and cell death. However, systems-level functions of Piezo in regulating organogenesis remain poorly understood. Here, we demonstrate that Piezo controls epithelial cell topology to ensure precise organ growth by integrating live-imaging experiments with pharmacological and genetic perturbations and computational modeling. Notably, the knockout or knockdown of Piezo increases bilateral asymmetry in wing size. Piezo's multifaceted functions can be deconstructed as either autonomous or non-autonomous based on a comparison between tissue-compartment-level perturbations or between genetic perturbation populations at the whole-tissue level. A computational model that posits cell proliferation and apoptosis regulation through modulation of the cutoff tension required for Piezo channel activation explains key cell and tissue phenotypes arising from perturbations of Piezo expression levels. Our findings demonstrate that Piezo promotes robustness in regulating epithelial topology and is necessary for precise organ size control.


Asunto(s)
Células Epiteliales , Canales Iónicos , Canales Iónicos/metabolismo , Canales Iónicos/genética , Animales , Tamaño de los Órganos , Células Epiteliales/metabolismo , Ratones , Proliferación Celular , Alas de Animales/metabolismo , Alas de Animales/crecimiento & desarrollo , Apoptosis , Humanos , Epitelio/metabolismo
16.
Cell Rep ; 43(6): 114302, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38824644

RESUMEN

Resident cardiac macrophages are critical mediators of cardiac function. Despite their known importance to cardiac electrophysiology and tissue maintenance, there are currently no stem-cell-derived models of human engineered cardiac tissues (hECTs) that include resident macrophages. In this study, we made an induced pluripotent stem cell (iPSC)-derived hECT model with a resident population of macrophages (iM0) to better recapitulate the native myocardium and characterized their impact on tissue function. Macrophage retention within the hECTs was confirmed via immunofluorescence after 28 days of cultivation. The inclusion of iM0s significantly impacted hECT function, increasing contractile force production. A potential mechanism underlying these changes was revealed by the interrogation of calcium signaling, which demonstrated the modulation of ß-adrenergic signaling in +iM0 hECTs. Collectively, these findings demonstrate that macrophages significantly enhance cardiac function in iPSC-derived hECT models, emphasizing the need to further explore their contributions not only in healthy hECT models but also in the contexts of disease and injury.


Asunto(s)
Células Madre Pluripotentes Inducidas , Macrófagos , Contracción Miocárdica , Ingeniería de Tejidos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Macrófagos/metabolismo , Ingeniería de Tejidos/métodos , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Miocardio/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Diferenciación Celular , Señalización del Calcio
17.
Cell Rep ; 43(4): 114049, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573853

RESUMEN

Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.


Asunto(s)
Linfangiogénesis , Células Madre Mesenquimatosas , Osificación Heterotópica , Factor C de Crecimiento Endotelial Vascular , Animales , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Osificación Heterotópica/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Ratones , Células Madre Mesenquimatosas/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Diferenciación Celular , Tenocitos/metabolismo , Osteogénesis , Haploinsuficiencia , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino
18.
Cell Rep ; 43(3): 113889, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38416646

RESUMEN

The mystery of appendage regeneration has fascinated humans for centuries, while the underlying regulatory mechanisms remain unclear. In this study, we establish a transcriptional landscape of regenerating leg in the American cockroach, Periplaneta americana, an ideal model in appendage regeneration studies showing remarkable regeneration capacity. Through a large-scale in vivo screening, we identify multiple signaling pathways and transcription factors controlling leg regeneration. Specifically, zfh-2 and bowl contribute to blastema cell proliferation and morphogenesis in two transcriptional cascades: bone morphogenetic protein (BMP)/JAK-STAT-zfh-2-B-H2 and Notch-drm/bowl-bab1. Notably, we find zfh-2 is working as a direct target of BMP signaling to promote cell proliferation in the blastema. These mechanisms might be conserved in the appendage regeneration of vertebrates from an evolutionary perspective. Overall, our findings reveal that two crucial transcriptional cascades orchestrate distinct cockroach leg regeneration processes, significantly advancing the comprehension of molecular mechanism in appendage regeneration.


Asunto(s)
Cucarachas , Animales , Humanos , Factores de Transcripción , Morfogénesis
19.
Cell Rep ; 43(3): 113955, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38507414

RESUMEN

Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.


Asunto(s)
Tejido Adiposo Beige , Pez Cebra , Animales , Humanos , Anciano , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Epicárdico , Tejido Adiposo/metabolismo , Pericardio/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo
20.
Cell Rep ; 43(8): 114556, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39096491

RESUMEN

Early caregiving adversity (ECA) is associated with social behavior deficits and later development of psychopathology. However, the infant neural substrates of ECA are poorly understood. The lateral habenula (LHb), a highly conserved brain region with consistent links to adult psychopathology, is understudied in development, when the brain is most vulnerable to environmental impacts. Here, we describe the structural and functional ontogeny of the LHb and its behavioral role in infant and juvenile rat pups. We show that the LHb promotes a developmental transition in social approach behavior under threat as typically reared infants mature. By contrast, we show that ECA disrupts habenular ontogeny, including volume, protein expression, firing properties, and corticohabenular connectivity. Furthermore, inhibiting a specific corticohabenular projection rescues infant social approach deficits following ECA. Together, these results identify immediate biomarkers of ECA in the LHb and highlight this region as a site of early social processing and behavior control.


Asunto(s)
Habénula , Conducta Social , Animales , Habénula/metabolismo , Ratas , Masculino , Femenino , Conducta Animal , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA