Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.119
Filter
1.
Curr Top Dev Biol ; 159: 30-58, 2024.
Article in English | MEDLINE | ID: mdl-38729679

ABSTRACT

Morphogenesis from cells to tissue gives rise to the complex architectures that make our organs. How cells and their dynamic behavior are translated into functional spatial patterns is only starting to be understood. Recent advances in quantitative imaging revealed that, although highly heterogeneous, cellular behaviors make reproducible tissue patterns. Emerging evidence suggests that mechanisms of cellular coordination, intrinsic variability and plasticity are critical for robust pattern formation. While pattern development shows a high level of fidelity, tissue organization has undergone drastic changes throughout the course of evolution. In addition, alterations in cell behavior, if unregulated, can cause developmental malformations that disrupt function. Therefore, comparative studies of different species and of disease models offer a powerful approach for understanding how novel spatial configurations arise from variations in cell behavior and the fundamentals of successful pattern formation. In this chapter, I dive into the development of the vertebrate nervous system to explore efforts to dissect pattern formation beyond molecules, the emerging core principles and open questions.


Subject(s)
Nervous System , Vertebrates , Animals , Vertebrates/physiology , Vertebrates/embryology , Nervous System/growth & development , Nervous System/embryology , Body Patterning , Humans , Morphogenesis
2.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38819456

ABSTRACT

Drosophila nervous system development progresses through a series of well-characterized steps in which homeodomain transcription factors (HDTFs) play key roles during most, if not all, phases. Strikingly, although some HDTFs have only one role, many others are involved in multiple steps of the developmental process. Most Drosophila HDTFs engaged in nervous system development are conserved in vertebrates and often play similar roles during vertebrate development. In this Spotlight, we focus on the role of HDTFs during embryogenesis, where they were first characterized.


Subject(s)
Drosophila Proteins , Homeodomain Proteins , Nervous System , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Nervous System/metabolism , Nervous System/embryology , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Drosophila/genetics , Drosophila/metabolism , Drosophila/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
3.
Gene Expr Patterns ; 45: 119257, 2022 09.
Article in English | MEDLINE | ID: mdl-35690356

ABSTRACT

The family of novel transmembrane proteins (TMEM) 132 have been associated with multiple neurological disorders and cancers in humans, but have hardly been studied in vivo. Here we report the expression patterns of the five Tmem132 genes (a, b, c, d and e) in developing mouse nervous system with RNA in situ hybridization in wholemount embryos and tissue sections. Our results reveal differential and partially overlapping expression of multiple Tmem132 family members in both the central and peripheral nervous system, suggesting potential partial redundancy among them.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins , Nervous System , Animals , Humans , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nervous System/embryology
4.
Science ; 375(6582): eabm4459, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35175798

ABSTRACT

The blood-brain barrier (BBB) protects the central nervous system (CNS) from harmful blood-borne factors. Although BBB dysfunction is a hallmark of several neurological disorders, therapies to restore BBB function are lacking. An attractive strategy is to repurpose developmental BBB regulators, such as Wnt7a, into BBB-protective agents. However, safe therapeutic use of Wnt ligands is complicated by their pleiotropic Frizzled signaling activities. Taking advantage of the Wnt7a/b-specific Gpr124/Reck co-receptor complex, we genetically engineered Wnt7a ligands into BBB-specific Wnt activators. In a "hit-and-run" adeno-associated virus-assisted CNS gene delivery setting, these new Gpr124/Reck-specific agonists protected BBB function, thereby mitigating glioblastoma expansion and ischemic stroke infarction. This work reveals that the signaling specificity of Wnt ligands is adjustable and defines a modality to treat CNS disorders by normalizing the BBB.


Subject(s)
Blood-Brain Barrier/physiology , GPI-Linked Proteins/agonists , Glioblastoma/therapy , Receptors, G-Protein-Coupled/agonists , Stroke/therapy , Wnt Proteins/genetics , Wnt Signaling Pathway , Animals , Brain/metabolism , Endothelial Cells/metabolism , Frizzled Receptors/metabolism , Glioblastoma/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mutagenesis , Nervous System/embryology , Protein Engineering , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stroke/metabolism , Wnt Proteins/chemistry , Wnt Proteins/metabolism , Xenopus laevis , Zebrafish
5.
Sci Rep ; 11(1): 19536, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599206

ABSTRACT

Differential expression of cell adhesion molecules in neuronal populations is one of the many mechanisms promoting the formation of functional neural circuits in the developing nervous system. The IgLON family consists of five cell surface immunoglobulin proteins that have been associated with various developmental disorders, such as autism spectrum disorder, schizophrenia, and major depressive disorder. However, there is still limited and fragmented information about their patterns of expression in certain regions of the developing nervous system and how their expression contributes to their function. Utilizing an in situ hybridization approach, we have analyzed the spatiotemporal expression of all IgLON family members in the developing mouse brain, spinal cord, eye, olfactory epithelium, and vomeronasal organ. At one prenatal (E16) and two postnatal (P0 and P15) ages, we show that each IgLON displays distinct expression patterns in the olfactory system, cerebral cortex, midbrain, cerebellum, spinal cord, and eye, indicating that they likely contribute to the wiring of specific neuronal circuitry. These analyses will inform future functional studies aimed at identifying additional roles for these proteins in nervous system development.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Gene Expression Regulation, Developmental , Multigene Family , Nervous System/embryology , Nervous System/metabolism , Neurogenesis/genetics , Animals , Female , In Situ Hybridization , Mice , Organ Specificity/genetics , Pregnancy , RNA, Messenger/genetics
6.
Adv Sci (Weinh) ; 8(23): e2101837, 2021 12.
Article in English | MEDLINE | ID: mdl-34693660

ABSTRACT

Neurovascular disorders, which involve the vascular and nervous systems, are common. Research on such disorders usually focuses on either vascular or nervous components, without looking at how they interact. Adopting a neurovascular perspective is essential to improve current treatments. Therefore, comparing molecular processes known to be involved in both systems separately can provide insight into promising areas of future research. Since development and regeneration share many mechanisms, comparing signaling molecules involved in both the developing vascular and nervous systems and shedding light to those that they have in common can reveal processes, which have not yet been studied from a regenerative perspective, yet hold great potential. Hence, this review discusses and compares processes involved in the development of the vascular and nervous systems, in order to provide an overview of the molecular mechanisms, which are most promising with regards to treatment for neurovascular disorders. Vascular endothelial growth factor, semaphorins, and ephrins are found to hold the most potential, while fibroblast growth factor, bone morphogenic protein, slits, and sonic hedgehog are shown to participate in both the developing vascular and nervous systems, yet have not been studied at the neurovascular level, therefore being of special interest for future research.


Subject(s)
Arteries/embryology , Nervous System/embryology , Regenerative Medicine/methods , Signal Transduction , Veins/embryology , Arteries/metabolism , Ephrins/metabolism , Female , Fibroblast Growth Factors/metabolism , Humans , Nervous System/metabolism , Semaphorins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Veins/metabolism
7.
Cells ; 10(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34572097

ABSTRACT

MT1-MMP/MMP14 belongs to a subgroup of the matrix metalloproteinases family that presents a transmembrane domain, with a cytosolic tail and the catalytic site exposed to the extracellular space. Deficient mice for this enzyme result in early postnatal death and display severe defects in skeletal, muscle and lung development. By using a transgenic line expressing the LacZ reporter under the control of the endogenous Mt1-mmp promoter, we reported a dynamic spatiotemporal expression pattern for Mt1-mmp from early embryonic to perinatal stages during cardiovascular development and brain formation. Thus, Mt1-mmp shows expression in the endocardium of the heart and the truncus arteriosus by E8.5, and is also strongly detected during vascular system development as well as in endothelial cells. In the brain, LacZ reporter expression was detected in the olfactory bulb, the rostral cerebral cortex and the caudal mesencephalic tectum. LacZ-positive cells were observed in neural progenitors of the spinal cord, neural crest cells and the intersomitic region. In the limb, Mt1-mmp expression was restricted to blood vessels, cartilage primordium and muscles. Detection of the enzyme was confirmed by Western blot and immunohistochemical analysis. We suggest novel functions for this metalloproteinase in angiogenesis, endocardial formation and vascularization during organogenesis. Moreover, Mt1-mmp expression revealed that the enzyme may contribute to heart, muscle and brain throughout development.


Subject(s)
Cardiovascular System/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Eye/metabolism , Matrix Metalloproteinase 14/metabolism , Morphogenesis , Nervous System/metabolism , Animals , Cardiovascular System/embryology , Cells, Cultured , Embryo, Mammalian/cytology , Extremities/embryology , Extremities/physiology , Eye/embryology , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Nervous System/embryology
8.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34576153

ABSTRACT

The endocannabinoid system plays a central role in the earliest stages of embryonic, postnatal and adolescent neurodevelopment. Aberrant activity of this system at key developmental phases has been shown to affect neural development. The aim of this review is to synthesise and analyse preclinical insights within rodent populations, focusing on the effects that perinatal (embryonic, gestational and early postnatal developmental stages) and adolescent (postnatal day 21-60) cannabinoid exposure impose across time on the subsequent activity of various drugs of abuse. Results in rodents show that exposure to cannabinoids during the perinatal and adolescent period can lead to multifaceted behavioural and molecular changes. In the perinatal period, significant effects of Δ9-THC exposure on subsequent opiate and amphetamine reward-related behaviours were observed primarily in male rodents. These effects were not extended to include cocaine or alcohol. In adolescence, various cannabinoid agonists were used experimentally. This array of cannabinoids demonstrated consistent effects on opioids across sex. In contrast, no significant effects were observed regarding the future activity of amphetamines and cocaine. However, these studies focused primarily on male rodents. In conclusion, numerous gaps and limitations are apparent in the current body of research. The sparsity of studies analysing the perinatal period must be addressed. Future research within both periods must also focus on delineating sex-specific effects, moving away from a male-centric focus. Studies should also aim to utilise more clinically relevant cannabinoid treatments.


Subject(s)
Cannabinoids/pharmacology , Illicit Drugs/adverse effects , Nervous System/embryology , Animals , Behavior , Humans , Nervous System/drug effects
9.
Cells ; 10(7)2021 06 30.
Article in English | MEDLINE | ID: mdl-34208876

ABSTRACT

During the development of the retina and the nervous system, high levels of energy are required by the axons of retinal ganglion cells (RGCs) to grow towards their brain targets. This energy demand leads to an increase of glycolysis and L-lactate concentrations in the retina. L-lactate is known to be the endogenous ligand of the GPR81 receptor. However, the role of L-lactate and its receptor in the development of the nervous system has not been studied in depth. In the present study, we used immunohistochemistry to show that GPR81 is localized in different retinal layers during development, but is predominantly expressed in the RGC of the adult rodent. Treatment of retinal explants with L-lactate or the exogenous GPR81 agonist 3,5-DHBA altered RGC growth cone (GC) morphology (increasing in size and number of filopodia) and promoted RGC axon growth. These GPR81-mediated modifications of GC morphology and axon growth were mediated by protein kinases A and C, but were absent in explants from gpr81-/- transgenic mice. Living gpr81-/- mice showed a decrease in ipsilateral projections of RGCs to the dorsal lateral geniculate nucleus (dLGN). In conclusion, present results suggest that L-lactate and its receptor GPR81 play an important role in the development of the visual nervous system.


Subject(s)
Lactates/metabolism , Nervous System/embryology , Receptors, G-Protein-Coupled/metabolism , Vision, Ocular/physiology , Animals , Axons/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Growth Cones/metabolism , Mice, Inbred C57BL , Phosphorylation , Protein Kinase C/metabolism , Retina/metabolism , Thalamus/metabolism
10.
Dev Biol ; 478: 183-204, 2021 10.
Article in English | MEDLINE | ID: mdl-34216573

ABSTRACT

The mechanisms regulating nervous system development are still unknown for a wide variety of taxa. In insects and vertebrates, bone morphogenetic protein (BMP) signaling plays a key role in establishing the dorsal-ventral (D-V) axis and limiting the neuroectoderm to one side of that axis, leading to speculation about the conserved evolution of centralized nervous systems. Studies outside of insects and vertebrates show a more diverse picture of what, if any role, BMP signaling plays in neural development across Bilateria. This is especially true in the morphologically diverse Spiralia (≈Lophotrochozoa). Despite several studies of D-V axis formation and neural induction in spiralians, there is no consensus for how these two processes are related, or whether BMP signaling may have played an ancestral role in either process. To determine the function of BMP signaling during early development of the spiralian annelid Capitella teleta, we incubated embryos and larvae in BMP4 protein for different amounts of time. Adding exogenous BMP protein to early-cleaving C. teleta embryos had a striking effect on formation of the brain, eyes, foregut, and ventral midline in a time-dependent manner. However, adding BMP did not block brain or VNC formation or majorly disrupt the D-V axis. We identified three key time windows of BMP activity. 1) BMP treatment around birth of the 3rd-quartet micromeres caused the loss of the eyes, radialization of the brain, and a reduction of the foregut, which we interpret as a loss of A- and C-quadrant identities with a possible trans-fate switch to a D-quadrant identity. 2) Treatment after the birth of micromere 4d induced formation of a third ectopic brain lobe, eye, and foregut lobe, which we interpret as a trans-fate switch of B-quadrant micromeres to a C-quadrant identity. 3) Continuous BMP treatment from late cleavage (4d â€‹+ â€‹12 â€‹h) through mid-larval stages resulted in a modest expansion of Ct-chrdl expression in the dorsal ectoderm and a concomitant loss of the ventral midline (neurotroch ciliary band). Loss of the ventral midline was accompanied by a collapse of the bilaterally-symmetric ventral nerve cord, although the total amount of neural tissue was not greatly affected. Our results compared with those from other annelids and molluscs suggest that BMP signaling was not ancestrally involved in delimiting neural tissue to one region of the D-V axis. However, the effects of ectopic BMP on quadrant-identity during cleavage stages may represent a non-axial organizing signal that was present in the last common ancestor of annelids and mollusks. Furthermore, in the last common ancestor of annelids, BMP signaling may have functioned in patterning ectodermal fates along the D-V axis in the trunk. Ultimately, studies on a wider range of spiralian taxa are needed to determine the role of BMP signaling during neural induction and neural patterning in the last common ancestor of this group. Ultimately, these comparisons will give us insight into the evolutionary origins of centralized nervous systems and body plans.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Proteins/metabolism , Polychaeta/embryology , Polychaeta/metabolism , Zebrafish Proteins/pharmacology , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/genetics , Brain/embryology , Digestive System/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development , Eye/embryology , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Polychaeta/drug effects , Polychaeta/growth & development , Recombinant Proteins/pharmacology , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism
11.
Int J Mol Med ; 48(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34132363

ABSTRACT

The developmental origins of health and disease concept illustrates that exposure in early life to various factors may affect the offspring's long­term susceptibility to disease. During development, the nervous system is sensitive and vulnerable to the environmental insults. Polychlorinated biphenyls (PCBs), which are divided into dioxin­like (DL­PCBs) and non­dioxin­like PCBs (NDL­PCBs), are synthetic persistent environmental endocrine­disrupting chemicals. The toxicological mechanisms of DL­PCBs have been associated with the activation of the aryl hydrocarbon receptor and NDL­PCBs have been associated with ryanodine receptor­mediated calcium ion channels, which affect neuronal migration, promote dendritic growth and alter neuronal connectivity. In addition, PCB accumulation in the placenta destroys the fetal placental unit and affects endocrine function, particularly thyroid hormones and the dopaminergic system, leading to neuroendocrine disorders. However, epidemiological investigations have not achieved a consistent result in different study cohorts. The present review summarizes the epidemiological differences and possible mechanisms of the effects of intrauterine PCB exposure on neurological development.


Subject(s)
Environmental Exposure/adverse effects , Environmental Pollutants/poisoning , Nervous System/drug effects , Polychlorinated Biphenyls/poisoning , Prenatal Exposure Delayed Effects/chemically induced , Female , Humans , Infant, Newborn , Nervous System/embryology , Nervous System Malformations/chemically induced , Nervous System Malformations/diagnosis , Nervous System Malformations/embryology , Pregnancy , Prenatal Exposure Delayed Effects/diagnosis
12.
Dev Biol ; 477: 37-48, 2021 09.
Article in English | MEDLINE | ID: mdl-33991533

ABSTRACT

Ras is the most commonly mutated oncogene in humans and uses three oncogenic effectors: Raf, PI3K, and RalGEF activation of Ral. Understanding the importance of RalGEF>Ral signaling in cancer is hampered by the paucity of knowledge about their function in animal development, particularly in cell movements. We found that mutations that disrupt function of RalGEF or Ral enhance migration phenotypes of mutants for genes with established roles in cell migration. We used as a model the migration of the canal associated neurons (CANs), and validated our results in HSN cell migration, neurite guidance, and general animal locomotion. These functions of RalGEF and Ral are specific to their control of Ral signaling output rather than other published functions of these proteins. In this capacity Ral functions cell autonomously as a permissive developmental signal. In contrast, we observed Ras, the canonical activator of RalGEF>Ral signaling in cancer, to function as an instructive signal. Furthermore, we unexpectedly identified a function for the close Ras relative, Rap1, consistent with activation of RalGEF>Ral. These studies define functions of RalGEF>Ral, Rap1 and Ras signaling in morphogenetic processes that fashion the nervous system. We have also defined a model for studying how small GTPases partner with downstream effectors. Taken together, this analysis defines novel molecules and relationships in signaling networks that control cell movements during development of the nervous system.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Guanine Nucleotide Exchange Factors/physiology , Nervous System/physiopathology , Signal Transduction , ral GTP-Binding Proteins/physiology , ras Proteins/physiology , Animals , CRISPR-Cas Systems , Caenorhabditis elegans/embryology , Embryonic Induction , Genes, ras , Nervous System/embryology , Neurons/physiology , ras Proteins/genetics
13.
Cells ; 10(4)2021 04 20.
Article in English | MEDLINE | ID: mdl-33924211

ABSTRACT

Understanding the autistic brain and the involvement of genetic, non-genetic, and numerous signaling pathways in the etiology and pathophysiology of autism spectrum disorder (ASD) is complex, as is evident from various studies. Apart from multiple developmental disorders of the brain, autistic subjects show a few characteristics like impairment in social communications related to repetitive, restricted, or stereotypical behavior, which suggests alterations in neuronal circuits caused by defects in various signaling pathways during embryogenesis. Most of the research studies on ASD subjects and genetic models revealed the involvement of mutated genes with alterations of numerous signaling pathways like Wnt, hedgehog, and Retinoic Acid (RA). Despite significant improvement in understanding the pathogenesis and etiology of ASD, there is an increasing awareness related to it as well as a need for more in-depth research because no effective therapy has been developed to address ASD symptoms. Therefore, identifying better therapeutic interventions like "novel drugs for ASD" and biomarkers for early detection and disease condition determination are required. This review article investigated various etiological factors as well as the signaling mechanisms and their alterations to understand ASD pathophysiology. It summarizes the mechanism of signaling pathways, their significance, and implications for ASD.


Subject(s)
Autism Spectrum Disorder/pathology , Embryonic Development , Nervous System/embryology , Nervous System/metabolism , Signal Transduction , Animals , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/physiopathology , Humans , Models, Biological
14.
Cells ; 10(4)2021 04 02.
Article in English | MEDLINE | ID: mdl-33918242

ABSTRACT

Bisphenol A (BPA) is considered as one of the most extensively synthesized and used chemicals for industrial and consumer products. Previous investigations have established that exposure to BPA has been linked to developmental, reproductive, cardiovascular, immune, and metabolic effects. Several jurisdictions have imposed restrictions and/or have banned the use of BPA in packaging material and other consumer goods. Hence, manufacturers have replaced BPA with its analogues that have a similar chemical structure. Some of these analogues have shown similar endocrine effects as BPA, while others have not been assessed. In this investigation, we compared the neurodevelopmental effects of BPA and its major replacement Bisphenol F (BPF) on rat fetal neural stem cells (rNSCs). rNSCs were exposed to cell-specific differentiation media with non-cytotoxic doses of BPA or BPF at the range of 0.05 M to 100 M concentrations and measured the degree of cell proliferation, differentiation, and morphometric parameters. Both of these compounds increased cell proliferation and impacted the differentiation rates of oligodendrocytes and neurons, in a concentration-dependent manner. Further, there were concentration-dependent decreases in the maturation of oligodendrocytes and neurons, with a concomitant increase in immature oligodendrocytes and neurons. In contrast, neither BPA nor BPF had any overall effect on cellular proliferation or the cytotoxicity of astrocytes. However, there was a concentration-dependent increase in astrocyte differentiation and morphological changes. Morphometric analysis for the astrocytes, oligodendrocytes, and neurons showed a reduction in the arborization. These data show that fetal rNSCs exposed to either BPA or BPF lead to comparable changes in the cellular differentiation, proliferation, and arborization processes.


Subject(s)
Benzhydryl Compounds/toxicity , Fetus/pathology , Models, Biological , Nervous System/embryology , Neural Stem Cells/drug effects , Phenols/toxicity , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Nervous System/drug effects , Neural Stem Cells/pathology , Neurons/drug effects , Neurons/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Rats
15.
Cells ; 10(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33920757

ABSTRACT

Cholinergic interneurons are "gatekeepers" for striatal circuitry and play pivotal roles in attention, goal-directed actions, habit formation, and behavioral flexibility. Accordingly, perturbations to striatal cholinergic interneurons have been associated with many neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. The role of acetylcholine in many of these disorders is well known, but the use of drugs targeting cholinergic systems fell out of favor due to adverse side effects and the introduction of other broadly acting compounds. However, in response to recent findings, re-examining the mechanisms of cholinergic interneuron dysfunction may reveal key insights into underlying pathogeneses. Here, we provide an update on striatal cholinergic interneuron function, connectivity, and their putative involvement in several disorders. In doing so, we aim to spotlight recurring physiological themes, circuits, and mechanisms that can be investigated in future studies using new tools and approaches.


Subject(s)
Cholinergic Agents/metabolism , Corpus Striatum/pathology , Interneurons/pathology , Mental Disorders/pathology , Nerve Degeneration/pathology , Nervous System/embryology , Animals , Corpus Striatum/physiopathology , Humans , Mental Disorders/physiopathology , Nerve Degeneration/physiopathology , Nervous System/physiopathology
16.
Dev Biol ; 476: 173-188, 2021 08.
Article in English | MEDLINE | ID: mdl-33839113

ABSTRACT

Mouse models of Spina bifida (SB) have been instrumental for identifying genes, developmental processes, and environmental factors that influence neurulation and neural tube closure. Beyond the prominent neural tube defects, other aspects of the nervous system can be affected in SB with significant changes in essential bodily functions such as urination. SB patients frequently experience bladder dysfunction and SB fetuses exhibit reduced density of bladder nerves and smooth muscle although the developmental origins of these deficits have not been determined. The Pax3 Splotch-delayed (Pax3Sp-d) mouse model of SB is one of a very few mouse SB models that survives to late stages of gestation. Through analysis of Pax3Sp-d mutants we sought to define how altered bladder innervation in SB might arise by tracing sacral neural crest (NC) development, pelvic ganglia neuronal differentiation, and assessing bladder nerve fiber density. In Pax3Sp-d/Sp-d fetal mice we observed delayed migration of Sox10+ NC-derived progenitors (NCPs), deficient pelvic ganglia neurogenesis, and reduced density of bladder wall innervation. We further combined NC-specific deletion of Pax3 with the constitutive Pax3Sp-d allele in an effort to generate viable Pax3 mutants to examine later stages of bladder innervation and postnatal bladder function. Neural crest specific deletion of a Pax3 flox allele, using a Sox10-cre driver, in combination with a constitutive Pax3Sp-d mutation produced postnatal viable offspring that exhibited altered bladder function as well as reduced bladder wall innervation and altered connectivity between accessory ganglia at the bladder neck. Combined, the results show that Pax3 plays critical roles within sacral NC that are essential for initiation of neurogenesis and differentiation of autonomic neurons within pelvic ganglia.


Subject(s)
Neural Crest/innervation , PAX3 Transcription Factor/genetics , Urinary Bladder/innervation , Animals , Cell Differentiation/physiology , Disease Models, Animal , Female , Ganglia , Male , Mice/embryology , Mice, Inbred C57BL , Nervous System/embryology , Neural Crest/physiology , Neural Tube Defects/genetics , Neurogenesis , PAX3 Transcription Factor/physiology , Paired Box Transcription Factors/genetics , SOXE Transcription Factors , Sacrococcygeal Region/innervation , Spinal Dysraphism/complications , Spinal Dysraphism/genetics , Urinary Bladder/embryology
17.
Toxicol Appl Pharmacol ; 417: 115477, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33667508

ABSTRACT

N-Ethylpentylone (NEP) is one of the most recent novel stimulants, and there is limited understanding of its toxicity. Here we employed zebrafish model for analyzing the effects of NEP on early embryos and cardiovascular and nervous systems at late developmental stages. We first observed multi-malformations in early embryos and larvae after NEP administration, together with significant deregulations of brain and heart development-associated genes (neurog1, her6, elavl3, nkx2.5, nppa, nppb, tnnt2a) at transcriptional level. Low-dosed NEP treatment induced an anxiety-like phenotype in zebrafish larvae, while higher doses of NEP exerted an inhibitory effect on locomotion and heart rate. Besides, the expression of th (tyrosine hydroxylase) and th2 (tyrosine hydroxylase 2), identifying dopamine (DA) release, were significantly increased during one-hour free swimming after effective low-dosed NEP administration, along with the upregulation of gene fosab and fosb related to stress and anxiety response. D1R antagonist SCH23390 and D2R antagonist sulpiride partially alleviated the aberrances of locomotion and heart rate, indicating dopaminergic receptors were involved in the bidirectional dosage-dependent pattern of NEP-induced performance. Meanwhile, sulpiride offset the upregulated expression of th, th2 and fosab in the group of 1.5 µM NEP, which highlighted the significant role of D2R in NEP-induced locomotive effects. This study systematically described the developmental, neuronal and cardiac toxicity of NEP in zebrafish, and identified the dopaminergic receptors as one of the downstream effectors of NEP administration.


Subject(s)
Benzodioxoles/toxicity , Butylamines/toxicity , Cardiovascular System/drug effects , Dopamine Agonists/toxicity , Dopamine/metabolism , Nervous System/drug effects , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Zebrafish Proteins/agonists , Animals , Animals, Genetically Modified , Cardiovascular System/embryology , Cardiovascular System/metabolism , Female , Gene Expression Regulation, Developmental , Heart Rate/drug effects , Larva/drug effects , Larva/metabolism , Locomotion/drug effects , Male , Nervous System/embryology , Nervous System/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Transcription, Genetic , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
18.
Int J Dev Biol ; 65(4-5-6): 251-261, 2021.
Article in English | MEDLINE | ID: mdl-32930355

ABSTRACT

The development of multicellular organisms involves three main events: differentiation, growth, and morphogenesis. These processes need to be coordinated for a correct developmental program to work. Mechanisms of cell segregation and the formation of boundaries during development play essential roles in this coordination, allowing the generation and maintenance of distinct regions in an organism. These mechanisms are also at work in the nervous system. The process of regionalization involves first the patterning of the developing organism through gradients and the expression of transcription factors in specific regions. Once different tissues have been induced, segregation mechanisms may operate to avoid cell mixing between different compartments. Three mechanisms have been proposed to achieve segregation: (1) differential affinity, which mainly involves the expression of distinct pools of adhesion molecules such as members of the cadherin superfamily; (2) contact inhibition, which is largely mediated by Eph-ephrin signaling; and (3) cortical tension, which involves the actomyosin cytoskeleton. In many instances, these mechanisms collaborate in cell segregation. In the last three decades, there have been several advances in our understanding of how cell segregation and boundaries participate in the development of the nervous system. Interestingly, as in other aspects of development, the molecular players are remarkably similar between vertebrates and invertebrates. Here we summarize the main concepts of cell segregation and boundary formation, focusing on the nervous system and highlighting the similarities between vertebrate and invertebrate model organisms.


Subject(s)
Ephrins , Nervous System/embryology , Organogenesis , Actomyosin , Animals , Invertebrates/embryology , Vertebrates/embryology
19.
Mol Neurobiol ; 58(3): 1062-1073, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33083964

ABSTRACT

The peptidyl-prolyl isomerase Pin1 is a unique enzyme catalyzing the isomerization of the peptide bond between phosphorylated serine-proline or threonine-proline motifs in proteins, thereby regulating a wide spectrum of protein functions, including folding, intracellular signaling, transcription, cell cycle progression, and apoptosis. Pin1 has been reported to act as a key molecular switch inducing cell-type-specific effects, critically depending on the different phosphorylation patterns of its targets within different biological contexts. While its implication in proliferating cells, and, in particular, in the field of cancer, has been widely characterized, less is known about Pin1 biological functions in terminally differentiated and post-mitotic neurons. Notably, Pin1 is widely expressed in the central and peripheral nervous system, where it regulates a variety of neuronal processes, including neuronal development, apoptosis, and synaptic activity. However, despite studies reporting the interaction of Pin1 with neuronal substrates or its involvement in specific signaling pathways, a more comprehensive understanding of its biological functions at neuronal level is still lacking. Besides its implication in physiological processes, a growing body of evidence suggests the crucial involvement of Pin1 in aging and age-related and neurodegenerative diseases, including Alzheimer's disease, Parkinson disease, frontotemporal dementias, Huntington disease, and amyotrophic lateral sclerosis, where it mediates profoundly different effects, ranging from neuroprotective to neurotoxic. Therefore, a more detailed understanding of Pin1 neuronal functions may provide relevant information on the consequences of Pin1 deregulation in age-related and neurodegenerative disorders.


Subject(s)
NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nerve Degeneration/enzymology , Nervous System/embryology , Neurons/enzymology , Neurons/pathology , Signal Transduction , Aging/pathology , Animals , Humans , Nerve Degeneration/pathology
20.
Gen Comp Endocrinol ; 300: 113633, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33031801

ABSTRACT

Pregnancy and lactation are reproductive processes that rely on physiological adaptations that should be timely and adequately triggered to guarantee both maternal and fetal health. Pineal melatonin is a hormone that presents daily and seasonal variations that synchronizes the organism's physiology to the different demands across time through its specific mechanisms and ways of action. The reproductive system is a notable target for melatonin as it actively participates on reproductive physiology and regulates the hypothalamus-pituitary-gonads axis, influencing gonadotropins and sexual hormones synthesis and release. For its antioxidant properties, melatonin is also vital for the oocytes and spermatozoa quality and viability, and for blastocyst development. Maternal pineal melatonin blood levels increase during pregnancy and triggers the maternal physiological alterations in energy metabolism both during pregnancy and lactation to cope with the energy demands of both periods and to promote adequate mammary gland development. Moreover, maternal melatonin freely crosses the placenta and is the only source of this hormone to the fetus. It importantly times the conceptus physiology and influences its development and programing of several functions that depend on neural and brain development, ultimately priming adult behavior and energy and glucose metabolism. The present review aims to explain the above listed melatonin functions, including the potential alterations observed in the progeny gestated under maternal chronodisruption and/or hypomelatoninemia.


Subject(s)
Fetal Development/physiology , Lactation/physiology , Melatonin/metabolism , Pineal Gland/metabolism , Animals , Female , Humans , Mammary Glands, Human/embryology , Nervous System/embryology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...