Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Evol ; 98(2): 61-75, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36574764

RESUMO

The amygdala is a complex brain structure in the vertebrate telencephalon, essential for regulating social behaviors, emotions, and (social) cognition. In contrast to the vast majority of neuron types described in the many nuclei of the mammalian amygdala, little is known about the neuronal diversity in non-mammals, making reconstruction of its evolution particularly difficult. Here, we characterize glutamatergic neuron types in the amygdala of the urodele amphibian Pleurodeles waltl. Our single-cell RNA sequencing data indicate the existence of at least ten distinct types and subtypes of glutamatergic neurons in the salamander amygdala. These neuron types are molecularly distinct from neurons in the ventral pallium (VP), suggesting that the pallial amygdala and the VP are two separate areas in the telencephalon. In situ hybridization for marker genes indicates that amygdalar glutamatergic neuron types are located in three major subdivisions: the lateral amygdala, the medial amygdala, and a newly defined area demarcated by high expression of the transcription factor Sim1. The gene expression profiles of these neuron types suggest similarities with specific neurons in the sauropsid and mammalian amygdala. In particular, we identify Sim1+ and Sim1+ Otp+ expressing neuron types, potentially homologous to the mammalian nucleus of the lateral olfactory tract (NLOT) and to hypothalamic-derived neurons of the medial amygdala, respectively. Taken together, our results reveal a surprising diversity of glutamatergic neuron types in the amygdala of salamanders, despite the anatomical simplicity of their brain. These results offer new insights on the cellular and anatomical complexity of the amygdala in tetrapod ancestors.


Assuntos
Tonsila do Cerebelo , Urodelos , Animais , Urodelos/metabolismo , Tonsila do Cerebelo/metabolismo , Fatores de Transcrição/genética , Telencéfalo/metabolismo , Neurônios/metabolismo , Mamíferos/metabolismo
2.
Nat Commun ; 13(1): 7392, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36450803

RESUMO

Octopuses are mollusks that have evolved intricate neural systems comparable with vertebrates in terms of cell number, complexity and size. The brain cell types that control their sophisticated behavioral repertoire are still unknown. Here, we profile the cell diversity of the paralarval Octopus vulgaris brain to build a cell type atlas that comprises mostly neural cells, but also multiple glial subtypes, endothelial cells and fibroblasts. We spatially map cell types to the vertical, subesophageal and optic lobes. Investigation of cell type conservation reveals a shared gene signature between glial cells of mouse, fly and octopus. Genes related to learning and memory are enriched in vertical lobe cells, which show molecular similarities with Kenyon cells in Drosophila. We construct a cell type taxonomy revealing transcriptionally related cell types, which tend to appear in the same brain region. Together, our data sheds light on cell type diversity and evolution in the octopus brain.


Assuntos
Octopodiformes , Animais , Camundongos , Octopodiformes/genética , Células Endoteliais , Encéfalo , Alimentos Marinhos , Neuroglia , Drosophila
3.
Science ; 377(6610): eabp9186, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-36048957

RESUMO

The evolution of advanced cognition in vertebrates is associated with two independent innovations in the forebrain: the six-layered neocortex in mammals and the dorsal ventricular ridge (DVR) in sauropsids (reptiles and birds). How these innovations arose in vertebrate ancestors remains unclear. To reconstruct forebrain evolution in tetrapods, we built a cell-type atlas of the telencephalon of the salamander Pleurodeles waltl. Our molecular, developmental, and connectivity data indicate that parts of the sauropsid DVR trace back to tetrapod ancestors. By contrast, the salamander dorsal pallium is devoid of cellular and molecular characteristics of the mammalian neocortex yet shares similarities with the entorhinal cortex and subiculum. Our findings chart the series of innovations that resulted in the emergence of the mammalian six-layered neocortex and the sauropsid DVR.


Assuntos
Evolução Biológica , Neurônios , Pleurodeles , Telencéfalo , Animais , Atlas como Assunto , Neocórtex/citologia , Neocórtex/fisiologia , Neurônios/metabolismo , Pleurodeles/fisiologia , Telencéfalo/citologia , Telencéfalo/fisiologia , Transcriptoma
4.
Elife ; 102021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34425939

RESUMO

Cephalopods have evolved nervous systems that parallel the complexity of mammalian brains in terms of neuronal numbers and richness in behavioral output. How the cephalopod brain develops has only been described at the morphological level, and it remains unclear where the progenitor cells are located and what molecular factors drive neurogenesis. Using histological techniques, we located dividing cells, neural progenitors and postmitotic neurons in Octopus vulgaris embryos. Our results indicate that an important pool of progenitors, expressing the conserved bHLH transcription factors achaete-scute or neurogenin, is located outside the central brain cords in the lateral lips adjacent to the eyes, suggesting that newly formed neurons migrate into the cords. Lineage-tracing experiments then showed that progenitors, depending on their location in the lateral lips, generate neurons for the different lobes, similar to the squid Doryteuthis pealeii. The finding that octopus newborn neurons migrate over long distances is reminiscent of vertebrate neurogenesis and suggests it might be a fundamental strategy for large brain development.


Octopuses have evolved incredibly large and complex nervous systems that allow them to perform impressive behaviors, like plan ahead, navigate and solve puzzles. The nervous system of the common octopus (also known as Octopus vulgaris) contains over half a billion nerves cells called neurons, similar to the number found in small primates. Two thirds of these cells reside in the octopuses' arms, while the rest make-up a central brain that sits between their eyes. Very little is known about how this central brain forms in the embryo, including where the cells originate and which molecular factors drive their maturation in to adult cells. To help answer these questions, Deryckere et al. studied the brain of Octopus vulgaris at different stages of early development using various cell staining and imaging techniques. The experiments identified an important pool of dividing cells which sit in an area outside the central brain called the 'lateral lips'. In these cells, genes known to play a role in neural development in other animals are active, indicating that the cells had not reached their final, mature state. In contrast, the central brain did not seem to contain any of these immature cells at the point when it was growing the most. To investigate this further, Deryckere et al. used fluorescent markers to track the progeny of the dividing cells during development. This revealed that cells in the lateral lips take on a specific neuronal fate before migrating to their target region in the central brain. Newly matured neurons have also been shown to travel large distances in the embryos of vertebrates, suggesting that this mechanism may be a common strategy for building large, complex brains. Although the nervous system of the common octopus is comparable to mammals, they evolved from a very distant branch of the tree of life; indeed, their last common ancestor was a worm-like animal that lived about 600 million years ago. Studying the brain of the common octopus, as done here, could therefore provide new insights into how complex nervous systems, including our own, evolved over time.


Assuntos
Migração Animal , Encéfalo/crescimento & desenvolvimento , Movimento Celular , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Octopodiformes/anatomia & histologia , Octopodiformes/fisiologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Encéfalo/citologia , Encéfalo/fisiologia
5.
BMC Dev Biol ; 20(1): 7, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32299349

RESUMO

BACKGROUND: Octopus vulgaris has been an iconic cephalopod species for neurobiology research as well as for cephalopod aquaculture. It is one of the most intelligent and well-studied invertebrates, possessing both long- and short-term memory and the striking ability to perform complex cognitive tasks. Nevertheless, how the common octopus developed these uncommon features remains enigmatic. O. vulgaris females spawn thousands of small eggs and remain with their clutch during their entire development, cleaning, venting and protecting the eggs. In fact, eggs incubated without females usually do not develop normally, mainly due to biological contamination (fungi, bacteria, etc.). This high level of parental care might have hampered laboratory research on the embryonic development of this intriguing cephalopod. RESULTS: Here, we present a completely parameter-controlled artificial seawater standalone egg incubation system that replaces maternal care and allows successful embryonic development of a small-egged octopus species until hatching in a laboratory environment. We also provide a practical and detailed staging atlas based on bright-field and light sheet fluorescence microscopy imaging for precise monitoring of embryonic development. The atlas has a comparative section to benchmark stages to the different scales published by Naef (1928), Arnold (1965) and Boletzky (2016). Finally, we provide methods to monitor health and wellbeing of embryos during organogenesis. CONCLUSION: Besides introducing the study of O. vulgaris embryonic development to a wider community, this work can be a high-quality reference for comparative evolutionary developmental biology.


Assuntos
Octopodiformes/embriologia , Animais , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Microscopia de Fluorescência
6.
Development ; 147(10)2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32253238

RESUMO

The transcription factor Zeb2 controls fate specification and subsequent differentiation and maturation of multiple cell types in various embryonic tissues. It binds many protein partners, including activated Smad proteins and the NuRD co-repressor complex. How Zeb2 subdomains support cell differentiation in various contexts has remained elusive. Here, we studied the role of Zeb2 and its domains in neurogenesis and neural differentiation in the young postnatal ventricular-subventricular zone (V-SVZ), in which neural stem cells generate olfactory bulb-destined interneurons. Conditional Zeb2 knockouts and separate acute loss- and gain-of-function approaches indicated that Zeb2 is essential for controlling apoptosis and neuronal differentiation of V-SVZ progenitors before and after birth, and we identified Sox6 as a potential downstream target gene of Zeb2. Zeb2 genetic inactivation impaired the differentiation potential of the V-SVZ niche in a cell-autonomous fashion. We also provide evidence that its normal function in the V-SVZ also involves non-autonomous mechanisms. Additionally, we demonstrate distinct roles for Zeb2 protein-binding domains, suggesting that Zeb2 partners co-determine neuronal output from the mouse V-SVZ in both quantitative and qualitative ways in early postnatal life.


Assuntos
Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Neurogênese/genética , Bulbo Olfatório/embriologia , Bulbo Olfatório/crescimento & desenvolvimento , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo , Animais , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Técnicas de Inativação de Genes , Interneurônios/metabolismo , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Bulbo Olfatório/metabolismo , Fatores de Transcrição SOXD/metabolismo , Transdução de Sinais/imunologia , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética
7.
Arterioscler Thromb Vasc Biol ; 40(4): e87-e104, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32078368

RESUMO

OBJECTIVE: Impaired ALK1 (activin receptor-like kinase-1)/Endoglin/BMP9 (bone morphogenetic protein 9) signaling predisposes to arteriovenous malformations (AVMs). Activation of SMAD1/5 signaling can be enhanced by shear stress. In the genetic disease hereditary hemorrhagic telangiectasia, which is characterized by arteriovenous malformations, the affected receptors are those involved in the activation of mechanosensitive SMAD1/5 signaling. To elucidate how genetic and mechanical signals interact in AVM development, we sought to identify targets differentially regulated by BMP9 and shear stress. Approach and Results: We identify Cx37 (Connexin37) as a differentially regulated target of ligand-induced and mechanotransduced SMAD1/5 signaling. We show that stimulation of endothelial cells with BMP9 upregulated Cx37, whereas shear stress inhibited this expression. This signaling was SMAD1/5-dependent, and in the absence of SMAD1/5, there was an inversion of the expression pattern. Ablated SMAD1/5 signaling alone caused AVM-like vascular malformations directly connecting the dorsal aorta to the inlet of the heart. In yolk sacs of mouse embryos with an endothelial-specific compound heterozygosity for SMAD1/5, addition of TNFα (tumor necrosis factor-α), which downregulates Cx37, induced development of these direct connections bypassing the yolk sac capillary bed. In wild-type embryos undergoing vascular remodeling, Cx37 was globally expressed by endothelial cells but was absent in regions of enlarging vessels. TNFα and endothelial-specific compound heterozygosity for SMAD1/5 caused ectopic regions lacking Cx37 expression, which correlated to areas of vascular malformations. Mechanistically, loss of Cx37 impairs correct directional migration under flow conditions. CONCLUSIONS: Our data demonstrate that Cx37 expression is differentially regulated by shear stress and SMAD1/5 signaling, and that reduced Cx37 expression is permissive for capillary enlargement into shunts.


Assuntos
Malformações Arteriovenosas/genética , Conexinas/genética , Regulação para Baixo , Mecanotransdução Celular , Proteína Smad1/genética , Proteína Smad5/genética , Regulação para Cima , Receptores de Activinas Tipo II/metabolismo , Animais , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/patologia , Capilares/patologia , Células Cultivadas , Conexinas/metabolismo , Embrião de Mamíferos , Endoglina/metabolismo , Células Endoteliais/metabolismo , Feminino , Fator 2 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Camundongos Knockout , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Remodelação Vascular
8.
Sci Rep ; 9(1): 8969, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31222074

RESUMO

To understand traumas to the nervous system, the relation between mechanical load and functional impairment needs to be explained. Cellular-level computational models are being used to capture the mechanism behind mechanically-induced injuries and possibly predict these events. However, uncertainties in the material properties used in computational models undermine the validity of their predictions. For this reason, in this study the squid giant axon was used as a model to provide a description of the axonal mechanical behavior in a large strain and high strain rate regime [Formula: see text], which is relevant for injury investigations. More importantly, squid giant axon membrane sheaths were isolated and tested under dynamic uniaxial tension and relaxation. From the lumen outward, the membrane sheath presents: an axolemma, a layer of Schwann cells followed by the basement membrane and a prominent layer of loose connective tissue consisting of fibroblasts and collagen. Our results highlight the load-bearing role of this enwrapping structure and provide a constitutive description that could in turn be used in computational models. Furthermore, tests performed on collagen-depleted membrane sheaths reveal both the substantial contribution of the endoneurium to the total sheath's response and an interesting increase in material nonlinearity when the collagen in this connective layer is digested. All in all, our results provide useful insights for modelling the axonal mechanical response and in turn will lead to a better understanding of the relationship between mechanical insult and electrophysiological outcome.


Assuntos
Axônios/fisiologia , Membrana Celular/metabolismo , Decapodiformes/fisiologia , Fenômenos Mecânicos , Bainha de Mielina/metabolismo , Algoritmos , Animais , Modelos Teóricos
9.
Front Physiol ; 9: 1160, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30246785

RESUMO

Within the clade of mollusks, cephalopods have developed an unusually large and complex nervous system. The increased complexity of the cephalopod centralized "brain" parallels an amazing amount of complex behaviors that culminate in one order, the octopods. The mechanisms that enable evolution of expanded brains in invertebrates remain enigmatic. While expression mapping of known molecular pathways demonstrated the conservation of major neurogenesis pathways and revealed neurogenic territories, it did not explain why cephalopods could massively increase their brain size compared to other mollusks. Such an increase is reminiscent of the expansion of the cerebral cortex in mammalians, which have enlarged their number and variety of neurogenic stem cells. We hypothesize that similar mechanisms might be at play in cephalopods and that focusing on the stem cell biology of cephalopod neurogenesis and genetic innovations might be smarter strategies to uncover the mechanism that has driven cephalopod brain expansion.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...