Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37.264
Filtrar
1.
Curr Top Membr ; 93: 85-116, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39181579

RESUMO

Lysosomes are more than just cellular recycling bins; they play a crucial role in regulating key cellular functions. Proper lysosomal function is essential for growth pathway regulation, cell proliferation, and metabolic homeostasis. Impaired lysosomal function is associated with lipid storage disorders and neurodegenerative diseases. Lysosomes form extensive and dynamic close contacts with the membranes of other organelles, including the endoplasmic reticulum, mitochondria, peroxisomes, and lipid droplets. These membrane contacts sites (MCSs) are vital for many lysosomal functions. In this chapter, we will explore lysosomal MCSs focusing on the machinery that mediates these contacts, how they are regulated, and their functional implications on physiology and pathology.


Assuntos
Comunicação Celular , Homeostase , Lisossomos , Lisossomos/metabolismo , Humanos , Animais , Membranas Intracelulares/metabolismo
2.
Nat Commun ; 15(1): 7101, 2024 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-39155292

RESUMO

The inference of cell-cell communication (CCC) is crucial for a better understanding of complex cellular dynamics and regulatory mechanisms in biological systems. However, accurately inferring spatial CCCs at single-cell resolution remains a significant challenge. To address this issue, we present a versatile method, called DeepTalk, to infer spatial CCC at single-cell resolution by integrating single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data. DeepTalk utilizes graph attention network (GAT) to integrate scRNA-seq and ST data, which enables accurate cell-type identification for single-cell ST data and deconvolution for spot-based ST data. Then, DeepTalk can capture the connections among cells at multiple levels using subgraph-based GAT, and further achieve spatially resolved CCC inference at single-cell resolution. DeepTalk achieves excellent performance in discovering meaningful spatial CCCs on multiple cross-platform datasets, which demonstrates its superior ability to dissect cellular behavior within intricate biological processes.


Assuntos
Comunicação Celular , Análise de Célula Única , Transcriptoma , Análise de Célula Única/métodos , Humanos , Perfilação da Expressão Gênica/métodos , Análise de Sequência de RNA/métodos , Animais , Algoritmos , Biologia Computacional/métodos
3.
Gene ; 929: 148820, 2024 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-39103059

RESUMO

BACKGROUND: Abdominal aortic aneurysm (AAA) is a complex vascular disorder characterized by the progressive dilation of the abdominal aorta, with a high risk of rupture and mortality. Understanding the cellular interactions and molecular mechanisms underlying AAA development is critical for identifying potential therapeutic targets. METHODS: This study utilized datasets GSE197748, GSE164678 and GSE183464 from the GEO database, encompassing bulk and single-cell RNA sequencing data from AAA and control samples. We performed principal component analysis, differential expression analysis, and functional enrichment analysis to identify key pathways involved in AAA. Cell-cell interactions were investigated using CellPhoneDB, focusing on fibroblasts, vascular smooth muscle cells (VSMCs), and macrophages. We further validated our findings using a mouse model of AAA induced by porcine pancreatic enzyme infusion, followed by gene expression analysis and co-immunoprecipitation experiments. RESULTS: Our analysis revealed significant alterations in gene expression profiles between AAA and control samples, with a pronounced immune response and cell adhesion pathways being implicated. Single-cell RNA sequencing data highlighted an increased proportion of pro-inflammatory macrophages, along with changes in the composition of fibroblasts and VSMCs in AAA. CellPhoneDB analysis identified critical ligand-receptor interactions, notably collagen type I alpha 1 chain (COL1A1)/COL1A2-CD18 and thrombospondin 1 (THBS1)-CD3, suggesting complex communication networks between fibroblasts and VSMCs. In vivo experiments confirmed the upregulation of these genes in AAA mice and demonstrated the functional interaction between COL1A1/COL1A2 and CD18. CONCLUSION: The interaction between fibroblasts and VSMCs, mediated by specific ligand-receptor pairs such as COL1A1/COL1A2-CD18 and THBS1-CD3, plays a pivotal role in AAA pathogenesis.


Assuntos
Aneurisma da Aorta Abdominal , Músculo Liso Vascular , Análise de Sequência de RNA , Análise de Célula Única , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/metabolismo , Animais , Camundongos , Análise de Célula Única/métodos , Humanos , Análise de Sequência de RNA/métodos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Macrófagos/metabolismo , Progressão da Doença , Fibroblastos/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Perfilação da Expressão Gênica/métodos , Comunicação Celular/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo
4.
Int J Nanomedicine ; 19: 8211-8236, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39157736

RESUMO

Hypoxia, as a prominent feature of the tumor microenvironment, has a profound impact on the multicomponent changes within this environment. Under hypoxic conditions, the malignant phenotype of tumor cells, the variety of cell types within the tumor microenvironment, as well as intercellular communication and material exchange, undergo complex alterations. These changes provide significant prospects for exploring the mechanisms of tumor development under different microenvironmental conditions and for devising therapeutic strategies. Exosomes secreted by tumor cells and stromal cells are integral components of the tumor microenvironment, serving as crucial mediators of intercellular communication and material exchange, and have consequently garnered increasing attention from researchers. This review focuses on the mechanisms by which hypoxic conditions promote the release of exosomes by tumor cells and alter their encapsulated contents. It also examines the effects of exosomes derived from tumor cells, immune cells, and other cell types under hypoxic conditions on the tumor microenvironment. Additionally, we summarize current research progress on the potential clinical applications of exosomes under hypoxic conditions and propose future research directions in this field.


Assuntos
Comunicação Celular , Exossomos , Neoplasias , Microambiente Tumoral , Exossomos/metabolismo , Humanos , Comunicação Celular/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Hipóxia Celular/fisiologia , Hipóxia Tumoral , Hipóxia/metabolismo
5.
Clin Transl Med ; 14(8): e1812, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39152680

RESUMO

The liver possesses a distinctive capacity for regeneration within the human body. Under normal circumstances, liver cells replicate themselves to maintain liver function. Compensatory replication of healthy hepatocytes is sufficient for the regeneration after acute liver injuries. In the late stage of chronic liver damage, a large number of hepatocytes die and hepatocyte replication is blocked. Liver regeneration has more complex mechanisms, such as the transdifferentiation between cell types or hepatic progenitor cells mediated. Dysregulation of liver regeneration causes severe chronic liver disease. Gaining a more comprehensive understanding of liver regeneration mechanisms would facilitate the advancement of efficient therapeutic approaches. This review provides an overview of the signalling pathways linked to different aspects of liver regeneration in various liver diseases. Moreover, new knowledge on cellular interactions during the regenerative process is also presented. Finally, this paper explores the potential applications of new technologies, such as nanotechnology, stem cell transplantation and organoids, in liver regeneration after injury, offering fresh perspectives on treating liver disease.


Assuntos
Regeneração Hepática , Regeneração Hepática/fisiologia , Humanos , Hepatopatias/terapia , Hepatopatias/fisiopatologia , Comunicação Celular/fisiologia , Fígado/lesões , Hepatócitos/metabolismo , Transdução de Sinais , Animais
6.
Front Immunol ; 15: 1408415, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39148736

RESUMO

Exosomes play a crucial role in various biological processes, such as human development, immune responses, and disease occurrence. The membrane proteins on exosomes are pivotal factors for their biological functionality. Currently, numerous membrane proteins have been identified on exosome membranes, participating in intercellular communication, mediating target cell recognition, and regulating immune processes. Furthermore, membrane proteins from exosomes derived from cancer cells can serve as relevant biomarkers for early cancer diagnosis. This article provides a comprehensive review of the composition of exosome membrane proteins and their diverse functions in the organism's biological processes. Through in-depth exploration of exosome membrane proteins, it is expected to offer essential foundations for the future development of novel biomedical diagnostics and therapies.


Assuntos
Exossomos , Proteínas de Membrana , Exossomos/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Animais , Neoplasias/imunologia , Neoplasias/metabolismo , Comunicação Celular , Biomarcadores Tumorais/metabolismo , Biomarcadores
7.
Methods Mol Biol ; 2828: 23-36, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39147967

RESUMO

Cell-cell interaction mediated by secreted and adhesive signaling molecules forms the basis of the coordinated cell movements (i.e., collective cell migration) observed in developing embryos, regenerating tissues, immune cells, and metastatic cancer. Decoding the underlying input/output rules at the single-cell level, however, remains a challenge due to the vast complexity in the extracellular environments that support such cellular behaviors. The amoebozoa Dictyostelium discoideum uses GPCR-mediated chemotaxis and cell-cell contact signals mediated by adhesion proteins with immunoglobulin-like folds to form a collectively migrating slug. Coordinated migration and repositioning of the cells in this relatively simple morphogenetic system are driven strictly by regulation of actin cytoskeleton by these signaling factors. Its unique position in the eukaryotic tree of life outside metazoa points to basic logics of tissue self-organization that are common across taxa. Here, we describe a method to reconstitute intercellular contact signals and the resulting cell polarization using purified adhesion proteins. In addition, a protocol using a microfluidic chamber is laid out where one can study how the cell-cell contact signal and chemoattractant signals, when simultaneously presented, are interpreted. Quantitative image analysis for obtaining cell morphology features is also provided. A similar approach should be applicable to study other collectively migrating cells.


Assuntos
Comunicação Celular , Movimento Celular , Quimiotaxia , Dictyostelium , Dictyostelium/fisiologia , Dictyostelium/citologia , Adesão Celular , Transdução de Sinais , Polaridade Celular
8.
Signal Transduct Target Ther ; 9(1): 196, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39107318

RESUMO

Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.


Assuntos
Comunicação Celular , Transdução de Sinais , Humanos , Comunicação Celular/genética , Diferenciação Celular , Animais
9.
Nat Commun ; 15(1): 6534, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095390

RESUMO

Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.


Assuntos
Doença de Huntington , Organoides , Fenótipo , Telencéfalo , Doença de Huntington/patologia , Doença de Huntington/genética , Doença de Huntington/metabolismo , Organoides/patologia , Organoides/metabolismo , Animais , Telencéfalo/patologia , Telencéfalo/citologia , Telencéfalo/metabolismo , Humanos , Camundongos , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Análise de Célula Única , Comunicação Celular , Mosaicismo , Neurônios/metabolismo , Neurônios/patologia
10.
Nat Commun ; 15(1): 6820, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39122702

RESUMO

Biomaterial wound dressings, such as hydrogels, interact with host cells to regulate tissue repair. This study investigates how crosslinking of gelatin-based hydrogels influences immune and stromal cell behavior and wound healing in female mice. We observe that softer, lightly crosslinked hydrogels promote greater cellular infiltration and result in smaller scars compared to stiffer, heavily crosslinked hydrogels. Using single-cell RNA sequencing, we further show that heavily crosslinked hydrogels increase inflammation and lead to the formation of a distinct macrophage subpopulation exhibiting signs of oxidative activity and cell fusion. Conversely, lightly crosslinked hydrogels are more readily taken up by macrophages and integrated within the tissue. The physical properties differentially affect macrophage and fibroblast interactions, with heavily crosslinked hydrogels promoting pro-fibrotic fibroblast activity that drives macrophage fusion through RANKL signaling. These findings suggest that tuning the physical properties of hydrogels can guide cellular responses and improve healing, offering insights for designing better biomaterials for wound treatment.


Assuntos
Fibroblastos , Hidrogéis , Macrófagos , Cicatrização , Animais , Hidrogéis/química , Cicatrização/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Feminino , Comunicação Celular/efeitos dos fármacos , Materiais Biocompatíveis/química , Ligante RANK/metabolismo , Camundongos Endogâmicos C57BL , Reagentes de Ligações Cruzadas/química , Gelatina/química , Inflamação/metabolismo , Inflamação/patologia
11.
Mol Cancer ; 23(1): 172, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174949

RESUMO

Exosomes mediate cell-to-cell crosstalk involving a variety of biomolecules through an intricate signaling network. In recent years, the pivotal role of exosomes and their non-coding RNAs cargo in the development and progression of several cancer types clearly emerged. In particular, tumor bulk and its microenvironment co-evolve through cellular communications where these nanosized extracellular vesicles are among the most relevant actors. Knowledge about the cellular, and molecular mechanisms involved in these communications will pave the way for novel exosome-based delivery of therapeutic RNAs as well as innovative prognostic/diagnostic tools. Despite the valuable therapeutic potential and clinical relevance of exosomes, their role on sarcoma has been vaguely reported because the rarity and high heterogeneity of this type of cancer. Here, we dissected the scientific literature to unravel the multifaceted role of exosomal non-coding RNAs as mediator of cell-to-cell communications in the sarcoma subtypes.


Assuntos
Comunicação Celular , Exossomos , RNA não Traduzido , Sarcoma , Humanos , Exossomos/metabolismo , Exossomos/genética , Sarcoma/genética , Sarcoma/patologia , Sarcoma/terapia , Sarcoma/metabolismo , RNA não Traduzido/genética , Animais , Microambiente Tumoral/genética , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais , Biomarcadores Tumorais/genética , Pesquisa Translacional Biomédica
12.
J Zhejiang Univ Sci B ; 25(8): 633-655, 2024 Aug 15.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39155778

RESUMO

Extracellular vesicles (EVs) are nano-sized bilayer vesicles that are shed or secreted by virtually every cell type. A variety of biomolecules, including proteins, lipids, coding and non-coding RNAs, and mitochondrial DNA, can be selectively encapsulated into EVs and delivered to nearby and distant recipient cells, leading to alterations in the recipient cells, suggesting that EVs play an important role in intercellular communication. EVs play effective roles in physiology and pathology and could be used as diagnostic and therapeutic tools. At present, although the mechanisms of exosome biogenesis and secretion in donor cells are well understood, the molecular mechanism of EV recognition and uptake by recipient cells is still unclear. This review summarizes the current understanding of the molecular mechanisms of EVs' biological journey in recipient cells, from recognition to uptake and cargo release. Furthermore, we highlight how EVs escape endolysosomal degradation after uptake and thus release cargo, which is crucial for studies applying EVs as drug-targeted delivery vehicles. Knowledge of the cellular processes that govern EV uptake is important to shed light on the functions of EVs as well as on related clinical applications.


Assuntos
Comunicação Celular , Vesículas Extracelulares , Vesículas Extracelulares/metabolismo , Humanos , Exossomos/metabolismo , Animais , Sistemas de Liberação de Medicamentos , Transporte Biológico
13.
Artigo em Chinês | MEDLINE | ID: mdl-39193734

RESUMO

Objective:To explore the gene expression characteristics of endothelial cells and fibroblasts in the microenvironment of SDHD-mutated carotid body tumors(SDHD-CBT), to fine the functional enrichment of each subcluster, and to further explore the network of cell-cell interactions in the microenvironment of SDHD-CBT. Methods:The bioinformatics analysis was used to download and reanalyze the single-nuclear RNA sequencing data of SDHD-CBT, SDHB mutated thoracic and abdominal paraganglioma(SDHB-ATPGL), SDHB-CBT, and normal adrenal medulla(NAM), to clarify the information of cell populations of the samples. We focused on exploring the gene expression profiles of endothelial cells and fibroblasts subclusters, and performed functional enrichment analysis based on Gene Ontology(GO) resources. CellChat was used to compare the cell-cell interactions networks of different clinical samples and predict significant signaling pathways in SDHD-CBT. Results:A total of 7 cell populations were profiled. The main subtypes of endothelial cells in SDHD-CBT are arterial and venous endothelial cells, and the main subtypes of fibroblasts are myofibroblasts and pericytes. Compared to NAM, SDHB-CBT and SDHB-ATPGL, cell communication involving endothelial cells and fibroblasts in SDHD-CBT is more abundant, with significant enrichment in pathways such as FGF, PTN, WNT, PROS, PERIOSTIN, and TGFb. Conclusion:Endothelial cells and fibroblasts in SDHD-CBT are heterogeneous and involved in important cellular interactionprocesses, in which the discovery of FGF,PTN,WNT,PROS,PERIOSTIN and TGFb signals may play an important role in the regulation of microenvironment of SDHD-CBT.


Assuntos
Células Endoteliais , Fibroblastos , Microambiente Tumoral , Humanos , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Tumor do Corpo Carotídeo/metabolismo , Tumor do Corpo Carotídeo/genética , Tumor do Corpo Carotídeo/patologia , Transdução de Sinais , Succinato Desidrogenase/metabolismo , Succinato Desidrogenase/genética , Biologia Computacional/métodos , Paraganglioma/genética , Paraganglioma/patologia , Paraganglioma/metabolismo , Comunicação Celular , Mutação , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/genética
14.
Front Immunol ; 15: 1373464, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39185406

RESUMO

Introduction: In systemic sclerosis (SSc), B-cells are activated and present in the skin and lung of patients where they can interact with fibroblasts. The precise impact and mechanisms of the interaction of B-cells and fibroblasts at the tissular level are poorly studied. Objective: We investigated the impact and mechanisms of B-cell/fibroblast interactions in cocultures between B-cells from patients with SSc and 3-dimensional reconstituted healthy skin model including fibroblasts, keratinocytes and extracellular matrix. Methods: The quantification and description of the B-cell infiltration in 3D cocultures were performed using cells imagery strategy and cytometry. The effect of coculture on the transcriptome of B-cells and fibroblasts was studied with bulk and single-cell RNA sequencing approaches. The mechanisms of this interaction were studied by blocking key cytokines like IL-6 and TNF. Results: We showed a significant infiltration of B-cells in the 3D healthy skin model. The amount but not the depth of infiltration was higher with B-cells from SSc patients and with activated B-cells. B-cell infiltrates were mainly composed of naïve and memory cells, whose frequencies differed depending on B-cells origin and activation state: infiltrated B-cells from patients with SSc showed an activated profile and an overexpression of immunoglobulin genes compared to circulating B-cells before infiltration. Our study has shown for the first time that activated B-cells modified the transcriptomic profile of both healthy and SSc fibroblasts, toward a pro-inflammatory (TNF and IL-17 signaling) and interferon profile, with a key role of the TNF pathway. Conclusion: B-cells and 3D skin cocultures allowed the modelization of B-cells infiltration in tissues observed in SSc, uncovering an influence of the underlying disease and the activation state of B-cells. We showed a pro-inflammatory effect on skin fibroblasts and pro-activation effect on infiltrating B-cells during coculture. This reinforces the role of B-cells in SSc and provide potential targets for future therapeutic approach in this disease.


Assuntos
Linfócitos B , Técnicas de Cocultura , Fibroblastos , Escleroderma Sistêmico , Pele , Humanos , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/metabolismo , Fibroblastos/imunologia , Fibroblastos/metabolismo , Pele/imunologia , Pele/patologia , Pele/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Feminino , Comunicação Celular/imunologia , Ativação Linfocitária/imunologia , Pessoa de Meia-Idade , Masculino , Células Cultivadas , Transcriptoma , Adulto , Queratinócitos/imunologia , Queratinócitos/metabolismo , Citocinas/metabolismo
15.
Proc Natl Acad Sci U S A ; 121(36): e2404790121, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39186653

RESUMO

Eukaryotic cells are characterized by multiple chemically distinct compartments, one of the most notable being the nucleus. Within these compartments, there is a continuous exchange of information, chemicals, and signaling molecules, essential for coordinating and regulating cellular activities. One of the main goals of bottom-up synthetic biology is to enhance the complexity of synthetic cells by establishing functional compartmentalization. There is a need to mimic autonomous signaling between compartments, which in living cells, is often regulated at the genetic level within the nucleus. This advancement is key to unlocking the potential of synthetic cells as cell models and as microdevices in biotechnology. However, a technological bottleneck exists preventing the creation of synthetic cells with a defined nucleus-like compartment capable of genetically programmed intercompartment signaling events. Here, we present an approach for creating synthetic cells with distinct nucleus-like compartments that can encapsulate different biochemical mixtures in discrete compartments. Our system enables in situ protein expression of membrane proteins, enabling autonomous chemical communication between nuclear and cytoplasmic compartments, leading to downstream activation of enzymatic pathways within the cell.


Assuntos
Células Artificiais , Núcleo Celular , Biologia Sintética , Biologia Sintética/métodos , Núcleo Celular/metabolismo , Núcleo Celular/genética , Células Artificiais/metabolismo , Transdução de Sinais , Citoplasma/metabolismo , Comunicação Celular
16.
Adv Neurobiol ; 39: 1-17, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190069

RESUMO

Proper functioning of the central nervous system depends on various tightly regulated phenomena, among which astrocyte-neuron interactions are of critical importance. Various studies across the species have highlighted the diverse yet crucial roles of astrocytes in regulating the nervous system development and functions. In simpler organisms like worms or insects, astrocyte-like cells govern basic functions such as structural support to neurons or regulation of extracellular ions. As the species complexity increases, so does the functional and morphological complexity of astrocytes. For example, in fish and amphibians, these cells are involved in synaptic development and ion homeostasis, while in reptiles and birds, astrocytes regulate synaptic transmission and plasticity and are reported to be involved in complex behaviors. Other species like those belonging to mammals and, in particular, primates have a heterogeneous population of astrocytes, exhibiting region-specific functional properties. In primates, these cells are responsible for proper synaptic transmission, neurotransmitter release and metabolism, and higher cognitive functions like learning, memory, or information processing. This chapter highlights the well-established and somewhat conserved roles of astrocytes and astrocyte-neuron interactions across the evolution of both invertebrates and vertebrates.


Assuntos
Astrócitos , Evolução Biológica , Plasticidade Neuronal , Neurônios , Transmissão Sináptica , Astrócitos/metabolismo , Animais , Neurônios/metabolismo , Transmissão Sináptica/fisiologia , Plasticidade Neuronal/fisiologia , Humanos , Comunicação Celular/fisiologia , Especificidade da Espécie
17.
Adv Neurobiol ; 39: 69-93, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190072

RESUMO

This chapter explores the intricate interactions between neurons and astrocytes within the nervous system with a particular emphasis on studies conducted in human tissue or with human cells. We specifically explore how neuron-astrocyte interactions relate to processes of cellular development, morphology, migration, synapse formation, and metabolism. These findings enrich our understanding of basic neurobiology and how disruptions in these processes are relevant to human diseases.The study of human neuron-astrocyte interactions is made possible because of transformative in vitro advancements that have facilitated the generation and sustained culture of human neural cells. In addition, the rise of techniques like sequencing at single-cell resolution has enabled the exploration of numerous human cell atlases and their comparisons to other animal model systems. Thus, the innovations outlined in this chapter illuminate the convergence and divergence of neuron-astrocyte interactions across species. As technologies progress, continually more sophisticated in vitro systems will increasingly reflect in vivo environments and deepen our command of neuron-glial interactions in human biology.


Assuntos
Astrócitos , Comunicação Celular , Neurônios , Humanos , Astrócitos/metabolismo , Neurônios/metabolismo , Comunicação Celular/fisiologia , Animais , Sinapses/metabolismo , Sinapses/fisiologia , Movimento Celular/fisiologia
18.
Adv Neurobiol ; 39: 165-191, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190075

RESUMO

Engagement of astrocytes within the brain's reward circuitry has been apparent for approximately 30 years, when noncontingent drug administration was observed to lead to cytological markers of reactive astrocytes. Since that time, advanced approaches in rodent behavior and astrocyte monitoring have revealed complex interactions between astrocytes with drug type, animal sex, brain region, and dose and duration of drug administration. A number of studies now collectively reveal that rodent drug self-administration followed by prolonged abstinence results in decreased features of structure and synaptic colocalization of astrocytes. In addition, stimulation of astrocytes in the nucleus accumbens with DREADD receptors or pharmacological compounds opposes drug-seeking behavior. These findings provide a clear path for ongoing investigation into astrocytes as mediators of drug action in the brain and underscore the potential therapeutic utility of astrocytes in the regulation of drug craving and relapse vulnerability.


Assuntos
Astrócitos , Neurônios , Transtornos Relacionados ao Uso de Substâncias , Astrócitos/metabolismo , Animais , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Humanos , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Comportamento de Procura de Droga , Encéfalo/metabolismo , Recompensa , Comunicação Celular/fisiologia
19.
Adv Neurobiol ; 39: 213-231, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190077

RESUMO

Spinal cord injuries cause irreversible loss of sensory and motor functions. In mammals, intrinsic and extrinsic inhibitions of neuronal regeneration obstruct neural repair after spinal cord injury. Although astrocytes have been involved in a growing list of vital homeostatic functions in the nervous system, their roles after injury have fascinated and puzzled scientists for decades. Astrocytes undergo long-lasting morphological and functional changes after injury, referred to as reactive astrogliosis. Although reactive astrogliosis is required to contain spinal cord lesions and restore the blood-spinal cord barrier, reactive astrocytes have detrimental effects that inhibit neuronal repair and remyelination. Intriguingly, elevated regenerative capacity is preserved in some non-mammalian vertebrates, where astrocyte-like glial cells display exclusively pro-regenerative effects after injury. A detailed molecular and phenotypic catalog of the continuum of astrocyte reactivity states is an essential first step toward the development of glial cell manipulations for spinal cord repair.


Assuntos
Astrócitos , Neurônios , Traumatismos da Medula Espinal , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/patologia , Astrócitos/metabolismo , Animais , Humanos , Neurônios/metabolismo , Gliose/metabolismo , Gliose/patologia , Regeneração Nervosa/fisiologia , Medula Espinal/metabolismo , Comunicação Celular/fisiologia
20.
Adv Neurobiol ; 39: 285-318, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190080

RESUMO

Amyotrophic lateral sclerosis (ALS) is a complex disease impacting motor neurons of the brain, brainstem, and spinal cord. Disease etiology is quite heterogeneous with over 40 genes causing the disease and a vast ~90% of patients having no prior family history. Astrocytes are major contributors to ALS, particularly through involvement in accelerating disease progression. Through study of genetic forms of disease including SOD1, TDP43, FUS, C9orf72, VCP, TBK1, and more recently patient-derived cells from sporadic individuals, many biological mechanisms have been identified to cause intrinsic or glial-mediated neurotoxicity to motor neurons. Overall, many of the normally supportive and beneficial roles that astrocytes contribute to neuronal health and survival instead switch to become deleterious and neurotoxic. While the exact pathways may differ based on disease-origin, altered astrocyte-neuron communication is a common feature of ALS. Within this chapter, distinct genetic forms are examined in detail, along with what is known from sporadic patient-derived cells. Overall, this chapter highlights the interplay between astrocytes and neurons in this complex disease and describes the key features underlying: astrocyte-mediated motor neuron toxicity, excitotoxicity, oxidative/nitrosative stress, protein dyshomeostasis, metabolic imbalance, inflammation, trophic factor withdrawal, blood-brain/blood-spinal cord barrier involvement, disease spreading, and the extracellular matrix/cell adhesion/TGF-ß signaling pathways.


Assuntos
Esclerose Lateral Amiotrófica , Astrócitos , Comunicação Celular , Progressão da Doença , Neurônios Motores , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Humanos , Astrócitos/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Comunicação Celular/fisiologia , Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA