Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
1.
Cells ; 13(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39272980

RESUMO

Direct neuronal reprogramming is a promising approach to replace neurons lost due to disease via the conversion of endogenous glia reacting to brain injury into neurons. However, it is essential to demonstrate that the newly generated neurons originate from glial cells and/or show that they are not pre-existing endogenous neurons. Here, we use controls for both requirements while comparing two viral vector systems (Mo-MLVs and AAVs) for the expression of the same neurogenic factor, the phosphorylation-resistant form of Neurogenin2. Our results show that Mo-MLVs targeting proliferating glial cells after traumatic brain injury reliably convert astrocytes into neurons, as assessed by genetic fate mapping of astrocytes. Conversely, expressing the same neurogenic factor in a flexed AAV system results in artefactual labelling of endogenous neurons fatemapped by birthdating in development that are negative for the genetic fate mapping marker induced in astrocytes. These results are further corroborated by chronic live in vivo imaging. Taken together, the phosphorylation-resistant form of Neurogenin2 is more efficient in reprogramming reactive glia into neurons than its wildtype counterpart in vivo using retroviral vectors (Mo-MLVs) targeting proliferating glia. Conversely, AAV-mediated expression generates artefacts and is not sufficient to achieve fate conversion.


Assuntos
Astrócitos , Reprogramação Celular , Córtex Cerebral , Dependovirus , Vetores Genéticos , Neurônios , Animais , Astrócitos/metabolismo , Neurônios/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Camundongos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Dependovirus/genética , Reprogramação Celular/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Camundongos Endogâmicos C57BL , Masculino , Retroviridae/genética
2.
Nat Neurosci ; 27(7): 1260-1273, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38956165

RESUMO

Direct neuronal reprogramming is a promising approach to regenerate neurons from local glial cells. However, mechanisms of epigenome remodeling and co-factors facilitating this process are unclear. In this study, we combined single-cell multiomics with genome-wide profiling of three-dimensional nuclear architecture and DNA methylation in mouse astrocyte-to-neuron reprogramming mediated by Neurogenin2 (Ngn2) and its phosphorylation-resistant form (PmutNgn2), respectively. We show that Ngn2 drives multilayered chromatin remodeling at dynamic enhancer-gene interaction sites. PmutNgn2 leads to higher reprogramming efficiency and enhances epigenetic remodeling associated with neuronal maturation. However, the differences in binding sites or downstream gene activation cannot fully explain this effect. Instead, we identified Yy1, a transcriptional co-factor recruited by direct interaction with Ngn2 to its target sites. Upon deletion of Yy1, activation of neuronal enhancers, genes and ultimately reprogramming are impaired without affecting Ngn2 binding. Thus, our work highlights the key role of interactors of proneural factors in direct neuronal reprogramming.


Assuntos
Astrócitos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Reprogramação Celular , Proteínas do Tecido Nervoso , Neurônios , Fator de Transcrição YY1 , Animais , Fator de Transcrição YY1/metabolismo , Fator de Transcrição YY1/genética , Astrócitos/metabolismo , Camundongos , Reprogramação Celular/fisiologia , Neurônios/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Epigenoma , Montagem e Desmontagem da Cromatina , Epigênese Genética , Células Cultivadas
3.
Redox Biol ; 75: 103211, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38908072

RESUMO

Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.


Assuntos
Ferroptose , Humanos , Animais , Ferro/metabolismo , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Peroxidação de Lipídeos , Oxirredução , Suscetibilidade a Doenças
4.
Nat Commun ; 15(1): 2866, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38570482

RESUMO

Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.


Assuntos
Lesões Encefálicas , Ferimentos Perfurantes , Animais , Camundongos , Masculino , Proteína Glial Fibrilar Ácida/metabolismo , Neuroglia/metabolismo , Lesões Encefálicas/metabolismo , Córtex Cerebral/metabolismo , Ferimentos Perfurantes/complicações , Ferimentos Perfurantes/metabolismo
5.
Genes Dev ; 38(3-4): 98-114, 2024 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-38485267

RESUMO

Cell diversification is at the base of increasing multicellular organism complexity in phylogeny achieved during ontogeny. However, there are also functions common to all cells, such as cell division, cell migration, translation, endocytosis, exocytosis, etc. Here we revisit the organelles involved in such common functions, reviewing recent evidence of unexpected differences of proteins at these organelles. For instance, centrosomes or mitochondria differ significantly in their protein composition in different, sometimes closely related, cell types. This has relevance for development and disease. Particularly striking is the high amount and diversity of RNA-binding proteins at these and other organelles, which brings us to review the evidence for RNA at different organelles and suborganelles. We include a discussion about (sub)organelles involved in translation, such as the nucleolus and ribosomes, for which unexpected cell type-specific diversity has also been reported. We propose here that the heterogeneity of these organelles and compartments represents a novel mechanism for regulating cell diversity. One reason is that protein functions can be multiplied by their different contributions in distinct organelles, as also exemplified by proteins with moonlighting function. The specialized organelles still perform pan-cellular functions but in a cell type-specific mode, as discussed here for centrosomes, mitochondria, vesicles, and other organelles. These can serve as regulatory hubs for the storage and transport of specific and functionally important regulators. In this way, they can control cell differentiation, plasticity, and survival. We further include examples highlighting the relevance for disease and propose to examine organelles in many more cell types for their possible differences with functional relevance.


Assuntos
Mitocôndrias , Organelas , Organelas/metabolismo , Mitocôndrias/metabolismo , Divisão Celular , Ribossomos/metabolismo , Diferenciação Celular
6.
Sci Adv ; 10(13): eadn9998, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38536915

RESUMO

Cortical neurogenesis follows a simple lineage: apical radial glia cells (RGCs) generate basal progenitors, and these produce neurons. How this occurs in species with expanded germinal zones and a folded cortex, such as human, remains unclear. We used single-cell RNA sequencing from individual cortical germinal zones in ferret and barcoded lineage tracking to determine the molecular diversity of progenitor cells and their lineages. We identified multiple RGC classes that initiate parallel lineages, converging onto a common class of newborn neuron. Parallel RGC classes and transcriptomic trajectories were repeated across germinal zones and conserved in ferret and human, but not in mouse. Neurons followed parallel differentiation trajectories in the gyrus and sulcus, with different expressions of human cortical malformation genes. Progenitor cell lineage multiplicity is conserved in the folded mammalian cerebral cortex.


Assuntos
Córtex Cerebral , Furões , Animais , Camundongos , Humanos , Linhagem da Célula/fisiologia , Neurônios/fisiologia , Diferenciação Celular , Neurogênese
7.
Neuron ; 112(7): 1117-1132.e9, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38266647

RESUMO

Mitochondria account for essential cellular pathways, from ATP production to nucleotide metabolism, and their deficits lead to neurological disorders and contribute to the onset of age-related diseases. Direct neuronal reprogramming aims at replacing neurons lost in such conditions, but very little is known about the impact of mitochondrial dysfunction on the direct reprogramming of human cells. Here, we explore the effects of mitochondrial dysfunction on the neuronal reprogramming of induced pluripotent stem cell (iPSC)-derived astrocytes carrying mutations in the NDUFS4 gene, important for Complex I and associated with Leigh syndrome. This led to the identification of the unfolded protein response as a major hurdle in the direct neuronal conversion of not only astrocytes and fibroblasts from patients but also control human astrocytes and fibroblasts. Its transient inhibition potently improves reprogramming by influencing the mitochondria-endoplasmic-reticulum-stress-mediated pathways. Taken together, disease modeling using patient cells unraveled novel general hurdles and ways to overcome these in human astrocyte-to-neuron reprogramming.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Mitocondriais , Humanos , Neurônios/fisiologia , Mitocôndrias/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Resposta a Proteínas não Dobradas , Astrócitos/metabolismo , Doenças Mitocondriais/metabolismo , Reprogramação Celular , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo
8.
J Biol Chem ; 300(2): 105648, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38219816

RESUMO

Cellular plasticity is crucial for adapting to ever-changing stimuli. As a result, cells consistently reshape their translatome, and, consequently, their proteome. The control of translational activity has been thoroughly examined at the stage of translation initiation. However, the regulation of ribosome speed in cells is widely unknown. In this study, we utilized a timed ribosome runoff approach, along with proteomics and transmission electron microscopy, to investigate global translation kinetics in cells. We found that ribosome speeds vary among various cell types, such as astrocytes, induced pluripotent human stem cells, human neural stem cells, and human and rat neurons. Of all cell types studied, mature cortical neurons exhibit the highest rate of translation. This finding is particularly remarkable because mature cortical neurons express the eukaryotic elongation factor 2 (eEF2) at lower levels than other cell types. Neurons solve this conundrum by inactivating a fraction of their ribosomes. As a result, the increase in eEF2 levels leads to a reduction of inactive ribosomes and an enhancement of active ones. Processes that alter the demand for active ribosomes, like neuronal excitation, cause increased inactivation of redundant ribosomes in an eEF2-dependent manner. Our data suggest a novel regulatory mechanism in which neurons dynamically inactivate ribosomes to facilitate translational remodeling. These findings have important implications for developmental brain disorders characterized by, among other things, aberrant translation.


Assuntos
Biossíntese de Proteínas , Ribossomos , Animais , Humanos , Ratos , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional , Ribossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
9.
Nat Med ; 29(12): 3149-3161, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38066208

RESUMO

The glial environment influences neurological disease progression, yet much of our knowledge still relies on preclinical animal studies, especially regarding astrocyte heterogeneity. In murine models of traumatic brain injury, beneficial functions of proliferating reactive astrocytes on disease outcome have been unraveled, but little is known regarding if and when they are present in human brain pathology. Here we examined a broad spectrum of pathologies with and without intracerebral hemorrhage and found a striking correlation between lesions involving blood-brain barrier rupture and astrocyte proliferation that was further corroborated in an assay probing for neural stem cell potential. Most importantly, proteomic analysis unraveled a crucial signaling pathway regulating this astrocyte plasticity with GALECTIN3 as a novel marker for proliferating astrocytes and the GALECTIN3-binding protein LGALS3BP as a functional hub mediating astrocyte proliferation and neurosphere formation. Taken together, this work identifies a therapeutically relevant astrocyte response and their molecular regulators in different pathologies affecting the human cerebral cortex.


Assuntos
Astrócitos , Células-Tronco Neurais , Humanos , Camundongos , Animais , Astrócitos/patologia , Proteômica , Encéfalo , Sistema Nervoso Central
10.
Postepy Dermatol Alergol ; 40(5): 661-669, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38028417

RESUMO

Introduction: The key to the correct diagnosis of shrimp allergy is a qualification to the most efficient diagnostic method and later interpretation of the result. To achieve this, it is necessary to apply a diagnostic strategy relevant to each patient's clinical situation and approach every case individually. Aim: In this study the allergen profile of shrimp-sensitized patients was analysed using ALEX2 Allergy Explorer. Material and methods: This study includes 50 adult patients with positive prick-by-prick tests with tiger shrimp bought from the local eco-market and an elevated concentration of IgE specific to the shrimp allergen extract (ImmunoCap). A total of 35 patients with negative skin prick tests with shrimp and not detectable sIgE shrimp in ImmunoCap were included in the control group. All patients had ALEX2 Allergy Explorer microarray test. Results: In the shrimp-sensitized group, 22 patients were sensitized to at least one allergen component of Penaeus monodon, 20 patients were sensitized to crab, and 20 were sensitized to lobster. Only 15 (30%) patients were sensitized to the Northern prawn (Pandalus borealis) allergen extract in ALEX2 and only 12 (24%) to Shrimp mix (Litopenaeus setiferus, Farfantepenaeus aztecus, Farfantepenaeus dourarum). Conclusions: Sensitization to shrimp tropomyosin in the research group was present only in 34% of cases. There may be other shrimp allergen components, not available in ALEX2, which are responsible for shrimp sensitization.

11.
Neuron ; 111(8): 1241-1263.e16, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-36796357

RESUMO

Cortical projection neurons polarize and form an axon while migrating radially. Even though these dynamic processes are closely interwoven, they are regulated separately-the neurons terminate their migration when reaching their destination, the cortical plate, but continue to grow their axons. Here, we show that in rodents, the centrosome distinguishes these processes. Newly developed molecular tools modulating centrosomal microtubule nucleation combined with in vivo imaging uncovered that dysregulation of centrosomal microtubule nucleation abrogated radial migration without affecting axon formation. Tightly regulated centrosomal microtubule nucleation was required for periodic formation of the cytoplasmic dilation at the leading process, which is essential for radial migration. The microtubule nucleating factor γ-tubulin decreased at neuronal centrosomes during the migratory phase. As distinct microtubule networks drive neuronal polarization and radial migration, this provides insight into how neuronal migratory defects occur without largely affecting axonal tracts in human developmental cortical dysgeneses, caused by mutations in γ-tubulin.


Assuntos
Neurônios , Tubulina (Proteína) , Humanos , Tubulina (Proteína)/metabolismo , Neurônios/fisiologia , Axônios/metabolismo , Microtúbulos/metabolismo , Centrossomo , Encéfalo/metabolismo
12.
Postepy Dermatol Alergol ; 39(5): 913-922, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36457677

RESUMO

Introduction: SARS-CoV-2 is responsible for causing the COVID-19 disease, which affected 174 million people worldwide. After vaccines were launched, the focus was on their effectiveness and the degree of their safety. Aim: The authors try to find factors that may affect the response to vaccination. Material and methods: The study was conducted in 47 adults (39 women and 8 men; age: 47.3 ±11.2). Participants were vaccinated with two doses of the Comirnaty mRNA vaccine. Each patient had a medical history taken and the concentration of specific sIgG antibodies against S1 protein and SARS-CoV-2 N protein, as well as of selected cytokines (IL-8, TGF-ß, IFN-γ) was determined before and 3 weeks after the first and second dose of the vaccine. Results: There were 18 convalescents among the respondents. A statistically significant increase in the concentration of specific sIgG S1 in subsequent determinations was observed. Higher levels of sIgG S1 were found after the first dose of the vaccine in COVID-19 convalescents. There was no statistically significant influence of age, body mass index and sex on the increase in the concentration of antibodies and the concentration of the determined cytokines. It was shown that the higher the initial TGF-ß concentration, the greater the increase in sIgG S1 after administration of the vaccine. Conclusions: Vaccination did not increase the levels of IL-8, IFN-ß and TGF-γ. A higher concentration of serum TGF-ß before vaccination correlated with the higher concentration of sIgG S1 antibodies after the first dose of the vaccine.

13.
Mol Syst Biol ; 18(9): e11129, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36106915

RESUMO

Despite the therapeutic promise of direct reprogramming, basic principles concerning fate erasure and the mechanisms to resolve cell identity conflicts remain unclear. To tackle these fundamental questions, we established a single-cell protocol for the simultaneous analysis of multiple cell fate conversion events based on combinatorial and traceable reprogramming factor expression: Collide-seq. Collide-seq revealed the lack of a common mechanism through which fibroblast-specific gene expression loss is initiated. Moreover, we found that the transcriptome of converting cells abruptly changes when a critical level of each reprogramming factor is attained, with higher or lower levels not contributing to major changes. By simultaneously inducing multiple competing reprogramming factors, we also found a deterministic system, in which titration of fates against each other yields dominant or colliding fates. By investigating one collision in detail, we show that reprogramming factors can disturb cell identity programs independent of their ability to bind their target genes. Taken together, Collide-seq has shed light on several fundamental principles of fate conversion that may aid in improving current reprogramming paradigms.


Assuntos
Reprogramação Celular , Fibroblastos , Diferenciação Celular/genética , Reprogramação Celular/genética , Fibroblastos/metabolismo , Transcriptoma/genética
14.
Sci Adv ; 8(23): eabg9445, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35687687

RESUMO

Cell transplantation is a promising approach for the reconstruction of neuronal circuits after brain damage. Transplanted neurons integrate with remarkable specificity into circuitries of the mouse cerebral cortex affected by neuronal ablation. However, it remains unclear how neurons perform in a local environment undergoing reactive gliosis, inflammation, macrophage infiltration, and scar formation, as in traumatic brain injury (TBI). To elucidate this, we transplanted cells from the embryonic mouse cerebral cortex into TBI-injured, inflamed-only, or intact cortex of adult mice. Brain-wide quantitative monosynaptic rabies virus (RABV) tracing unraveled graft inputs from correct regions across the brain in all conditions, with pronounced quantitative differences: scarce in intact and inflamed brain versus exuberant after TBI. In the latter, the initial overshoot is followed by pruning, with only a few input neurons persisting at 3 months. Proteomic profiling identifies candidate molecules for regulation of the synaptic yield, a pivotal parameter to tailor for functional restoration of neuronal circuits.

15.
Sci Adv ; 8(23): eabg9287, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35687689

RESUMO

Transplantation is a clinically relevant approach for brain repair, but much remains to be understood about influences of the disease environment on transplant connectivity. To explore the effect of amyloid pathology in Alzheimer's disease (AD) and aging, we examined graft connectivity using monosynaptic rabies virus tracing in APP/PS1 mice and in 16- to 18-month-old wild-type (WT) mice. Transplanted neurons differentiated within 4 weeks and integrated well into the host visual cortex, receiving input from the appropriate brain regions for this area. Unexpectedly, we found a prominent several-fold increase in local inputs, in both amyloid-loaded and aged environments. State-of-the-art deep proteome analysis using mass spectrometry highlights complement system activation as a common denominator of environments promoting excessive local input connectivity. These data therefore reveal the key role of the host pathology in shaping the input connectome, calling for caution in extrapolating results from one pathological condition to another.

16.
Science ; 376(6599): eabf9088, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35709258

RESUMO

The centrosome provides an intracellular anchor for the cytoskeleton, regulating cell division, cell migration, and cilia formation. We used spatial proteomics to elucidate protein interaction networks at the centrosome of human induced pluripotent stem cell-derived neural stem cells (NSCs) and neurons. Centrosome-associated proteins were largely cell type-specific, with protein hubs involved in RNA dynamics. Analysis of neurodevelopmental disease cohorts identified a significant overrepresentation of NSC centrosome proteins with variants in patients with periventricular heterotopia (PH). Expressing the PH-associated mutant pre-mRNA-processing factor 6 (PRPF6) reproduced the periventricular misplacement in the developing mouse brain, highlighting missplicing of transcripts of a microtubule-associated kinase with centrosomal location as essential for the phenotype. Collectively, cell type-specific centrosome interactomes explain how genetic variants in ubiquitous proteins may convey brain-specific phenotypes.


Assuntos
Centrossomo , Células-Tronco Neurais , Neurogênese , Neurônios , Heterotopia Nodular Periventricular , Mapas de Interação de Proteínas , Processamento Alternativo , Animais , Encéfalo/anormalidades , Centrossomo/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Camundongos , Microtúbulos/metabolismo , Neurônios/metabolismo , Heterotopia Nodular Periventricular/metabolismo , Proteoma/metabolismo , Fatores de Processamento de RNA/metabolismo , Fatores de Transcrição/metabolismo
17.
Trends Cell Biol ; 32(8): 707-719, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35750615

RESUMO

Stem cells are at the source of creating cellular diversity. Multiple mechanisms, including basic cell biological processes, regulate their fate. The centrosome is at the core of many stem cell functions and recent work highlights the association of distinct proteins at the centrosome in stem cell differentiation. As showcased by a novel centrosome protein regulating neural stem cell differentiation, it is timely to review the heterogeneity of the centrosome at protein and RNA levels and how this impacts their function in stem and progenitor cells. Together with evidence for heterogeneity of other organelles so far considered as similar between cells, we call for exploring the cell type-specific composition of organelles as a way to expand protein function in development with relevance to regenerative medicine.


Assuntos
Centrossomo , Células-Tronco Neurais , Diferenciação Celular/fisiologia , Centrossomo/metabolismo , Humanos , Sistema Nervoso , Organelas
18.
Adv Sci (Weinh) ; 9(16): e2104979, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35398994

RESUMO

Astrocytes have crucial functions in the central nervous system (CNS) and are major players in many CNS diseases. Research on astrocyte-centered diseases requires efficient and well-characterized gene transfer vectors. Vectors derived from the Adeno-associated virus serotype 9 (AAV9) target astrocytes in the brains of rodents and nonhuman primates. A recombinant (r) synthetic peptide-displaying AAV9 variant, rAAV9P1, that efficiently and selectively transduces cultured human astrocytes, has been described previously. Here, it is shown that rAAV9P1 retains astrocyte-targeting properties upon intravenous injection in mice. Detailed analysis of putative receptors on human astrocytes shows that rAAV9P1 utilizes integrin subunits αv, ß8, and either ß3 or ß5 as well as the AAV receptor AAVR. This receptor pattern is distinct from that of vectors derived from wildtype AAV2 or AAV9. Furthermore, a CRISPR/Cas9 genome-wide knockout screening revealed the involvement of several astrocyte-associated intracellular signaling pathways in the transduction of human astrocytes by rAAV9P1. This study delineates the unique receptor and intracellular pathway signatures utilized by rAAV9P1 for targeting human astrocytes. These results enhance the understanding of the transduction biology of synthetic rAAV vectors for astrocytes and can promote the development of advanced astrocyte-selective gene delivery vehicles for research and clinical applications.


Assuntos
Astrócitos , Vetores Genéticos , Animais , Astrócitos/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Camundongos , Transdução Genética
19.
EMBO Mol Med ; 14(5): e14797, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35373464

RESUMO

Direct reprogramming based on genetic factors resembles a promising strategy to replace lost cells in degenerative diseases such as Parkinson's disease. For this, we developed a knock-in mouse line carrying a dual dCas9 transactivator system (dCAM) allowing the conditional in vivo activation of endogenous genes. To enable a translational application, we additionally established an AAV-based strategy carrying intein-split-dCas9 in combination with activators (AAV-dCAS). Both approaches were successful in reprogramming striatal astrocytes into induced GABAergic neurons confirmed by single-cell transcriptome analysis of reprogrammed neurons in vivo. These GABAergic neurons functionally integrate into striatal circuits, alleviating voluntary motor behavior aspects in a 6-OHDA Parkinson's disease model. Our results suggest a novel intervention strategy beyond the restoration of dopamine levels. Thus, the AAV-dCAS approach might enable an alternative route for clinical therapies of Parkinson's disease.


Assuntos
Doença de Parkinson , Animais , Astrócitos , Corpo Estriado , Dopamina , Neurônios Dopaminérgicos , Neurônios GABAérgicos , Camundongos , Doença de Parkinson/genética , Doença de Parkinson/terapia
20.
Cell Mol Life Sci ; 79(5): 244, 2022 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-35430697

RESUMO

Adult neurogenesis has been described in two canonical regions of the adult central nervous system (CNS) of rodents, the subgranular zone (SGZ) of the hippocampus and the subependymal zone (SEZ) of the lateral ventricles. The stem cell niche of the SEZ provides a privileged environment composed of a specialized extracellular matrix (ECM) that comprises the glycoproteins tenascin-C (Tnc) and laminin-1 (LN1). In the present study, we investigated the function of these ECM glycoproteins in the adult stem cell niche. Adult neural stem/progenitor cells (aNSPCs) of the SEZ were prepared from wild type (Tnc+/+) and Tnc knockout (Tnc-/-) mice and analyzed using molecular and cell biological approaches. A delayed maturation of aNSPCs in Tnc-/- tissue was reflected by a reduced capacity to form neurospheres in response to epidermal growth factor (EGF). To examine a potential influence of the ECM on cell proliferation, aNSPCs of both genotypes were studied by cell tracking using digital video microscopy. aNSPCs were cultivated on three different substrates, namely, poly-D-lysine (PDL) and PDL replenished with either LN1 or Tnc for up to 6 days in vitro. On each of the three substrates aNSPCs displayed lineage trees that could be investigated with regard to cell cycle length. The latter appeared reduced in Tnc-/- aNSPCs on PDL and LN1 substrates, less so on Tnc that seemed to compensate the absence of the ECM compound to some extent. Close inspection of the lineage trees revealed a subpopulation of late dividing aNSPCslate that engaged into cycling after a notable delay. aNSPCslate exhibited a clearly different morphology, with a larger cell body and conspicuous processes. aNSPCslate reiterated the reduction in cell cycle length on all substrates tested, which was not rescued on Tnc substrates. When the migratory activity of aNSPC-derived progeny was determined, Tnc-/- neuroblasts displayed significantly longer migration tracks. This was traced to an increased rate of migration episodes compared to the wild-type cells that rested for longer time periods. We conclude that Tnc intervenes in the proliferation of aNSPCs and modulates the motility of neuroblasts in the niche of the SEZ.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Células-Tronco Adultas/metabolismo , Animais , Divisão Celular , Matriz Extracelular/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Tenascina/genética , Tenascina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA