Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-37398301

RESUMO

CRISPR-based genetic screening directly in mammalian tissues in vivo is challenging due to the need for scalable, cell-type selective delivery and recovery of guide RNA libraries. We developed an in vivo adeno-associated virus-based and Cre recombinase-dependent workflow for cell type-selective CRISPR interference screening in mouse tissues. We demonstrate the power of this approach by identifying neuron-essential genes in the mouse brain using a library targeting over 2000 genes.

2.
Nat Commun ; 13(1): 6581, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36323693

RESUMO

Astrocytes are critical components of the neurovascular unit that support blood-brain barrier (BBB) function. Pathological transformation of astrocytes to reactive states can be protective or harmful to BBB function. Here, using a human induced pluripotent stem cell (iPSC)-derived BBB co-culture model, we show that tumor necrosis factor (TNF) transitions astrocytes to an inflammatory reactive state that causes BBB dysfunction through activation of STAT3 and increased expression of SERPINA3, which encodes alpha 1-antichymotrypsin (α1ACT). To contextualize these findings, we correlated astrocytic STAT3 activation to vascular inflammation in postmortem human tissue. Further, in murine brain organotypic cultures, astrocyte-specific silencing of Serpina3n reduced vascular inflammation after TNF challenge. Last, treatment with recombinant Serpina3n in both ex vivo explant cultures and in vivo was sufficient to induce BBB dysfunction-related molecular changes. Overall, our results define the TNF-STAT3-α1ACT signaling axis as a driver of an inflammatory reactive astrocyte signature that contributes to BBB dysfunction.


Assuntos
Barreira Hematoencefálica , Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Barreira Hematoencefálica/metabolismo , Astrócitos/metabolismo , alfa 1-Antiquimotripsina/metabolismo , Células Cultivadas , Células-Tronco Pluripotentes Induzidas/metabolismo , Inflamação/patologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Transcrição STAT3/metabolismo
3.
Cancer Discov ; 12(5): 1314-1335, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35262173

RESUMO

Brain metastasis is a significant cause of morbidity and mortality in multiple cancer types and represents an unmet clinical need. The mechanisms that mediate metastatic cancer growth in the brain parenchyma are largely unknown. Melanoma, which has the highest rate of brain metastasis among common cancer types, is an ideal model to study how cancer cells adapt to the brain parenchyma. Our unbiased proteomics analysis of melanoma short-term cultures revealed that proteins implicated in neurodegenerative pathologies are differentially expressed in melanoma cells explanted from brain metastases compared with those derived from extracranial metastases. We showed that melanoma cells require amyloid beta (Aß) for growth and survival in the brain parenchyma. Melanoma-secreted Aß activates surrounding astrocytes to a prometastatic, anti-inflammatory phenotype and prevents phagocytosis of melanoma by microglia. Finally, we demonstrate that pharmacologic inhibition of Aß decreases brain metastatic burden. SIGNIFICANCE: Our results reveal a novel mechanistic connection between brain metastasis and Alzheimer's disease, two previously unrelated pathologies; establish Aß as a promising therapeutic target for brain metastasis; and demonstrate suppression of neuroinflammation as a critical feature of metastatic adaptation to the brain parenchyma. This article is highlighted in the In This Issue feature, p. 1171.


Assuntos
Neoplasias Encefálicas , Melanoma , Peptídeos beta-Amiloides/uso terapêutico , Astrócitos/metabolismo , Neoplasias Encefálicas/genética , Humanos , Melanoma/tratamento farmacológico , Metástase Neoplásica , Doenças Neuroinflamatórias
4.
Sci Signal ; 12(569)2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30783009

RESUMO

Astrocytes and microglia play critical roles in brain inflammation. Here, we report that glutathione S-transferases (GSTs), particularly GSTM1, promote proinflammatory signaling in astrocytes and contribute to astrocyte-mediated microglia activation during brain inflammation. In vivo, astrocyte-specific knockdown of GSTM1 in the prefrontal cortex attenuated microglia activation in brain inflammation induced by systemic injection of lipopolysaccharides (LPS). Knocking down GSTM1 in astrocytes also attenuated LPS-induced production of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) by microglia when the two cell types were cocultured. In astrocytes, GSTM1 was required for the activation of nuclear factor κB (NF-κB) and the production of proinflammatory mediators, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and C-C motif chemokine ligand 2 (CCL2), both of which enhance microglia activation. Our study suggests that GSTs play a proinflammatory role in priming astrocytes and enhancing microglia activation in a microglia-astrocyte positive feedback loop during brain inflammation.


Assuntos
Astrócitos/metabolismo , Encefalite/metabolismo , Glutationa Transferase/metabolismo , Microglia/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Citocinas/metabolismo , Encefalite/genética , Encefalite/patologia , Feminino , Glutationa Transferase/genética , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/citologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos
5.
Schizophr Bull ; 45(1): 7-16, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30239909

RESUMO

Extracellular vesicles (EVs) have gained increasing attention as underexplored intercellular communication mechanisms in basic science and as potential diagnostic tools in translational studies, particularly those related to cancers and neurological disorders. This article summarizes accumulated findings in the basic biology of EVs, EV research methodology, and the roles of EVs in brain cell function and dysfunction, as well as emerging EV studies in human brain disorders. Further research on EVs in neurobiology and psychiatry may open the door to a better understanding of intercellular communications in healthy and diseased brains, and the discovery of novel biomarkers and new therapeutic strategies in psychiatric disorders.


Assuntos
Biomarcadores , Encéfalo/fisiologia , Vesículas Extracelulares/fisiologia , Transtornos Mentais/diagnóstico , Humanos
6.
eNeuro ; 4(6)2017.
Artigo em Inglês | MEDLINE | ID: mdl-29379874

RESUMO

Interleukin (IL)-33 is a member of the IL-1 family of cytokines. IL-33 is expressed in nuclei and secreted as alarmin upon cellular damage to deliver a danger signal to the surrounding cells. Previous studies showed that IL-33 is expressed in the brain and that it is involved in neuroinflammatory and neurodegenerative processes in both humans and rodents. Nevertheless, the role of IL-33 in physiological brain function and behavior remains unclear. Here, we have investigated the behaviors of mice lacking IL-33 (Il33-/- mice). IL-33 is constitutively expressed throughout the adult mouse brain, mainly in oligodendrocyte-lineage cells and astrocytes. Notably, Il33-/- mice exhibited reduced anxiety-like behaviors in the elevated plus maze (EPM) and the open field test (OFT), as well as deficits in social novelty recognition, despite their intact sociability, in the three-chamber social interaction test. The immunoreactivity of c-Fos proteins, an indicator of neuronal activity, was altered in several brain regions implicated in anxiety-related behaviors, such as the medial prefrontal cortex (mPFC), amygdala, and piriform cortex (PCX), in Il33-/- mice after the EPM. Altered c-Fos immunoreactivity in Il33-/- mice was not correlated with IL-33 expression in wild-type (WT) mice nor was IL-33 expression affected by the EPM in WT mice. Thus, our study has revealed that Il33-/- mice exhibit multiple behavioral deficits, such as reduced anxiety and impaired social recognition. Our findings also indicate that IL-33 may regulate the development and/or maturation of neuronal circuits, rather than control neuronal activities in adult brains.


Assuntos
Comportamento Animal/fisiologia , Interleucina-33/deficiência , Camundongos Knockout/psicologia , Animais , Ansiedade/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Interleucina-33/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reconhecimento Psicológico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA