Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 44(29)2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38830764

RESUMO

Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-ß (Aß) pathology (PS2APP) or combined Aß and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.


Assuntos
Doença de Alzheimer , Glicoproteínas de Membrana , Microglia , Esclerose Múltipla , Receptores Imunológicos , Animais , Receptores Imunológicos/agonistas , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Glicoproteínas de Membrana/agonistas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Camundongos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Feminino , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Anticorpos/farmacologia , Humanos , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/metabolismo
2.
Nat Neurosci ; 26(3): 430-446, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36732642

RESUMO

Complex diseases are characterized by spatiotemporal cellular and molecular changes that may be difficult to comprehensively capture. However, understanding the spatiotemporal dynamics underlying pathology can shed light on disease mechanisms and progression. Here we introduce STARmap PLUS, a method that combines high-resolution spatial transcriptomics with protein detection in the same tissue section. As proof of principle, we analyze brain tissues of a mouse model of Alzheimer's disease at 8 and 13 months of age. Our approach provides a comprehensive cellular map of disease progression. It reveals a core-shell structure where disease-associated microglia (DAM) closely contact amyloid-ß plaques, whereas disease-associated astrocyte-like (DAA-like) cells and oligodendrocyte precursor cells (OPCs) are enriched in the outer shells surrounding the plaque-DAM complex. Hyperphosphorylated tau emerges mainly in excitatory neurons in the CA1 region and correlates with the local enrichment of oligodendrocyte subtypes. The STARmap PLUS method bridges single-cell gene expression profiles with tissue histopathology at subcellular resolution, providing a tool to pinpoint the molecular and cellular changes underlying pathology.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Doença de Alzheimer/genética , Modelos Animais de Doenças , Peptídeos beta-Amiloides , Astrócitos , Placa Amiloide , Precursor de Proteína beta-Amiloide , Camundongos Transgênicos , Encéfalo
3.
Cell Rep ; 40(8): 111189, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-36001972

RESUMO

Oligodendrocyte dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, so understanding oligodendrocyte activation states would shed light on disease processes. We identify three distinct activation states of oligodendrocytes from single-cell RNA sequencing (RNA-seq) of mouse models of Alzheimer's disease (AD) and multiple sclerosis (MS): DA1 (disease-associated1, associated with immunogenic genes), DA2 (disease-associated2, associated with genes influencing survival), and IFN (associated with interferon response genes). Spatial analysis of disease-associated oligodendrocytes (DAOs) in the cuprizone model reveals that DA1 and DA2 are established outside of the lesion area during demyelination and that DA1 repopulates the lesion during remyelination. Independent meta-analysis of human single-nucleus RNA-seq datasets reveals that the transcriptional responses of MS oligodendrocytes share features with mouse models. In contrast, the oligodendrocyte activation signature observed in human AD is largely distinct from those observed in mice. This catalog of oligodendrocyte activation states (http://research-pub.gene.com/OligoLandscape/) will be important to understand disease progression and develop therapeutic interventions.


Assuntos
Doenças Desmielinizantes , Esclerose Múltipla , Doenças Neurodegenerativas , Animais , Cuprizona/uso terapêutico , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Oligodendroglia
4.
Cell Rep ; 37(13): 110158, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34965428

RESUMO

Non-neuronal responses in neurodegenerative disease have received increasing attention as important contributors to disease pathogenesis and progression. Here we utilize single-cell RNA sequencing to broadly profile 13 cell types in three different mouse models of Alzheimer disease (AD), capturing the effects of tau-only, amyloid-only, or combined tau-amyloid pathology. We highlight microglia, oligodendrocyte, astrocyte, and T cell responses and compare them across these models. Notably, we identify two distinct transcriptional states for oligodendrocytes emerging differentially across disease models, and we determine their spatial distribution. Furthermore, we explore the impact of Trem2 deletion in the context of combined pathology. Trem2 knockout mice exhibit severely blunted microglial responses to combined tau and amyloid pathology, but responses from non-microglial cell types (oligodendrocytes, astrocytes, and T cells) are relatively unchanged. These results delineate core transcriptional states that are engaged in response to AD pathology, and how they are influenced by a key AD risk gene, Trem2.


Assuntos
Doença de Alzheimer/patologia , Amiloide/química , Astrócitos/patologia , Glicoproteínas de Membrana/fisiologia , Oligodendroglia/patologia , Receptores Imunológicos/fisiologia , Linfócitos T/imunologia , Proteínas tau/metabolismo , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodendroglia/imunologia , Oligodendroglia/metabolismo
5.
Neuron ; 109(8): 1283-1301.e6, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33675684

RESUMO

Loss-of-function TREM2 mutations strongly increase Alzheimer's disease (AD) risk. Trem2 deletion has revealed protective Trem2 functions in preclinical models of ß-amyloidosis, a prominent feature of pre-diagnosis AD stages. How TREM2 influences later AD stages characterized by tau-mediated neurodegeneration is unclear. To understand Trem2 function in the context of both ß-amyloid and tau pathologies, we examined Trem2 deficiency in the pR5-183 mouse model expressing mutant tau alone or in TauPS2APP mice, in which ß-amyloid pathology exacerbates tau pathology and neurodegeneration. Single-cell RNA sequencing in these models revealed robust disease-associated microglia (DAM) activation in TauPS2APP mice that was amyloid-dependent and Trem2-dependent. In the presence of ß-amyloid pathology, Trem2 deletion further exacerbated tau accumulation and spreading and promoted brain atrophy. Without ß-amyloid pathology, Trem2 deletion did not affect these processes. Therefore, TREM2 may slow AD progression and reduce tau-driven neurodegeneration by restricting the degree to which ß-amyloid facilitates the spreading of pathogenic tau.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Atrofia/genética , Atrofia/metabolismo , Atrofia/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Receptores Imunológicos/genética , Proteínas tau/genética
6.
J Exp Med ; 217(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32357367

RESUMO

Myeloid cells play critical and diverse roles in mammalian physiology, including tissue development and repair, innate defense against pathogens, and generation of adaptive immunity. As cells that show prolonged recruitment to sites of injury or pathology, myeloid cells represent therapeutic targets for a broad range of diseases. However, few approaches have been developed for gene editing of these cell types, likely owing to their sensitivity to foreign genetic material or virus-based manipulation. Here we describe optimized strategies for gene disruption in primary myeloid cells of human and murine origin. Using nucleofection-based delivery of Cas9-ribonuclear proteins (RNPs), we achieved near population-level genetic knockout of single and multiple targets in a range of cell types without selection or enrichment. Importantly, we show that cellular fitness and response to immunological stimuli is not significantly impacted by the gene editing process. This provides a significant advance in the study of myeloid cell biology, thus enabling pathway discovery and drug target validation across species in the field of innate immunity.


Assuntos
Sistemas CRISPR-Cas/genética , Técnicas de Inativação de Genes , Técnicas de Transferência de Genes , Células Mieloides/metabolismo , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Deleção de Genes , Edição de Genes , Engenharia Genética , Genoma , Humanos , Macrófagos/metabolismo , Camundongos , Monócitos/metabolismo , Fagocitose , Fenótipo , RNA Guia de Cinetoplastídeos/genética , Ribonucleoproteínas/metabolismo , Vírus/metabolismo
7.
Annu Rev Genet ; 53: 263-288, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31518519

RESUMO

Advances in human genetics have implicated a growing number of genes in neurodegenerative diseases, providing insight into pathological processes. For Alzheimer disease in particular, genome-wide association studies and gene expression studies have emphasized the pathogenic contributions from microglial cells and motivated studies of microglial function/dysfunction. Here, we summarize recent genetic evidence for microglial involvement in neurodegenerative disease with a focus on Alzheimer disease, for which the evidence is most compelling. To provide context for these genetic discoveries, we discuss how microglia influence brain development and homeostasis, how microglial characteristics change in disease, and which microglial activities likely influence the course of neurodegeneration. In all, we aim to synthesize varied aspects of microglial biology and highlight microglia as possible targets for therapeutic interventions in neurodegenerative disease.


Assuntos
Encéfalo/crescimento & desenvolvimento , Microglia/patologia , Microglia/fisiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Envelhecimento/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiologia , Sistema Nervoso Central/metabolismo , Via Clássica do Complemento/fisiologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Homeostase , Humanos , Macrófagos/fisiologia , Placa Amiloide/fisiopatologia , Fator de Crescimento Transformador beta/metabolismo
8.
Cell Rep ; 27(10): 2895-2908.e4, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31167136

RESUMO

Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.


Assuntos
Encéfalo/diagnóstico por imagem , AMP Cíclico/metabolismo , Microglia/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microtúbulos/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Pseudópodes/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
9.
Curr Protoc Immunol ; 125(1): e70, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30414379

RESUMO

Microglia represent 5-10% of cells in the central nervous system and contribute to the development, homeostasis, injury, and repair of neural tissues. As the tissue-resident macrophages of the central nervous system, microglia execute core innate immune functions such as detection of pathogens/damage, cytokine secretion, and phagocytosis. However, additional properties that are specific to microglia and their neural environment are beginning to be appreciated. This article describes approaches for purification of microglia by fluorescence-activated cell sorting using microglia-specific surface markers and for enrichment of microglia by magnetic sorting and immunopanning. Detailed information about culturing primary microglia at various developmental stages is also provided. Throughout, we focus on special considerations for handling microglia and compare the relative strengths or disadvantages of different protocols. © 2018 by John Wiley & Sons, Inc.


Assuntos
Técnicas de Cultura de Células , Citometria de Fluxo , Microglia , Animais , Camundongos , Ratos
10.
Nat Genet ; 50(12): 1716-1727, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30397336

RESUMO

Phagocytosis is required for a broad range of physiological functions, from pathogen defense to tissue homeostasis, but the mechanisms required for phagocytosis of diverse substrates remain incompletely understood. Here, we developed a rapid magnet-based phenotypic screening strategy, and performed eight genome-wide CRISPR screens in human cells to identify genes regulating phagocytosis of distinct substrates. After validating select hits in focused miniscreens, orthogonal assays and primary human macrophages, we show that (1) the previously uncharacterized gene NHLRC2 is a central player in phagocytosis, regulating RhoA-Rac1 signaling cascades that control actin polymerization and filopodia formation, (2) very-long-chain fatty acids are essential for efficient phagocytosis of certain substrates and (3) the previously uncharacterized Alzheimer's disease-associated gene TM2D3 can preferentially influence uptake of amyloid-ß aggregates. These findings illuminate new regulators and core principles of phagocytosis, and more generally establish an efficient method for unbiased identification of cellular uptake mechanisms across diverse physiological and pathological contexts.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Magnetismo/métodos , Fagocitose/genética , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Regulação da Expressão Gênica , Estudos de Associação Genética/métodos , Genoma Humano , Ensaios de Triagem em Larga Escala/métodos , Humanos , Camundongos , Células RAW 264.7 , Transdução de Sinais/genética , Células U937
11.
J Vis Exp ; (133)2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29578519

RESUMO

Microglia represent 5 - 10% of all central nervous system (CNS) cells and are increasingly drawing attention due to their contributions during development, homeostasis, and disease. Although macrophages have been studied in detail for decades, specialized features of microglia, the tissue-resident macrophages of the CNS, have remained largely mysterious, in part due to limitations in the ability to recapitulate mature microglial properties in culture. Here, we illustrate a straightforward procedure for the rapid isolation of pure microglia from the mature rodent brain. We also describe serum-free culture conditions that support high levels of microglial viability over time. Microglia cultured under these defined-medium conditions exhibit elaborate ramified processes and dynamic surveillance behavior. We illustrate some effects of serum exposure on cultured microglia and discuss how these serum-free cultures compare to both serum-exposed cultures as well as microglia in vivo.


Assuntos
Meios de Cultivo Condicionados/metabolismo , Microglia/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Microglia/citologia , Roedores
12.
Neuron ; 94(4): 759-773.e8, 2017 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-28521131

RESUMO

Microglia, the resident macrophages of the CNS, engage in various CNS-specific functions that are critical for development and health. To better study microglia and the properties that distinguish them from other tissue macrophage populations, we have optimized serum-free culture conditions to permit robust survival of highly ramified adult microglia under defined-medium conditions. We find that astrocyte-derived factors prevent microglial death ex vivo and that this activity results from three primary components, CSF-1/IL-34, TGF-ß2, and cholesterol. Using microglial cultures that have never been exposed to serum, we demonstrate a dramatic and lasting change in phagocytic capacity after serum exposure. Finally, we find that mature microglia rapidly lose signature gene expression after isolation, and that this loss can be reversed by engrafting cells back into an intact CNS environment. These data indicate that the specialized gene expression profile of mature microglia requires continuous instructive signaling from the intact CNS.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Colesterol/farmacologia , Interleucinas/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Microglia/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Animais , Astrócitos/metabolismo , Técnicas de Cultura de Células , Colesterol/metabolismo , Meios de Cultivo Condicionados/metabolismo , Humanos , Interleucinas/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Microglia/citologia , Microglia/imunologia , Microglia/metabolismo , Fagocitose/imunologia , Ratos , Soro , Transcriptoma , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta2/metabolismo
13.
Nature ; 541(7638): 481-487, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28099414

RESUMO

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Assuntos
Astrócitos/classificação , Astrócitos/patologia , Morte Celular , Sistema Nervoso Central/patologia , Microglia/patologia , Neurônios/patologia , Animais , Astrócitos/metabolismo , Axotomia , Técnicas de Cultura de Células , Sobrevivência Celular , Complemento C1q/metabolismo , Progressão da Doença , Humanos , Inflamação/patologia , Interleucina-1alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/patologia , Oligodendroglia/patologia , Fagocitose , Fenótipo , Ratos , Ratos Sprague-Dawley , Sinapses/patologia , Toxinas Biológicas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
Nat Med ; 23(2): 164-173, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28092666

RESUMO

Opioid pain medications have detrimental side effects including analgesic tolerance and opioid-induced hyperalgesia (OIH). Tolerance and OIH counteract opioid analgesia and drive dose escalation. The cell types and receptors on which opioids act to initiate these maladaptive processes remain disputed, which has prevented the development of therapies to maximize and sustain opioid analgesic efficacy. We found that µ opioid receptors (MORs) expressed by primary afferent nociceptors initiate tolerance and OIH development. RNA sequencing and histological analysis revealed that MORs are expressed by nociceptors, but not by spinal microglia. Deletion of MORs specifically in nociceptors eliminated morphine tolerance, OIH and pronociceptive synaptic long-term potentiation without altering antinociception. Furthermore, we found that co-administration of methylnaltrexone bromide, a peripherally restricted MOR antagonist, was sufficient to abrogate morphine tolerance and OIH without diminishing antinociception in perioperative and chronic pain models. Collectively, our data support the idea that opioid agonists can be combined with peripheral MOR antagonists to limit analgesic tolerance and OIH.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos/genética , Hiperalgesia/genética , Microglia/metabolismo , Morfina/farmacologia , Nociceptores/metabolismo , Receptores Opioides mu/genética , Analgesia , Animais , Dor Crônica , Modelos Animais de Doenças , Deleção de Genes , Hiperalgesia/induzido quimicamente , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Nociceptividade/efeitos dos fármacos , Dor Pós-Operatória , Compostos de Amônio Quaternário/farmacologia , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais , Medula Espinal/citologia , Medula Espinal/metabolismo
15.
Proc Natl Acad Sci U S A ; 113(12): E1738-46, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26884166

RESUMO

The specific function of microglia, the tissue resident macrophages of the brain and spinal cord, has been difficult to ascertain because of a lack of tools to distinguish microglia from other immune cells, thereby limiting specific immunostaining, purification, and manipulation. Because of their unique developmental origins and predicted functions, the distinction of microglia from other myeloid cells is critically important for understanding brain development and disease; better tools would greatly facilitate studies of microglia function in the developing, adult, and injured CNS. Here, we identify transmembrane protein 119 (Tmem119), a cell-surface protein of unknown function, as a highly expressed microglia-specific marker in both mouse and human. We developed monoclonal antibodies to its intracellular and extracellular domains that enable the immunostaining of microglia in histological sections in healthy and diseased brains, as well as isolation of pure nonactivated microglia by FACS. Using our antibodies, we provide, to our knowledge, the first RNAseq profiles of highly pure mouse microglia during development and after an immune challenge. We used these to demonstrate that mouse microglia mature by the second postnatal week and to predict novel microglial functions. Together, we anticipate these resources will be valuable for the future study and understanding of microglia in health and disease.


Assuntos
Encéfalo/citologia , Proteínas de Membrana/análise , Microglia/química , Proteínas do Tecido Nervoso/análise , Idoso , Animais , Anticorpos Monoclonais/imunologia , Biomarcadores , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Divisão Celular , Linhagem da Célula , Criança , Endotoxemia/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lipopolissacarídeos/toxicidade , Macrófagos/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Microglia/fisiologia , Pessoa de Meia-Idade , Compressão Nervosa , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Traumatismos do Nervo Óptico/patologia , Especificidade de Órgãos , Coelhos , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Análise de Sequência de RNA , Lobo Temporal/metabolismo , Transcriptoma
16.
Cell ; 156(4): 717-29, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24507937

RESUMO

Acid-sensing ion channels (ASICs) detect extracellular protons produced during inflammation or ischemic injury and belong to the superfamily of degenerin/epithelial sodium channels. Here, we determine the cocrystal structure of chicken ASIC1a with MitTx, a pain-inducing toxin from the Texas coral snake, to define the structure of the open state of ASIC1a. In the MitTx-bound open state and in the previously determined low-pH desensitized state, TM2 is a discontinuous α helix in which the Gly-Ala-Ser selectivity filter adopts an extended, belt-like conformation, swapping the cytoplasmic one-third of TM2 with an adjacent subunit. Gly 443 residues of the selectivity filter provide a ring of three carbonyl oxygen atoms with a radius of ∼3.6 Å, presenting an energetic barrier for hydrated ions. The ASIC1a-MitTx complex illuminates the mechanism of MitTx action, defines the structure of the selectivity filter of voltage-independent, sodium-selective ion channels, and captures the open state of an ASIC.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Proteínas Aviárias/química , Galinhas , Venenos Elapídicos/química , Elapidae , Canais Iônicos Sensíveis a Ácido/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Aviárias/metabolismo , Cristalografia por Raios X , Venenos Elapídicos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Alinhamento de Sequência , Canais de Sódio/química
17.
Toxicon ; 60(3): 254-64, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22538196

RESUMO

Venoms often target vital processes to cause paralysis or death, but many types of venom also elicit notoriously intense pain. While these pain-producing effects can result as a byproduct of generalized tissue trauma, there are now multiple examples of venom-derived toxins that target somatosensory nerve terminals in order to activate nociceptive (pain-sensing) neural pathways. Intriguingly, investigation of the venom components that are responsible for evoking pain has revealed novel roles and/or configurations of well-studied toxin motifs. This review serves to highlight pain-producing toxins that target the capsaicin receptor, TRPV1, or members of the acid-sensing ion channel family, and to discuss the utility of venom-derived multivalent and multimeric complexes.


Assuntos
Dor Aguda/induzido quimicamente , Proteínas do Tecido Nervoso/agonistas , Nociceptores/efeitos dos fármacos , Proteínas/toxicidade , Agonistas de Canais de Sódio , Canais de Cátion TRPV/agonistas , Peçonhas/toxicidade , Canais Iônicos Sensíveis a Ácido , Dor Aguda/etiologia , Dor Aguda/metabolismo , Animais , Mordeduras e Picadas/metabolismo , Mordeduras e Picadas/fisiopatologia , Gânglios Sensitivos/efeitos dos fármacos , Gânglios Sensitivos/metabolismo , Humanos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Nociceptores/metabolismo , Conformação Proteica , Subunidades Proteicas/análise , Subunidades Proteicas/química , Subunidades Proteicas/toxicidade , Proteínas/análise , Proteínas/química , Canais de Sódio/química , Canais de Sódio/metabolismo , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/metabolismo , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Peçonhas/química , Peçonhas/enzimologia
18.
J Am Soc Mass Spectrom ; 23(5): 923-34, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22351294

RESUMO

A 7 kDa toxin isolated from the venom of the Texas coral snake (Micrurus tener tener) was subjected to collision-induced dissociation (CID) and electron-transfer dissociation (ETD) analyses both before and after reduction at low pH. Manual and automated approaches to de novo sequencing are compared in detail. Manual de novo sequencing utilizing the combination of high accuracy CID and ETD data and an acid-related cleavage yielded the N-terminal half of the sequence from the reduced species. The intact polypeptide, containing 3 disulfide bridges produced a series of unusual fragments in ion trap CID experiments: abundant internal amino acid losses were detected, and also one of the disulfide-linkage positions could be determined from fragments formed by the cleavage of two bonds. In addition, internal and c-type fragments were also observed.


Assuntos
Dissulfetos/química , Venenos Elapídicos/química , Análise de Sequência de Proteína/métodos , Espectrometria de Massas em Tandem/métodos , Animais , Elapidae , Proteômica/métodos
19.
Archaea ; 2011: 891531, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22162665

RESUMO

RNase P is a highly conserved ribonucleoprotein enzyme that represents a model complex for understanding macromolecular RNA-protein interactions. Archaeal RNase P consists of one RNA and up to five proteins (Pop5, RPP30, RPP21, RPP29, and RPP38/L7Ae). Four of these proteins function in pairs (Pop5-RPP30 and RPP21-RPP29). We have used nuclear magnetic resonance (NMR) spectroscopy and isothermal titration calorimetry (ITC) to characterize the interaction between Pop5 and RPP30 from the hyperthermophilic archaeon Pyrococcus furiosus (Pfu). NMR backbone resonance assignments of free RPP30 (25 kDa) indicate that the protein is well structured in solution, with a secondary structure matching that observed in a closely related crystal structure. Chemical shift perturbations upon the addition of Pop5 (14 kDa) reveal its binding surface on RPP30. ITC experiments confirm a net 1 : 1 stoichiometry for this tight protein-protein interaction and exhibit complex isotherms, indicative of higher-order binding. Indeed, light scattering and size exclusion chromatography data reveal the complex to exist as a 78 kDa heterotetramer with two copies each of Pop5 and RPP30. These results will inform future efforts to elucidate the functional role of the Pop5-RPP30 complex in RNase P assembly and catalysis.


Assuntos
Multimerização Proteica , Pyrococcus furiosus/enzimologia , Ribonuclease P/metabolismo , Calorimetria , Espectroscopia de Ressonância Magnética , Subunidades Proteicas/metabolismo
20.
Nature ; 479(7373): 410-4, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22094702

RESUMO

Natural products that elicit discomfort or pain represent invaluable tools for probing molecular mechanisms underlying pain sensation. Plant-derived irritants have predominated in this regard, but animal venoms have also evolved to avert predators by targeting neurons and receptors whose activation produces noxious sensations. As such, venoms provide a rich and varied source of small molecule and protein pharmacophores that can be exploited to characterize and manipulate key components of the pain-signalling pathway. With this in mind, here we perform an unbiased in vitro screen to identify snake venoms capable of activating somatosensory neurons. Venom from the Texas coral snake (Micrurus tener tener), whose bite produces intense and unremitting pain, excites a large cohort of sensory neurons. The purified active species (MitTx) consists of a heteromeric complex between Kunitz- and phospholipase-A2-like proteins that together function as a potent, persistent and selective agonist for acid-sensing ion channels (ASICs), showing equal or greater efficacy compared with acidic pH. MitTx is highly selective for the ASIC1 subtype at neutral pH; under more acidic conditions (pH < 6.5), MitTx massively potentiates (>100-fold) proton-evoked activation of ASIC2a channels. These observations raise the possibility that ASIC channels function as coincidence detectors for extracellular protons and other, as yet unidentified, endogenous factors. Purified MitTx elicits robust pain-related behaviour in mice by activation of ASIC1 channels on capsaicin-sensitive nerve fibres. These findings reveal a mechanism whereby snake venoms produce pain, and highlight an unexpected contribution of ASIC1 channels to nociception.


Assuntos
Venenos Elapídicos/química , Venenos Elapídicos/farmacologia , Elapidae , Proteínas do Tecido Nervoso/metabolismo , Dor/induzido quimicamente , Multimerização Proteica , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Sequência de Aminoácidos , Animais , Capsaicina/farmacologia , Células Cultivadas , Membro Posterior/efeitos dos fármacos , Membro Posterior/fisiopatologia , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Oócitos , Dor/metabolismo , Dor/fisiopatologia , Estrutura Quaternária de Proteína , Prótons , Ratos , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Agonistas de Canais de Sódio , Canais de Sódio/deficiência , Canais de Sódio/genética , Canais de Cátion TRPV/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...