Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-38558977

RESUMO

Spared regions of the damaged central nervous system undergo dynamic remodeling and exhibit a remarkable potential for therapeutic exploitation. Here, lesion-remote astrocytes (LRAs), which interact with viable neurons, glia and neural circuitry, undergo reactive transformations whose molecular and functional properties are poorly understood. Using multiple transcriptional profiling methods, we interrogated LRAs from spared regions of mouse spinal cord following traumatic spinal cord injury (SCI). We show that LRAs acquire a spectrum of molecularly distinct, neuroanatomically restricted reactivity states that evolve after SCI. We identify transcriptionally unique reactive LRAs in degenerating white matter that direct the specification and function of local microglia that clear lipid-rich myelin debris to promote tissue repair. Fueling this LRA functional adaptation is Ccn1 , which encodes for a secreted matricellular protein. Loss of astrocyte CCN1 leads to excessive, aberrant activation of local microglia with (i) abnormal molecular specification, (ii) dysfunctional myelin debris processing, and (iii) impaired lipid metabolism, culminating in blunted debris clearance and attenuated neurological recovery from SCI. Ccn1 -expressing white matter astrocytes are specifically induced by local myelin damage and generated in diverse demyelinating disorders in mouse and human, pointing to their fundamental, evolutionarily conserved role in white matter repair. Our findings show that LRAs assume regionally divergent reactivity states with functional adaptations that are induced by local context-specific triggers and influence disorder outcome. Astrocytes tile the central nervous system (CNS) where they serve vital roles that uphold healthy nervous system function, including regulation of synapse development, buffering of neurotransmitters and ions, and provision of metabolic substrates 1 . In response to diverse CNS insults, astrocytes exhibit disorder-context specific transformations that are collectively referred to as reactivity 2-5 . The characteristics of regionally and molecularly distinct reactivity states are incompletely understood. The mechanisms through which distinct reactivity states arise, how they evolve or resolve over time, and their consequences for local cell function and CNS disorder progression remain enigmatic. Immediately adjacent to CNS lesions, border-forming astrocytes (BFAs) undergo transcriptional reprogramming and proliferation to form a neuroprotective barrier that restricts inflammation and supports axon regeneration 6-9 . Beyond the lesion, spared but dynamic regions of the injured CNS exhibit varying degrees of synaptic circuit remodeling and progressive cellular responses to secondary damage that have profound consequences for neural repair and recovery 10,11 . Throughout these cytoarchitecturally intact, but injury-reactive regions, lesion-remote astrocytes (LRAs) intermingle with neurons and glia, undergo little to no proliferation, and exhibit varying degrees of cellular hypertrophy 7,12,13 . The molecular and functional properties of LRAs remain grossly undefined. Therapeutically harnessing spared regions of the injured CNS will require a clearer understanding of the accompanying cellular and molecular landscape. Here, we leveraged integrative transcriptional profiling methodologies to identify multiple spatiotemporally resolved, molecularly distinct states of LRA reactivity within the injured spinal cord. Computational modeling of LRA-mediated heterotypic cell interactions, astrocyte-specific conditional gene deletion, and multiple mouse models of acute and chronic CNS white matter degeneration were used to interrogate a newly identified white matter degeneration-reactive astrocyte subtype. We define how this reactivity state is induced and its role in governing the molecular and functional specification of local microglia that clear myelin debris from the degenerating white matter to promote repair.

2.
Sci Transl Med ; 15(725): eadg7020, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-38055799

RESUMO

Low back pain (LBP) is often associated with the degeneration of human intervertebral discs (IVDs). However, the pain-inducing mechanism in degenerating discs remains to be elucidated. Here, we identified a subtype of locally residing human nucleus pulposus cells (NPCs), generated by certain conditions in degenerating discs, that was associated with the onset of discogenic back pain. Single-cell transcriptomic analysis of human tissues showed a strong correlation between a specific cell subtype and the pain condition associated with the human degenerated disc, suggesting that they are pain-triggering. The application of IVD degeneration-associated exogenous stimuli to healthy NPCs in vitro recreated a pain-associated phenotype. These stimulated NPCs activated functional human iPSC-derived sensory neuron responses in an in vitro organ-chip model. Injection of stimulated NPCs into the healthy rat IVD induced local inflammatory responses and increased cold sensitivity and mechanical hypersensitivity. Our findings reveal a previously uncharacterized pain-inducing mechanism mediated by NPCs in degenerating IVDs. These findings could aid in the development of NPC-targeted therapeutic strategies for the clinically unmet need to attenuate discogenic LBP.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Dor Lombar , Núcleo Pulposo , Humanos , Ratos , Animais , Degeneração do Disco Intervertebral/complicações , Degeneração do Disco Intervertebral/terapia , Dor Lombar/complicações , Crescimento Neuronal
4.
Nature ; 606(7914): 557-564, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614216

RESUMO

Astrocytes respond to injury and disease in the central nervous system with reactive changes that influence the outcome of the disorder1-4. These changes include differentially expressed genes (DEGs) whose contextual diversity and regulation are poorly understood. Here we combined biological and informatic analyses, including RNA sequencing, protein detection, assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and conditional gene deletion, to predict transcriptional regulators that differentially control more than 12,000 DEGs that are potentially associated with astrocyte reactivity across diverse central nervous system disorders in mice and humans. DEGs associated with astrocyte reactivity exhibited pronounced heterogeneity across disorders. Transcriptional regulators also exhibited disorder-specific differences, but a core group of 61 transcriptional regulators was identified as common across multiple disorders in both species. We show experimentally that DEG diversity is determined by combinatorial, context-specific interactions between transcriptional regulators. Notably, the same reactivity transcriptional regulators can regulate markedly different DEG cohorts in different disorders; changes in the access of transcriptional regulators to DNA-binding motifs differ markedly across disorders; and DEG changes can crucially require multiple reactivity transcriptional regulators. We show that, by modulating reactivity, transcriptional regulators can substantially alter disorder outcome, implicating them as therapeutic targets. We provide searchable resources of disorder-related reactive astrocyte DEGs and their predicted transcriptional regulators. Our findings show that transcriptional changes associated with astrocyte reactivity are highly heterogeneous and are customized from vast numbers of potential DEGs through context-specific combinatorial transcriptional-regulator interactions.


Assuntos
Astrócitos , Doenças do Sistema Nervoso Central , Regulação da Expressão Gênica , Fatores de Transcrição , Transcrição Gênica , Animais , Astrócitos/metabolismo , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/patologia , Cromatina/genética , Cromatina/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Análise de Sequência de RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Nature ; 561(7723): 396-400, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30158698

RESUMO

Transected axons fail to regrow across anatomically complete spinal cord injuries (SCI) in adults. Diverse molecules can partially facilitate or attenuate axon growth during development or after injury1-3, but efficient reversal of this regrowth failure remains elusive4. Here we show that three factors that are essential for axon growth during development but are attenuated or lacking in adults-(i) neuron intrinsic growth capacity2,5-9, (ii) growth-supportive substrate10,11 and (iii) chemoattraction12,13-are all individually required and, in combination, are sufficient to stimulate robust axon regrowth across anatomically complete SCI lesions in adult rodents. We reactivated the growth capacity of mature descending propriospinal neurons with osteopontin, insulin-like growth factor 1 and ciliary-derived neurotrophic factor before SCI14,15; induced growth-supportive substrates with fibroblast growth factor 2 and epidermal growth factor; and chemoattracted propriospinal axons with glial-derived neurotrophic factor16,17 delivered via spatially and temporally controlled release from biomaterial depots18,19, placed sequentially after SCI. We show in both mice and rats that providing these three mechanisms in combination, but not individually, stimulated robust propriospinal axon regrowth through astrocyte scar borders and across lesion cores of non-neural tissue that was over 100-fold greater than controls. Stimulated, supported and chemoattracted propriospinal axons regrew a full spinal segment beyond lesion centres, passed well into spared neural tissue, formed terminal-like contacts exhibiting synaptic markers and conveyed a significant return of electrophysiological conduction capacity across lesions. Thus, overcoming the failure of axon regrowth across anatomically complete SCI lesions after maturity required the combined sequential reinstatement of several developmentally essential mechanisms that facilitate axon growth. These findings identify a mechanism-based biological repair strategy for complete SCI lesions that could be suitable to use with rehabilitation models designed to augment the functional recovery of remodelling circuits.


Assuntos
Axônios/fisiologia , Regeneração Nervosa/fisiologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Animais , Astrócitos/patologia , Cicatriz/patologia , Eletrofisiologia , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hidrogéis , Laminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Proteoglicanas/metabolismo , Ratos , Ratos Endogâmicos Lew , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/reabilitação , Regeneração da Medula Espinal , Células Estromais/patologia
6.
J Clin Invest ; 127(9): 3259-3270, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28737515

RESUMO

Spinal cord injury (SCI) lesions present diverse challenges for repair strategies. Anatomically complete injuries require restoration of neural connectivity across lesions. Anatomically incomplete injuries may benefit from augmentation of spontaneous circuit reorganization. Here, we review SCI cell biology, which varies considerably across three different lesion-related tissue compartments: (a) non-neural lesion core, (b) astrocyte scar border, and (c) surrounding spared but reactive neural tissue. After SCI, axon growth and circuit reorganization are determined by neuron-cell-autonomous mechanisms and by interactions among neurons, glia, and immune and other cells. These interactions are shaped by both the presence and the absence of growth-modulating molecules, which vary markedly in different lesion compartments. The emerging understanding of how SCI cell biology differs across lesion compartments is fundamental to developing rationally targeted repair strategies.


Assuntos
Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia , Animais , Astrócitos/citologia , Astrócitos/patologia , Axônios/metabolismo , Axônios/patologia , Axônios/fisiologia , Proliferação de Células , Inflamação , Camundongos , Regeneração Nervosa/fisiologia , Neuroglia/citologia , Neuroglia/patologia , Neurônios/citologia , Neurônios/patologia , Medula Espinal/citologia , Medula Espinal/patologia , Sinapses
7.
Cell Res ; 27(6): 726-727, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28303889

RESUMO

After injury and in disease of the central nervous system (CNS), local cells called astrocytes respond with diverse molecular changes whose functional consequences are incompletely understood. A combined genomic and experimental analysis shows that classically-activated microglia, which are innate immune cells resident in CNS neural tissue, release molecules that drive astrocytes into a neurotoxic state, raising important questions about potential adaptive and maladaptive functions of such a mechanism.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Animais , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Microglia/citologia , Microglia/metabolismo
8.
Sci Rep ; 7: 41122, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28117356

RESUMO

Ependyma have been proposed as adult neural stem cells that provide the majority of newly proliferated scar-forming astrocytes that protect tissue and function after spinal cord injury (SCI). This proposal was based on small, midline stab SCI. Here, we tested the generality of this proposal by using a genetic knock-in cell fate mapping strategy in different murine SCI models. After large crush injuries across the entire spinal cord, ependyma-derived progeny remained local, did not migrate and contributed few cells of any kind and less than 2%, if any, of the total newly proliferated and molecularly confirmed scar-forming astrocytes. Stab injuries that were near to but did not directly damage ependyma, contained no ependyma-derived cells. Our findings show that ependymal contribution of progeny after SCI is minimal, local and dependent on direct ependymal injury, indicating that ependyma are not a major source of endogenous neural stem cells or neuroprotective astrocytes after SCI.


Assuntos
Astrócitos/fisiologia , Diferenciação Celular , Cicatriz/fisiopatologia , Epêndima/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/fisiologia
9.
Nature ; 532(7598): 195-200, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27027288

RESUMO

Transected axons fail to regrow in the mature central nervous system. Astrocytic scars are widely regarded as causal in this failure. Here, using three genetically targeted loss-of-function manipulations in adult mice, we show that preventing astrocyte scar formation, attenuating scar-forming astrocytes, or ablating chronic astrocytic scars all failed to result in spontaneous regrowth of transected corticospinal, sensory or serotonergic axons through severe spinal cord injury (SCI) lesions. By contrast, sustained local delivery via hydrogel depots of required axon-specific growth factors not present in SCI lesions, plus growth-activating priming injuries, stimulated robust, laminin-dependent sensory axon regrowth past scar-forming astrocytes and inhibitory molecules in SCI lesions. Preventing astrocytic scar formation significantly reduced this stimulated axon regrowth. RNA sequencing revealed that astrocytes and non-astrocyte cells in SCI lesions express multiple axon-growth-supporting molecules. Our findings show that contrary to the prevailing dogma, astrocyte scar formation aids rather than prevents central nervous system axon regeneration.


Assuntos
Astrócitos/patologia , Axônios/fisiologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiologia , Cicatriz/patologia , Modelos Biológicos , Regeneração Nervosa , Animais , Sistema Nervoso Central/citologia , Proteoglicanas de Sulfatos de Condroitina/biossíntese , Cicatriz/prevenção & controle , Feminino , Genômica , Camundongos , Reprodutibilidade dos Testes , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
10.
Exp Neurol ; 275 Pt 3: 305-315, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25828533

RESUMO

Astrocytes sense changes in neural activity and extracellular space composition. In response, they exert homeostatic mechanisms critical for maintaining neural circuit function, such as buffering neurotransmitters, modulating extracellular osmolarity and calibrating neurovascular coupling. In addition to upholding normal brain activities, astrocytes respond to diverse forms of brain injury with heterogeneous and progressive changes of gene expression, morphology, proliferative capacity and function that are collectively referred to as reactive astrogliosis. Traumatic brain injury (TBI) sets in motion complex events in which noxious mechanical forces cause tissue damage and disrupt central nervous system (CNS) homeostasis, which in turn trigger diverse multi-cellular responses that evolve over time and can lead either to neural repair or secondary cellular injury. In response to TBI, astrocytes in different cellular microenvironments tune their reactivity to varying degrees of axonal injury, vascular disruption, ischemia and inflammation. Here we review different forms of TBI-induced astrocyte reactivity and the functional consequences of these responses for TBI pathobiology. Evidence regarding astrocyte contribution to post-traumatic tissue repair and synaptic remodeling is examined, and the potential for targeting specific aspects of astrogliosis to ameliorate TBI sequelae is considered.


Assuntos
Astrócitos/patologia , Astrócitos/fisiologia , Lesões Encefálicas/patologia , Animais , Encéfalo/patologia , Encéfalo/fisiologia , Gliose/patologia , Humanos , Plasticidade Neuronal/fisiologia
11.
ACS Biomater Sci Eng ; 1(8): 705-717, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-27547820

RESUMO

Biomaterial vehicles have the potential to facilitate cell transplantation in the central nervous system (CNS). We have previously shown that highly tunable ionic diblock copolypeptide hydrogels (DCH) can provide sustained release of hydrophilic and hydrophobic molecules in the CNS. Here, we show that recently developed non-ionic and thermoresponsive DCH called DCHT exhibit excellent cytocompatibility. Neural stem cell (NSC) suspensions in DCHT were easily injected as liquids at room temperature. DCHT with a viscosity tuned to prevent cell sedimentation and clumping significantly increased the survival of NSC passed through injection cannulae. At body temperature, DCHT self-assembled into hydrogels with a stiffness tuned to that of CNS tissue. After injection in vivo, DCHT significantly increased by three-fold the survival of NSC grafted into healthy CNS. In injured CNS, NSC injected as suspensions in DCHT distributed well in non-neural lesion cores, integrated with healthy neural cells at lesion perimeters and supported regrowing host nerve fibers. Our findings show that non-ionic DCHT have numerous advantageous properties that make them useful tools for in vivo delivery of cells and molecules in the CNS for experimental investigations and potential therapeutic strategies.

12.
Neuron ; 81(2): 229-48, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24462092

RESUMO

The CNS is prone to heterogeneous insults of diverse etiologies that elicit multifaceted responses. Acute and focal injuries trigger wound repair with tissue replacement. Diffuse and chronic diseases provoke gradually escalating tissue changes. The responses to CNS insults involve complex interactions among cells of numerous lineages and functions, including CNS intrinsic neural cells, CNS intrinsic nonneural cells, and CNS extrinsic cells that enter from the circulation. The contributions of diverse nonneuronal cell types to outcome after acute injury, or to the progression of chronic disease, are of increasing interest as the push toward understanding and ameliorating CNS afflictions accelerates. In some cases, considerable information is available, in others, comparatively little, as examined and reviewed here.


Assuntos
Doenças do Sistema Nervoso Central/complicações , Doenças do Sistema Nervoso Central/patologia , Gliose/etiologia , Neuroglia/patologia , Animais , Humanos , Neuroglia/fisiologia , Neurônios/patologia , Neurônios/fisiologia
13.
J Neuropathol Exp Neurol ; 72(11): 1072-89, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24128681

RESUMO

Kallikreins (KLKs) are a family of 15 secreted serine proteases with emerging roles in neurologic diseases. To illuminate their contributions to the pathophysiology of spinal cord injury (SCI), we evaluated acute through chronic changes in the immunohistochemical appearance of 6 KLKs (KLK1, KLK5, KLK6, KLK7, KLK8, and KLK9) in postmortem human traumatic SCI cases, quantified their RNA expression levels in experimental murine SCI, and assessed the impact of recombinant forms of each enzyme toward murine cortical neurons in vitro. Temporally and spatially distinct changes in KLK expression were observed with partially overlapping patterns between human and murine SCI, including peak elevations (or reductions) during the acute and subacute periods. Kallikrein 9 showed the most marked changes and remained chronically elevated. Importantly, a subset of KLKs (KLK1, KLK5, KLK6, KLK7, and KLK9) were neurotoxic toward primary neurons in vitro. Kallikrein immunoreactivity was also observed in association with swollen axons and retraction bulbs in the human SCI cases examined. Together, these findings demonstrate that elevated levels of a significant subset of KLKs are positioned to contribute to neurodegenerative changes in cases of CNS trauma and disease and, therefore, represent new potential targets for the development of neuroprotective strategies.


Assuntos
Axônios/metabolismo , Calicreínas/metabolismo , Degeneração Neural/metabolismo , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Adolescente , Adulto , Idoso , Animais , Axônios/patologia , Criança , Pré-Escolar , Feminino , Humanos , Calicreínas/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Degeneração Neural/genética , Degeneração Neural/patologia , Transdução de Sinais/fisiologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
14.
Glia ; 61(9): 1456-70, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23832758

RESUMO

Kallikrein 6 (KLK6) is a secreted serine protease preferentially expressed by oligodendroglia in CNS white matter. Elevated levels of KLK6 occur in actively demyelinating multiple sclerosis (MS) lesions and in cases of spinal cord injury (SCI), stroke, and glioblastoma. Taken with recent evidence establishing KLK6 as a CNS-endogenous activator of protease-activated receptors (PARs), we hypothesized that KLK6 activates a subset of PARs to regulate oligodendrocyte physiology and potentially pathophysiology. Here, primary oligodendrocyte cultures derived from wild type or PAR1-deficient mice and the murine oligodendrocyte cell line, Oli-neu, were used to demonstrate that Klk6 (rodent form) mediates loss of oligodendrocyte processes and impedes morphological differentiation of oligodendrocyte progenitor cells (OPCs) in a PAR1-dependent fashion. Comparable gliopathy was also elicited by the canonical PAR1 agonist, thrombin, as well as PAR1-activating peptides (PAR1-APs). Klk6 also exacerbated ATP-mediated oligodendrogliopathy in vitro, pointing to a potential role in augmenting excitotoxicity. In addition, Klk6 suppressed the expression of proteolipid protein (PLP) RNA in cultured oligodendrocytes by a mechanism involving PAR1-mediated Erk1/2 signaling. Microinjection of PAR1 agonists, including Klk6 or PAR1-APs, into the dorsal column white matter of PAR1(+/+) but not PAR1(-/-) mice promoted vacuolating myelopathy and a loss of immunoreactivity for myelin basic protein (MBP) and CC-1(+) oligodendrocytes. These results demonstrate a functional role for Klk6-PAR1 signaling in oligodendroglial pathophysiology and suggest that antagonists of PAR1 or its protease agonists may represent new modalities to moderate demyelination and to promote myelin regeneration in cases of CNS white matter injury or disease.


Assuntos
Calicreínas/metabolismo , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Receptor PAR-1/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Trifosfato de Adenosina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Calicreínas/genética , Calicreínas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Básica da Mielina , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Receptor PAR-1/agonistas , Receptor PAR-1/deficiência , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Trombina/farmacologia
15.
Biol Chem ; 393(5): 355-67, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22505518

RESUMO

Kallikrein-related peptidase 6 (KLK6) is a trypsin-like serine protease upregulated at sites of central nervous system (CNS) injury, including de novo expression by reactive astrocytes, yet its physiological actions are largely undefined. Taken with recent evidence that KLK6 activates G-protein-coupled protease-activated receptors (PARs), we hypothesized that injury-induced elevations in KLK6 contribute to the development of astrogliosis and that this occurs in a PAR-dependent fashion. Using primary murine astrocytes and the Neu7 astrocyte cell line, we show that KLK6 causes astrocytes to transform from an epitheliod to a stellate morphology and to secrete interleukin 6 (IL-6). By contrast, KLK6 reduced expression of glial fibrillary acidic protein (GFAP). The stellation-promoting activities of KLK6 were shown to be dependent on activation of the thrombin receptor, PAR1, as a PAR1-specific inhibitor, SCH79797, blocked KLK6-induced morphological changes. The ability of KLK6 to promote astrocyte stellation was also shown to be linked to activation of protein kinase C (PKC). These studies indicate that KLK6 is positioned to serve as a molecular trigger of select physiological processes involved in the development of astrogliosis and that this is likely to occur at least in part by activation of the G-protein-coupled receptor, PAR1.


Assuntos
Gliose/enzimologia , Gliose/patologia , Calicreínas/metabolismo , Animais , Astrócitos/enzimologia , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/enzimologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Transporte/metabolismo , Proliferação de Células , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/metabolismo , Humanos , Hipertrofia , Interleucina-6/metabolismo , Camundongos , Proteína Quinase C/metabolismo , Receptor PAR-1/metabolismo , Medula Espinal/enzimologia , Medula Espinal/metabolismo , Medula Espinal/patologia
16.
Am J Physiol Gastrointest Liver Physiol ; 296(4): G717-26, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19221017

RESUMO

Inflammatory bowel diseases (IBD) can involve widespread gastrointestinal dysfunction, even in cases in which inflammation is localized to a single site. The underlying pathophysiology of dysfunction in noninflamed regions is unclear. We examined whether colitis is associated with altered electrogenic ion transport in the ileal mucosa and/or changes in the properties of ileal submucosal neurons. Colitis was induced by administration of trinitrobenzene sulfonic acid (TNBS), and the uninflamed ileum from animals was examined 3, 7, and 28 days later. Electrogenic ion transport was assessed in Ussing chambers. Intracellular microelectrode recordings were used to examine the neurophysiology of the submucosal plexus of the ileum in animals with colitis. Noncholinergic secretion was reduced by 33% in the ileum from animals 7 days after the induction of colitis. The epithelial response to vasoactive intestinal peptide (VIP) was unaltered in animals with colitis, but the response to carbachol was enhanced. Slow excitatory synaptic transmission was dramatically reduced in VIP-expressing, noncholinergic secretomotor neurons. This change was detected as early as 3 days following TNBS treatment. No changes to fast synaptic transmission or the number of VIP neurons were observed. In addition, cholinergic secretomotor neurons fired more action potentials during a given stimulus, and intrinsic primary afferent neurons had broader action potentials in animals with colitis. These findings implicate changes to enteric neural circuits as contributing factors in inflammation-induced secretory dysfunction at sites proximal to a localized inflammatory insult.


Assuntos
Colite/fisiopatologia , Íleo/inervação , Íleo/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Animais , Colite/induzido quimicamente , Cobaias , Mucosa Intestinal/inervação , Mucosa Intestinal/metabolismo , Masculino , Ácido Trinitrobenzenossulfônico/toxicidade , Peptídeo Intestinal Vasoativo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA