Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Neurosci ; 125: 103842, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36924917

RESUMO

Chemical platforms that facilitate both the identification and elucidation of new areas for therapeutic development are necessary but lacking. Activity-based protein profiling (ABPP) leverages active site-directed chemical probes as target discovery tools that resolve activity from expression and immediately marry the targets identified with lead compounds for drug design. However, this approach has traditionally focused on predictable and intrinsic enzyme functionality. Here, we applied our activity-based proteomics discovery platform to map non-encoded and post-translationally acquired enzyme functionalities (e.g. cofactors) in vivo using chemical probes that exploit the nucleophilic hydrazine pharmacophores found in a classic antidepressant drug (e.g. phenelzine, Nardil®). We show the probes are in vivo active and can map proteome-wide tissue-specific target engagement of the drug. In addition to engaging targets (flavoenzymes monoamine oxidase A/B) that are associated with the known therapeutic mechanism as well as several other members of the flavoenzyme family, the probes captured the previously discovered N-terminal glyoxylyl (Glox) group of Secernin-3 (SCRN3) in vivo through a divergent mechanism, indicating this functional feature has biochemical activity in the brain. SCRN3 protein is ubiquitously expressed in the brain, yet gene expression is regulated by inflammatory stimuli. In an inflammatory pain mouse model, behavioral assessment of nociception showed Scrn3 male knockout mice selectively exhibited impaired thermal nociceptive sensitivity. Our study provides a guided workflow to entangle molecular (off)targets and pharmacological mechanisms for therapeutic development.


Assuntos
Nociceptividade , Fenelzina , Animais , Camundongos , Masculino , Fenelzina/farmacologia , Proteoma , Proteínas do Tecido Nervoso
2.
J Neurosci ; 38(45): 9618-9634, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30242049

RESUMO

Although age-at-injury influences chronic recovery from traumatic brain injury (TBI), the differential effects of age on early outcome remain understudied. Using a male murine model of moderate contusion injury, we investigated the underlying mechanism(s) regulating the distinct response between juvenile and adult TBI. We demonstrate similar biomechanical and physical properties of naive juvenile and adult brains. However, following controlled cortical impact (CCI), juvenile mice displayed reduced cortical lesion formation, cell death, and behavioral deficits at 4 and 14 d. Analysis of high-resolution laser Doppler imaging showed a similar loss of cerebral blood flow (CBF) in the ipsilateral cortex at 3 and 24 h post-CCI, whereas juvenile mice showed enhanced subsequent restoration at 2-4 d compared with adults. These findings correlated with reduced blood-brain barrier (BBB) disruption and increased perilesional vessel density. To address whether an age-dependent endothelial cell (EC) response affects vessel stability and tissue outcome, we magnetically isolated CD31+ ECs from sham and injured cortices and evaluated mRNA expression. Interestingly, we found increased transcripts for BBB stability-related genes and reduced expression of BBB-disrupting genes in juveniles compared with adults. These differences were concomitant with significant changes in miRNA-21-5p and miR-148a levels. Accompanying these findings was robust GFAP immunoreactivity, which was not resolved by day 35. Importantly, pharmacological inhibition of EC-specific Tie2 signaling abolished the juvenile protective effects. These findings shed new mechanistic light on the divergent effects that age plays on acute TBI outcome that are both spatial and temporal dependent.SIGNIFICANCE STATEMENT Although a clear "window of susceptibility" exists in the developing brain that could deter typical developmental trajectories if exposed to trauma, a number of preclinical models have demonstrated evidence of early recovery in younger patients. Our findings further demonstrate acute neuroprotection and improved restoration of cerebral blood flow in juvenile mice subjected to cortical contusion injury compared with adults. We also demonstrate a novel role for endothelial cell-specific Tie2 signaling in this age-related response, which is known to promote barrier stability, is heightened in the injured juvenile vasculature, and may be exploited for therapeutic interventions across the age spectrum following traumatic brain injury.


Assuntos
Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/metabolismo , Circulação Cerebrovascular/fisiologia , Receptor TIE-2/metabolismo , Fatores Etários , Animais , Células Cultivadas , Masculino , Camundongos
3.
Circ Res ; 120(6): 960-977, 2017 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-28302742

RESUMO

In the past 2 decades, it has become evident that individuals born with congenital heart disease (CHD) are at risk of developing life-long neurological deficits. Multifactorial risk factors contributing to neurodevelopmental abnormalities associated with CHD have been identified; however, the underlying causes remain largely unknown, and efforts to address this issue have only recently begun. There has been a dramatic shift in focus from newly acquired brain injuries associated with corrective and palliative heart surgery to antenatal and preoperative factors governing altered brain maturation in CHD. In this review, we describe key time windows of development during which the immature brain is vulnerable to injury. Special emphasis is placed on the dynamic nature of cellular events and how CHD may adversely impact the cellular units and networks necessary for proper cognitive and motor function. In addition, we describe current gaps in knowledge and offer perspectives about what can be done to improve our understanding of neurological deficits in CHD. Ultimately, a multidisciplinary approach will be essential to prevent or improve adverse neurodevelopmental outcomes in individuals surviving CHD.


Assuntos
Encéfalo/crescimento & desenvolvimento , Cardiopatias Congênitas/diagnóstico , Transtornos do Neurodesenvolvimento/diagnóstico , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Circulação Cerebrovascular , Cardiopatias Congênitas/complicações , Humanos , Transtornos do Neurodesenvolvimento/epidemiologia , Transtornos do Neurodesenvolvimento/etiologia
4.
J Neurosci ; 33(24): 9932-6, 2013 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23761888
5.
Glia ; 62(3): 452-67, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24357067

RESUMO

Astrocytes respond to insult with a process of cellular activation known as reactive astrogliosis. One of the key signals regulating this phenomenon is the transcription factor nuclear factor-kappa B (NF-κB), which is responsible for modulating inflammation, cell survival, and cell death. In astrocytes, following trauma or disease, the expression of NF-κB-dependent genes is highly activated. We previously demonstrated that inactivation of astroglial NF-κB in vivo (GFAP-IκBα-dn mice) leads to improved functional outcome in experimental autoimmune encephalomyelitis (EAE), and this is accompanied by reduction of pro-inflammatory gene expression in the CNS. Here we extend our studies to show that recovery from EAE in GFAP-IκBα-dn mice is associated with reduction of peripheral immune cell infiltration into the CNS at the chronic phase of EAE. This is not dependent on a less permeable blood-brain barrier, but rather on a reduced immune cell mobilization from the periphery. Furthermore, once inside the CNS, the ability of T cells to produce pro-inflammatory cytokines is diminished during acute disease. In parallel, we found that the number of total and activated microglial cells is reduced, suggesting that functional improvement in GFAP-IκBα-dn mice is dependent upon reduction of the overall inflammatory response within the CNS sustained by both resident and infiltrating cells. This results in preservation of myelin compaction and enhanced remyelination, as shown by electron microscopy analysis of the spinal cord. Collectively our data indicate that astrocytes are key players in driving CNS inflammation and are directly implicated in the pathophysiology of EAE, since blocking their pro-inflammatory capability results in protection from the disease.


Assuntos
Astrócitos/fisiologia , Sistema Nervoso Central/metabolismo , Encefalomielite Autoimune Experimental/fisiopatologia , Regulação da Expressão Gênica/imunologia , Inflamação/etiologia , Inflamação/patologia , Animais , Astrócitos/ultraestrutura , Sistema Nervoso Central/imunologia , Claudina-5/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas I-kappa B/genética , Imunoglobulina G/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Bainha de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidade , Inibidor de NF-kappaB alfa , Fragmentos de Peptídeos/toxicidade , Medula Espinal , Linfócitos T/metabolismo
6.
J Neurosci Res ; 91(6): 828-37, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23553703

RESUMO

Spinal cord injury results in irreversible paralysis, axonal injury, widespread oligodendrocyte death, and white matter damage. Although the mechanisms underlying these phenomena are poorly understood, previous studies from our laboratory indicate that inhibiting activation of the nuclear factor-κB transcription factor in astrocytes reduces white matter damage and improves functional recovery following spinal cord injury. In the current study, we demonstrate that activation of the nuclear factor-κB transcription factor within astrocytes results in a significant increase in oligodendrocyte death following trauma by reducing extracellular zinc levels and inducing glutamate excitotoxicity. By using an ionotropic glutamate receptor antagonist (CNQX), we show that astroglial nuclear factor-κB-mediated oligodendrocyte death is dependent on glutamate signaling despite no change in extracellular glutamate concentrations. Further analysis demonstrated a reduction in levels of extracellular zinc in astrocyte cultures with functional nuclear factor-κB signaling following trauma. Cotreatment of oligodendrocytes with glutamate and zinc showed a significant increase in oligodendrocyte toxicity under low-zinc conditions, suggesting that the presence of zinc at specific concentrations can prevent glutamate excitotoxicity. These studies demonstrate a novel role for zinc in regulating oligodendrocyte excitotoxicity and identify new therapeutic targets to prevent oligodendrocyte cell death in central nervous system trauma and disease.


Assuntos
Morte Celular/fisiologia , Líquido Extracelular/química , Oligodendroglia/metabolismo , Traumatismos da Medula Espinal/metabolismo , Zinco/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Líquido Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Oligodendroglia/patologia , Traumatismos da Medula Espinal/patologia
7.
bioRxiv ; 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36778412

RESUMO

Chemical platforms that facilitate both the identification and elucidation of new areas for therapeutic development are necessary but lacking. Activity-based protein profiling (ABPP) leverages active site-directed chemical probes as target discovery tools that resolve activity from expression and immediately marry the targets identified with lead compounds for drug design. However, this approach has traditionally focused on predictable and intrinsic enzyme functionality. Here, we applied our activity-based proteomics discovery platform to map non-encoded and post-translationally acquired enzyme functionalities (e.g. cofactors) in vivo using chemical probes that exploit the nucleophilic hydrazine pharmacophores found in a classic antidepressant drug (e.g. phenelzine, Nardil ® ). We show the probes are in vivo active and can map proteome-wide tissue-specific target engagement of the drug. In addition to engaging targets (flavoenzymes monoamine oxidase A/B) that are associated with the known therapeutic mechanism as well as several other members of the flavoenzyme family, the probes captured the previously discovered N -terminal glyoxylyl (Glox) group of Secernin-3 (SCRN3) in vivo through a divergent mechanism, indicating this functional feature has biochemical activity in the brain. SCRN3 protein is ubiquitously expressed in the brain, yet gene expression is regulated by inflammatory stimuli. In an inflammatory pain mouse model, behavioral assessment of nociception showed Scrn3 male knockout mice selectively exhibited impaired thermal nociceptive sensitivity. Our study provides a guided workflow to entangle molecular (off)targets and pharmacological mechanisms for therapeutic development.

8.
Glia ; 60(4): 639-50, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22275133

RESUMO

Schwann cells (SCs) are crucial for peripheral nerve development and regeneration; however, the intrinsic regulatory mechanisms governing postinjury responses are poorly understood. Activation and deacetylation of nuclear factor-κB (NF- κB) in SCs have been implicated as prerequisites for peripheral nerve myelination. Using GFAP-IκBα-dn mice in which NF- κB transcriptional activation is inhibited in SCs, we found no discernable differences in the quantity or structure of myelinated axons in adult facial nerves. Following crush injury, axonal regeneration was impaired at 31 days and significantly enhanced at 65 days in transgenic animals. Compact remyelination and Remak bundle organization were significantly compromised at 31 days and restored by 65 days post injury. Together, these data indicate that inhibition of NF-κB activation in SCs transiently delays axonal regeneration and compact remyelination. Manipulating the temporal activation of nuclear factor-κB in Schwann cells may offer new therapeutic avenues for PNS and CNS regeneration.


Assuntos
Proteínas I-kappa B/metabolismo , Bainha de Mielina/metabolismo , Regeneração Nervosa/fisiologia , Células de Schwann/metabolismo , Degeneração Walleriana/patologia , Análise de Variância , Animais , Axotomia/métodos , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Traumatismos do Nervo Facial/complicações , Traumatismos do Nervo Facial/etiologia , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas I-kappa B/genética , Macrófagos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Neurônios Motores/patologia , Proteína P0 da Mielina/metabolismo , Inibidor de NF-kappaB alfa , Proteínas de Neurofilamentos/metabolismo , Tempo de Reação/genética , Recuperação de Função Fisiológica/genética , Células de Schwann/patologia , Células de Schwann/ultraestrutura , Estilbamidinas , Fatores de Tempo , Degeneração Walleriana/etiologia
9.
Cells ; 11(21)2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36359758

RESUMO

Neurological disorders are highly prevalent and often lead to chronic debilitating disease. Neuroinflammation is a major driver across the spectrum of disorders, and microglia are key mediators of this response, gaining wide acceptance as a druggable cell target. Moreover, clinical providers have limited ability to objectively quantify patient-specific changes in microglia status, which can be a predictor of illness and recovery. This necessitates the development of diagnostic biomarkers and imaging techniques to monitor microglia-mediated neuroinflammation in coordination with neurological outcomes. New insights into the polarization status of microglia have shed light on the regulation of disease progression and helped identify a modifiable target for therapeutics. Thus, the detection and monitoring of microglia activation through the inclusion of diagnostic biomarkers and imaging techniques will provide clinical tools to aid our understanding of the neurologic sequelae and improve long-term clinical care for patients. Recent achievements demonstrated by pre-clinical studies, using novel depletion and cell-targeted approaches as well as single-cell RNAseq, underscore the mechanistic players that coordinate microglial activation status and offer a future avenue for therapeutic intervention.


Assuntos
Microglia , Doenças do Sistema Nervoso , Animais , Camundongos , Humanos , Microglia/fisiologia , Camundongos Endogâmicos C57BL , Células Mieloides , Biomarcadores
10.
Stem Cell Reports ; 17(9): 2097-2110, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-35985331

RESUMO

In the past decade it has become evident that neuroblasts continue to supply the human cortex with interneurons via unique migratory streams shortly following birth. Owing to the size of the human brain, these newborn neurons must migrate long distances through complex cellular landscapes to reach their final locations. This process is poorly understood, largely because of technical difficulties in acquiring and studying neurotypical postmortem human samples along with diverging developmental features of well-studied mouse models. We reasoned that migratory streams of neuroblasts utilize cellular substrates, such as blood vessels, to guide their trek from the subventricular zone to distant cortical targets. Here, we evaluate the association between young interneuronal migratory streams and their preferred cellular substrates in gyrencephalic piglets during the developmental equivalent of human birth, infancy, and toddlerhood.


Assuntos
Células-Tronco Neurais , Animais , Encéfalo , Movimento Celular/fisiologia , Humanos , Ventrículos Laterais , Camundongos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Bulbo Olfatório , Suínos
11.
Am J Orthod Dentofacial Orthop ; 140(6): 762-70, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22133940

RESUMO

INTRODUCTION: Chronic snoring is considered abnormal in a pediatric population. This disorder is often attributed to enlarged tonsils and adenoids, but multiple anatomic obstructions should also be considered. Facial and dental morphometry associations with various sleep-disordered breathing symptoms were investigated at an orthodontic clinic. METHODS: Parents or guardians were asked to complete a 4-part questionnaire on behalf of their children (n = 604; <18 years of age), including medical and dental history, bruxism and temporomandibular disorder habits, sleep and daytime behavior, and sleep duration and quality. All subjects underwent a clinical screening assessment by the same orthodontist to identify standard dental, skeletal, functional, and esthetic factors. RESULTS: In contrast to sleep-disordered breathing or sleep apnea in adults, which is predominantly associated with obesity, sleep-disordered breathing symptoms in this pediatric cohort were primarily associated with adenotonsillar hypertrophy, morphologic features related to a long and narrow face (dolichofacial, high mandibular plane angle, narrow palate, and severe crowding in the maxilla and the mandible), allergies, frequent colds, and habitual mouth breathing. CONCLUSIONS: Because of the recognized impact of pediatric snoring on children's health, the determination of these good predictors can help in preventing and managing sleep-disordered breathing. If a health professional notices signs and symptoms of sleep-disordered breathing, the young patient should be referred to a sleep medicine specialist in conjunction with an orthodontist if there are dentoskeletal abnormalities.


Assuntos
Tonsila Faríngea/patologia , Face/anatomia & histologia , Má Oclusão/complicações , Respiração Bucal/etiologia , Tonsila Palatina/patologia , Síndromes da Apneia do Sono/etiologia , Ronco/etiologia , Adolescente , Bruxismo/complicações , Distribuição de Qui-Quadrado , Criança , Estudos Transversais , Feminino , Humanos , Hipertrofia/complicações , Masculino , Mandíbula/anatomia & histologia , Programas de Rastreamento , Obstrução Nasal/complicações , Razão de Chances , Palato Duro/patologia , Inquéritos e Questionários , Dimensão Vertical
12.
Front Cell Neurosci ; 15: 807170, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35027884

RESUMO

Abnormalities in the prefrontal cortex (PFC), as well as the underlying white matter (WM) tracts, lie at the intersection of many neurodevelopmental disorders. The influence of microorganisms on brain development has recently been brought into the clinical and research spotlight as alterations in commensal microbiota are implicated in such disorders, including autism spectrum disorders, schizophrenia, depression, and anxiety via the gut-brain axis. In addition, gut dysbiosis is common in preterm birth patients who often display diffuse WM injury and delayed WM maturation in critical tracts including those within the PFC and corpus callosum. Microbial colonization of the gut aligns with ongoing postnatal processes of oligodendrogenesis and the peak of brain myelination in humans; however, the influence of microbiota on gyral WM development remains elusive. Here, we develop and validate a neonatal germ-free swine model to address these issues, as piglets share key similarities in WM volume, developmental trajectories, and distribution to humans. We find significant region-specific reductions, and sexually dimorphic trends, in WM volume, oligodendrogenesis, and mature oligodendrocyte numbers in germ-free piglets during a key postnatal epoch of myelination. Our findings indicate that microbiota plays a critical role in promoting WM development during early life when the brain is vulnerable to environmental insults that can result in an array of disabilities manifesting later in life.

13.
J Neurosci Methods ; 334: 108594, 2020 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-31945400

RESUMO

Modern clearing techniques enable high resolution visualization and 3D reconstruction of cell populations and their structural details throughout large biological samples, including intact organs and even entire organisms. In the past decade, these methods have become more tractable and are now being utilized to provide unforeseen insights into the complexities of the nervous system. While several iterations of optical clearing techniques have been developed, some are more suitable for specific applications than others depending on the type of specimen under study. Here we review findings from select studies utilizing clearing methods to visualize the developing, injured, and diseased nervous system within numerous model systems and species. We note trends and imbalances in the types of research questions being addressed with clearing methods across these fields in neuroscience. In addition, we discuss restrictions in applying optical clearing methods for postmortem tissue from humans and large animals and emphasize the lack in continuity between studies of these species. We aim for this review to serve as a key outline of available tissue clearing methods used successfully to address issues across neuronal development, injury/repair, and aging/disease.

14.
J Am Heart Assoc ; 8(15): e012711, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31331224

RESUMO

Background Reduced oxygen delivery in congenital heart disease causes delayed brain maturation and white matter abnormalities in utero. No treatment currently exists. Tetrahydrobiopterin (BH4) is a cofactor for neuronal nitric oxide synthase. BH4 availability is reduced upon NOS activation, such as during hypoxic conditions, and leads to toxin production. We hypothesize that BH4 levels are depleted in the hypoxic brain and that BH4 replacement therapy mitigates the toxic effects of hypoxia on white matter. Methods and Results Transgenic mice were used to visualize oligodendrocytes. Hypoxia was introduced during a period of white matter development equivalent to the human third trimester. BH4 was administered during hypoxia. BH4 levels were depleted in the hypoxic brain by direct quantification (n=7-12). The proliferation (n=3-6), apoptosis (n=3-6), and developmental stage (n=5-8) of oligodendrocytes were determined immunohistologically. Total oligodendrocytes increased after hypoxia, consistent with hypoxia-induced proliferation seen previously; however, mature oligodendrocytes were less prevalent in hypoxia, and there was accumulation of immature oligodendrocytes. BH4 treatment improved the mature oligodendrocyte number such that it did not differ from normoxia, and accumulation of immature oligodendrocytes was not observed. These results persisted beyond the initial period of hypoxia (n=3-4). Apoptosis increased with hypoxia but decreased with BH4 treatment to normoxic levels. White matter myelin levels decreased following hypoxia by western blot. BH4 treatment normalized myelination (n=6-10). Hypoxia worsened sensory-motor coordination on balance beam tasks, and BH4 therapy normalized performance (n=5-9). Conclusions Suboptimal BH4 levels influence hypoxic white matter abnormalities. Repurposing BH4 for use during fetal brain development may limit white matter dysmaturation in congenital heart disease.


Assuntos
Biopterinas/análogos & derivados , Doenças Fetais/fisiopatologia , Cardiopatias/congênito , Cardiopatias/fisiopatologia , Hipóxia/fisiopatologia , Substância Branca/efeitos dos fármacos , Substância Branca/crescimento & desenvolvimento , Animais , Biopterinas/farmacologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Transgênicos
15.
Cell Rep ; 29(1): 225-235.e5, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577952

RESUMO

PINK1 and Parkin are established mediators of mitophagy, the selective removal of damaged mitochondria by autophagy. PINK1 and Parkin have been proposed to act as tumor suppressors, as loss-of-function mutations are correlated with enhanced tumorigenesis. However, it is unclear how PINK1 and Parkin act in coordination during mitophagy to influence the cell cycle. Here we show that PINK1 and Parkin genetically interact with proteins involved in cell cycle regulation, and loss of PINK1 and Parkin accelerates cell growth. PINK1- and Parkin-mediated activation of TBK1 at the mitochondria during mitophagy leads to a block in mitosis due to the sequestration of TBK1 from its physiological role at centrosomes during mitosis. Our study supports a diverse role for the far-reaching, regulatory effects of mitochondrial quality control in cellular homeostasis and demonstrates that the PINK1/Parkin pathway genetically interacts with the cell cycle, providing a framework for understanding the molecular basis linking PINK1 and Parkin to mitosis.


Assuntos
Ciclo Celular/genética , Mitocôndrias/genética , Mitose/genética , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Ubiquitina-Proteína Ligases/genética , Autofagia/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Células HCT116 , Células HEK293 , Células HeLa , Homeostase/genética , Humanos , Mitofagia/genética
16.
Cell Death Differ ; 24(2): 288-299, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27911443

RESUMO

We observed that the transient induction of mtDNA double strand breaks (DSBs) in cultured cells led to activation of cell cycle arrest proteins (p21/p53 pathway) and decreased cell growth, mediated through reactive oxygen species (ROS). To investigate this process in vivo we developed a mouse model where we could transiently induce mtDNA DSBs ubiquitously. This transient mtDNA damage in mice caused an accelerated aging phenotype, preferentially affecting proliferating tissues. One of the earliest phenotypes was accelerated thymus shrinkage by apoptosis and differentiation into adipose tissue, mimicking age-related thymic involution. This phenotype was accompanied by increased ROS and activation of cell cycle arrest proteins. Treatment with antioxidants improved the phenotype but the knocking out of p21 or p53 did not. Our results demonstrate that transient mtDNA DSBs can accelerate aging of certain tissues by increasing ROS. Surprisingly, this mtDNA DSB-associated senescence phenotype does not require p21/p53, even if this pathway is activated in the process.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA Mitocondrial/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilcisteína/farmacologia , Envelhecimento , Animais , Apoptose , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mifepristona/toxicidade , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Timócitos/citologia , Timócitos/efeitos dos fármacos , Timócitos/metabolismo , Proteína Supressora de Tumor p53/genética
17.
Sci Transl Med ; 9(374)2017 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28123074

RESUMO

Long-term neurological deficits due to immature cortical development are emerging as a major challenge in congenital heart disease (CHD). However, cellular mechanisms underlying dysregulation of perinatal corticogenesis in CHD remain elusive. The subventricular zone (SVZ) represents the largest postnatal niche of neural stem/progenitor cells (NSPCs). We show that the piglet SVZ resembles its human counterpart and displays robust postnatal neurogenesis. We present evidence that SVZ NSPCs migrate to the frontal cortex and differentiate into interneurons in a region-specific manner. Hypoxic exposure of the gyrencephalic piglet brain recapitulates CHD-induced impaired cortical development. Hypoxia reduces proliferation and neurogenesis in the SVZ, which is accompanied by reduced cortical growth. We demonstrate a similar reduction in neuroblasts within the SVZ of human infants born with CHD. Our findings demonstrate that SVZ NSPCs contribute to perinatal corticogenesis and suggest that restoration of SVZ NSPCs' neurogenic potential is a candidate therapeutic target for improving cortical growth in CHD.


Assuntos
Lobo Frontal/patologia , Insuficiência Cardíaca/patologia , Neurogênese , Animais , Animais Recém-Nascidos , Movimento Celular , Lobo Frontal/crescimento & desenvolvimento , Insuficiência Cardíaca/congênito , Hipóxia/patologia , Interneurônios/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Suínos
18.
Sci Rep ; 7(1): 17624, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29247175

RESUMO

Local and controlled delivery of therapeutic agents directly into focally afflicted tissues is the ideal for the treatment of diseases that require direct interventions. However, current options are obtrusive, difficult to implement, and limited in their scope of utilization; the optimal solution requires a method that may be optimized for available therapies and is designed for exact delivery. To address these needs, we propose the Biocage, a customizable implantable local drug delivery platform. The device is a needle-sized porous container capable of encasing therapeutic molecules and matrices of interest to be eluted into the region of interest over time. The Biocage was fabricated using the Nanoscribe Photonic Professional GT 3D laser lithography system, a two-photon polymerization (2PP) 3D printer capable of micron-level precision on a millimeter scale. We demonstrate the build consistency and features of the fabricated device; its ability to release molecules; and a method for its accurate, stable delivery in mouse brain tissue. The Biocage provides a powerful tool for customizable and precise delivery of therapeutic agents into target tissues.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Sistemas de Liberação de Medicamentos/métodos , Preparações Farmacêuticas/administração & dosagem , Sefarose/administração & dosagem , Animais , Camundongos , Camundongos Endogâmicos C57BL , Impressão Tridimensional
19.
J Am Heart Assoc ; 6(8)2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28862938

RESUMO

BACKGROUND: Newly developed white matter (WM) injury is common after cardiopulmonary bypass (CPB) in severe/complex congenital heart disease. Fractional anisotropy (FA) allows measurement of macroscopic organization of WM pathology but has rarely been applied after CPB. The aims of our animal study were to define CPB-induced FA alterations and to determine correlations between these changes and cellular events after congenital heart disease surgery. METHODS AND RESULTS: Normal porcine WM development was first assessed between 3 and 7 weeks of age: 3-week-old piglets were randomly assigned to 1 of 3 CPB-induced insults. FA was analyzed in 31 WM structures. WM oligodendrocytes, astrocytes, and microglia were assessed immunohistologically. Normal porcine WM development resembles human WM development in early infancy. We found region-specific WM vulnerability to insults associated with CPB. FA changes after CPB were also insult dependent. Within various WM areas, WM within the frontal cortex was susceptible, suggesting that FA in the frontal cortex should be a biomarker for WM injury after CPB. FA increases occur parallel to cellular processes of WM maturation during normal development; however, they are altered following surgery. CPB-induced oligodendrocyte dysmaturation, astrogliosis, and microglial expansion affect these changes. FA enabled capturing CPB-induced cellular events 4 weeks postoperatively. Regions most resilient to CPB-induced FA reduction were those that maintained mature oligodendrocytes. CONCLUSIONS: Reducing alterations of oligodendrocyte development in the frontal cortex can be both a metric and a goal to improve neurodevelopmental impairment in the congenital heart disease population. Studies using this model can provide important data needed to better interpret human imaging studies.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Diferenciação Celular , Lobo Frontal/patologia , Leucoencefalopatias/etiologia , Oligodendroglia/patologia , Substância Branca/patologia , Fatores Etários , Animais , Anisotropia , Astrócitos/patologia , Biomarcadores/metabolismo , Imagem de Difusão por Ressonância Magnética , Imagem de Tensor de Difusão , Lobo Frontal/diagnóstico por imagem , Lobo Frontal/metabolismo , Imuno-Histoquímica , Leucoencefalopatias/diagnóstico por imagem , Leucoencefalopatias/metabolismo , Leucoencefalopatias/patologia , Microglia/patologia , Modelos Animais , Oligodendroglia/metabolismo , Sus scrofa , Fatores de Tempo , Substância Branca/diagnóstico por imagem , Substância Branca/metabolismo
20.
J Thorac Cardiovasc Surg ; 151(1): 265-72.e1-3, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26412317

RESUMO

OBJECTIVES: White-matter injury after surgery is common in neonates with cerebral immaturity secondary to in utero hypoxia. Astrocytes play a central role in brain protection; however, the reaction of astrocytes to hypothermic circulatory arrest (HCA) remains unknown. We investigated the role of astrocytes in white-matter injury after HCA and determined the effects of preoperative hypoxia on this role, using a novel mouse model. METHODS: Mice were exposed to hypoxia from days 3 to 11, which is equivalent to the third trimester in humans (prehypoxia, n = 49). Brain slices were transferred to a chamber perfused by cerebrospinal fluid. Oxygen-glucose deprivation (OGD) was performed to simulate ischemia-reperfusion/reoxygenation resulting from circulatory arrest under hypothermia. Astrocyte reactions were compared with preoperative normoxia (prenormoxia; n = 45). RESULTS: We observed astrocyte activation after 25°C ischemia-reperfusion/reoxygenation in prenormoxia (P < .01). Astrocyte number after OGD correlated with caspase-3(+) cells (rho = .77, P = .001), confirming that astrogliosis is an important response after HCA. At 3 hours after OGD, astrocytes in prenormoxia had already proliferated and become activated (P < .05). Conversely, astrocytes that developed under hypoxia did not display these responses. At 20 hours after ischemia-reperfusion/reoxygenation, astrogliosis was not observed in prehypoxia, demonstrating that hypoxia altered the response of astrocytes to insult. In contrast to prenormoxia, caspase-3(+) cells in prehypoxia increased after ischemia reperfusion/reoxygenation, compared with control (P < .01). Caspase-3(+) cells were more common with prehypoxia than with prenormoxia (P < .001), suggesting that lack of astrogliosis permits increased white-matter injury. CONCLUSIONS: Preoperative hypoxia alters the neuroprotective function of astrocytes. Restoring this function before surgery may be a therapeutic option to reduce postoperative white-matter injury in the immature brain.


Assuntos
Astrócitos/patologia , Hipóxia Fetal/patologia , Hipóxia-Isquemia Encefálica/patologia , Leucoencefalopatias/patologia , Traumatismo por Reperfusão/patologia , Substância Branca/patologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Caspase 3/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Hipóxia Fetal/metabolismo , Proteína Glial Fibrilar Ácida/genética , Gliose , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/prevenção & controle , Técnicas In Vitro , Leucoencefalopatias/metabolismo , Leucoencefalopatias/prevenção & controle , Camundongos Transgênicos , Regiões Promotoras Genéticas , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Fatores de Tempo , Substância Branca/crescimento & desenvolvimento , Substância Branca/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA