Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37917202

RESUMO

Virophagy, the selective autophagosomal engulfment and lysosomal degradation of viral components, is crucial for neuronal cell survival and antiviral immunity. However, the mechanisms leading to viral antigen recognition and capture by autophagic machinery remain poorly understood. Here, we identified cyclin-dependent kinase-like 5 (CDKL5), known to function in neurodevelopment, as an essential regulator of virophagy. Loss-of-function mutations in CDKL5 are associated with a severe neurodevelopmental encephalopathy. We found that deletion of CDKL5 or expression of a clinically relevant pathogenic mutant of CDKL5 reduced virophagy of Sindbis virus (SINV), a neurotropic RNA virus, and increased intracellular accumulation of SINV capsid protein aggregates and cellular cytotoxicity. Cdkl5-knockout mice displayed increased viral antigen accumulation and neuronal cell death after SINV infection and enhanced lethality after infection with several neurotropic viruses. Mechanistic studies demonstrated that CDKL5 directly binds the canonical selective autophagy receptor p62 and phosphorylates p62 at T269/S272 to promote its interaction with viral capsid aggregates. We found that CDKL5-mediated phosphorylation of p62 facilitated the formation of large p62 inclusion bodies that captured viral capsids to initiate capsid targeting to autophagic machinery. Overall, these findings identify a cell-autonomous innate immune mechanism for autophagy activation to clear intracellular toxic viral protein aggregates during infection.


Assuntos
Agregados Proteicos , Vírus , Camundongos , Animais , Autofagia/genética , Fosforilação , Camundongos Knockout , Proteínas do Capsídeo , Antígenos Virais , Proteínas Serina-Treonina Quinases/genética
2.
Proc Natl Acad Sci U S A ; 120(12): e2211522120, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36917672

RESUMO

Ribosome-associated quality control (RQC) pathway is responsible for degradation of nascent polypeptides in aberrantly stalled ribosomes, and its defects may lead to neurological diseases. However, the underlying molecular mechanism of how RQC dysfunction elicits neurological disorders remains poorly understood. Here we revealed that neurons with knockout (KO) of ubiquitin ligase LTN1, a key gene in the RQC pathway, show developmental defects in neurons via upregulation of TTC3 and UFMylation signaling proteins. The abnormally enhanced TTC3 protein in Ltn1 KO neurons reduced further accumulation of translationally arrested products by preventing translation initiation of selective genes. However, the overaccumulated TTC3 protein in turn caused dendritic abnormalities and reduced surface-localized GABAA receptors during neuronal development. Ltn1 KO mice showed behavioral deficits associated with cognitive disorders, a subset of which were restored by TTC3 knockdown in medial prefrontal cortex. Together, the overactivated cellular compensatory mechanism against defective RQC through TTC3 overaccumulation induced synaptic and cognitive deficits. More broadly, these findings represent a novel cellular mechanism underlying neuronal dysfunctions triggered by exaggerated cellular stress response to accumulated abnormal translation products in neurons.


Assuntos
Disfunção Cognitiva , Ribossomos , Ubiquitina-Proteína Ligases , Animais , Camundongos , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Biossíntese de Proteínas , Ribossomos/genética , Ribossomos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
3.
Front Mol Neurosci ; 15: 893111, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35875665

RESUMO

Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.

4.
Exp Neurol ; 351: 114010, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35167826

RESUMO

Cisplatin is a member of a widely utilized class of chemotherapeutic agent that initiates DNA damage response, cell cycle arrest, and p53-dependent apoptotic cell death in concert with DNA­platinum adduct formation. While normal programmed cell death (PCD) can occur in the developing neuroepithelium in the absence of caspase-3 within certain genetic backgrounds, we observed an absolute dependency upon this executioner caspase with respect to cisplatin-induced PCD in the developing central nervous system (CNS). We therefore examined the nature of this genotoxic injury in the CNS in vivo, in which cisplatin treatment causes widespread cellular injury consistent with hallmarks of apoptosis which are averted upon caspase-3 inhibition. Examination of cisplatin-mediated injury as a function of time revealed the presence of an alternative, delayed form of necroptosis-like cell death which manifests in Casp3-/- neuroepithelia for several days following the normal pattern of apoptosis. Together, these findings suggest a coordinated regulation of these disparate PCD pathways in response to genotoxic stress in vivo and highlight the unique and critical role which caspase-3 plays among executioner caspases in coordinating apoptotic versus necroptotic responsiveness of the developing CNS to genotoxic injury.


Assuntos
Caspases , Cisplatino , Apoptose/fisiologia , Encéfalo/metabolismo , Caspase 3/metabolismo , Caspases/metabolismo , Cisplatino/toxicidade
5.
Biol Psychiatry ; 91(4): 335-345, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34836635

RESUMO

Recent genetic approaches have demonstrated that genetic factors contribute to the pathologic origins of neuropsychiatric disorders. Nevertheless, the exact pathophysiological mechanism for most cases remains unclear. Recent studies have demonstrated alterations in pathways of protein homeostasis (proteostasis) and identified several proteins that are misfolded and/or aggregated in the brains of patients with neuropsychiatric disorders, thus providing early evidence that disrupted proteostasis may be a contributing factor to their pathophysiology. Unlike neurodegenerative disorders in which massive neuronal and synaptic losses are observed, proteostasis impairments in neuropsychiatric disorders do not lead to robust neuronal death, but rather likely act via loss- and gain-of-function effects to disrupt neuronal and synaptic functions. Furthermore, abnormal activation of or overwhelmed endoplasmic reticulum and mitochondrial quality control pathways may exacerbate the pathophysiological changes initiated by impaired proteostasis, as these organelles are critical for proper neuronal functions and involved in the maintenance of proteostasis. This perspective article reviews recent findings implicating proteostasis impairments in the pathophysiology of neuropsychiatric disorders and explores how neuronal and synaptic functions may be impacted by disruptions in protein homeostasis. A greater understanding of the contributions by proteostasis impairment in neuropsychiatric disorders will help guide future studies to identify additional candidate proteins and new targets for therapeutic development.


Assuntos
Doenças Neurodegenerativas , Proteostase , Retículo Endoplasmático/metabolismo , Humanos , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Resposta a Proteínas não Dobradas
6.
Neurosci Biobehav Rev ; 110: 77-91, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30470595

RESUMO

Recent human genetics studies have identified many genetic variants that may be responsible for autism spectrum disorder (ASD). ASD mouse models with genetic modifications mimicking these rare genetic variants have provided invaluable mechanistic insights into the disruption of various biological processes and brain areas/circuitry affected in ASD patients. In this review, we begin by reviewing several mouse models for ASD-associated copy number variations (CNVs) to illustrate how they have been employed to establish causal links between their behavioral phenotypes and the affected genes. We then focus on studies using one of the principal behavioral abnormalities associated with ASD, social behavior, to identify the molecular and circuit-level deficits involved. Finally, we end by discussing other mouse models designed to probe how the disruption of specific biological processes such as autophagy and neurogenesis may contribute to ASD pathogenesis. By achieving a greater understanding of the pathophysiology and pathogenic mechanisms involved in ASD and related disorders, novel therapeutic strategies may be devised for ASD patients in the near future.


Assuntos
Transtorno do Espectro Autista/patologia , Transtorno Autístico/patologia , Lesões Encefálicas/patologia , Encéfalo/patologia , Animais , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/genética , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/genética , Encéfalo/fisiopatologia , Lesões Encefálicas/fisiopatologia , Predisposição Genética para Doença/genética , Humanos , Comportamento Social
7.
Biomolecules ; 9(11)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31683805

RESUMO

De novo protein synthesis by the ribosome and its multitude of co-factors must occur in a tightly regulated manner to ensure that the correct proteins are produced accurately at the right time and, in some cases, also in the proper location. With novel techniques such as ribosome profiling and cryogenic electron microscopy, our understanding of this basic biological process is better than ever and continues to grow. Concurrently, increasing attention is focused on how translational regulation in the brain may be disrupted during the progression of various neurological disorders. In fact, translational dysregulation is now recognized as the de facto pathogenic cause for some disorders. Novel mechanisms including ribosome stalling, ribosome-associated quality control, and liquid-liquid phase separation are closely linked to translational regulation, and may thus be involved in the pathogenic process. The relationships between translational dysregulation and neurological disorders, as well as the ways through which we may be able to reverse those detrimental effects, will be examined in this review.


Assuntos
Doenças do Sistema Nervoso/genética , Biossíntese de Proteínas , Ribossomos/genética , Animais , Encéfalo/metabolismo , Humanos , Doenças do Sistema Nervoso/metabolismo , Proteínas/genética , Proteínas/metabolismo , Ribossomos/metabolismo
8.
Health Econ Rev ; 9(1): 29, 2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31667671

RESUMO

BACKGROUND: Knowledge of the costs of health services improves health facility management and aids in health financing for universal health coverage. Because of resource requirements that are often not present in low- and middle-income countries, costing exercises are rare and infrequent. Here we report findings from the initial phase of establishing a routine costing system for health services implemented in three provinces in Cambodia. METHODS: Data was collected for the 2016 financial year from 20 health centres (including four with beds) and five hospitals (three district hospitals and two provincial hospitals). The costs to the providers for health centres were calculated using step-down allocations for selected costing units, including preventive and curative services, delivery, and patient contact, while for hospitals this was complemented with bed-day and inpatient day per department. Costs were compared by type of facility and between provinces. RESULTS: All required information was not readily available at health facilities and had to be recovered from various sources. Costs per outpatient consultation at health centres varied between provinces (from US$2.33 to US$4.89), as well as within provinces. Generally, costs were inversely correlated with the quantity of service output. Costs per contact were higher at health centres with beds than health centres without beds (US$4.59, compared to US$3.00). Conversely, costs for delivery were lower in health centres with beds (US$128.7, compared to US$413.7), mainly because of low performing health centres without beds. Costs per inpatient-day varied from US$27.61 to US$55.87 and were most expensive at the lowest level hospital. CONCLUSIONS: Establishing a routine health service costing system appears feasible if recording and accounting procedures are improved. Information on service costs by health facility level can provide useful information to optimise the use of available financial and human resources.

9.
Autophagy ; 15(10): 1848-1849, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31280658

RESUMO

The disruption of MTOR-regulated macroautophagy/autophagy was previously shown to cause autistic-like abnormalities; however, the underlying molecular defects remained largely unresolved. In a recent study, we demonstrated that autophagy deficiency induced by conditional Atg7 deletion in either forebrain GABAergic inhibitory or excitatory neurons leads to a similar set of autistic-like behavioral abnormalities even when induced following the peak period of synaptic pruning during postnatal neurodevelopment. Our proteomic analysis and molecular dissection further revealed a mechanism in which the GABAA receptor trafficking function of GABARAP (gamma-aminobutyric acid receptor associated protein) family proteins was compromised as they became sequestered by SQSTM1/p62-positive aggregates formed due to autophagy deficiency. Our discovery of autophagy as a link between MTOR and GABA signaling may have implications not limited to neurodevelopmental and neuropsychiatric disorders, but could potentially be involved in other human pathologies such as cancer and diabetes in which both pathways are implicated.


Assuntos
Autofagia/fisiologia , Encéfalo/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Encéfalo/patologia , Humanos , Camundongos , Camundongos Knockout , Receptores de GABA-A/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética
10.
Sci Adv ; 5(4): eaau8237, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30989111

RESUMO

Dysfunctional mTOR signaling is associated with the pathogenesis of neurodevelopmental and neuropsychiatric disorders. However, it is unclear what molecular mechanisms and pathogenic mediators are involved and whether mTOR-regulated autophagy continues to be crucial beyond neurodevelopment. Here, we selectively deleted Atg7 in forebrain GABAergic interneurons in adolescent mice and unexpectedly found that these mice showed a set of behavioral deficits similar to Atg7 deletion in forebrain excitatory neurons. By unbiased quantitative proteomic analysis, we identified γ-aminobutyric acid receptor-associated protein-like 2 (GABARAPL2) to differentially form high-molecular weight species in autophagy-deficient brains. Further functional analyses revealed a novel pathogenic mechanism involving the p62-dependent sequestration of GABARAP family proteins, leading to the reduction of surface GABAA receptor levels. Our work demonstrates a novel physiological role for autophagy in regulating GABA signaling beyond postnatal neurodevelopment, providing a potential mechanism for the reduced inhibitory inputs observed in neurodevelopmental and neuropsychiatric disorders with mTOR hyperactivation.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Encéfalo/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Receptores de GABA-A/metabolismo , Comportamento Social , Animais , Humanos , Interneurônios/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Prosencéfalo/fisiologia , Agregados Proteicos , Ligação Proteica , Transporte Proteico
11.
J Cell Mol Med ; 23(3): 1784-1797, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30548903

RESUMO

Aberrant regulation of programmed cell death (PCD) has been tied to an array of human pathologies ranging from cancers to autoimmune disorders to diverse forms of neurodegeneration. Pharmacologic modulation of PCD signalling is therefore of central interest to a number of clinical and biomedical applications. A key component of PCD signalling involves the modulation of pro- and anti-apoptotic Bcl-2 family members. Among these, Bax translocation represents a critical regulatory phase in PCD. In the present study, we have employed a high-content high-throughput screen to identify small molecules which inhibit the cellular process of Bax re-distribution to the mitochondria following commitment of the cell to die. Screening of 6246 Generally Recognized As Safe compounds from four chemical libraries post-induction of cisplatin-mediated PCD resulted in the identification of 18 compounds which significantly reduced levels of Bax translocation. Further examination revealed protective effects via reduction of executioner caspase activity and enhanced mitochondrial function. Consistent with their effects on Bax translocation, these compounds exhibited significant rescue against in vitro and in vivo cisplatin-induced apoptosis. Altogether, our findings identify a new set of clinically useful small molecules PCD inhibitors and highlight the role which cAMP plays in regulating Bax-mediated PCD.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas de Fluorescência Verde/antagonistas & inibidores , Ensaios de Triagem em Larga Escala/métodos , Transporte Proteico/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína X Associada a bcl-2/antagonistas & inibidores , Animais , Células CHO , Cricetulus , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína X Associada a bcl-2/metabolismo
12.
Biol Psychiatry ; 84(7): 509-521, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29752072

RESUMO

BACKGROUND: Neurodegenerative diseases involving protein aggregation often accompany psychiatric symptoms. Frontotemporal lobar degeneration (FTLD) associated with TAR DNA-binding protein 43 (TDP-43) aggregation is characterized by progressive neuronal atrophy in frontal and temporal lobes of cerebral cortex. Furthermore, patients with FTLD display mental dysfunction in multiple behavioral dimensions. Nevertheless, their molecular origin for psychiatric symptoms remains unclear. METHODS: In FTLD neurons and mouse models with TDP-43 aggregates, we examined coaggregation between TDP-43 and disrupted in schizophrenia 1 (DISC1), a key player in the pathology of mental conditions and its effects on local translation in dendrites and psychiatric behaviors. The protein coaggregation and the expression level of synaptic proteins were also investigated with postmortem brains from patients with FTLD (n = 6). RESULTS: We found cytosolic TDP-43/DISC1 coaggregates in brains of both FTLD mouse model and patients with FTLD. At the mechanistic levels, the TDP-43/DISC1 coaggregates disrupted the activity-dependent dendritic local translation through impairment of translation initiation and, in turn, reduced synaptic protein expression. Behavioral deficits detected in FTLD model mice were ameliorated by exogenous DISC1 expression. CONCLUSIONS: Our findings reveal a novel role of the aggregate-prone TDP-43/DISC1 protein complex in regulating local translation, which affects aberrant behaviors relevant to multiple psychiatric dimensions.


Assuntos
Comportamento Animal , Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , Biossíntese de Proteínas , Animais , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Degeneração Lobar Frontotemporal/fisiopatologia , Humanos , Camundongos
13.
J Clin Invest ; 127(4): 1438-1450, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28263187

RESUMO

Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Doença de Huntington/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Agregação Patológica de Proteínas/enzimologia , Animais , Feminino , Células HEK293 , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Camundongos Transgênicos , Mutação
14.
Am J Pathol ; 185(2): 305-13, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25433221

RESUMO

Alzheimer disease (AD) is biochemically characterized by increased levels of amyloid ß (Aß) peptide, which aggregates into extracellular Aß plaques in AD brains. Before plaque formation, Aß accumulates intracellularly in both AD brains and in the brains of AD model mice, which may contribute to disease progression. Autophagy, which is impaired in AD, clears cellular protein aggregates and participates in Aß metabolism. In addition to a degradative role of autophagy in Aß metabolism we recently showed that Aß secretion is inhibited in mice lacking autophagy-related gene 7 (Atg7) in excitatory neurons in the mouse forebrain. This inhibition of Aß secretion leads to intracellular accumulation of Aß. Here, we used fluorescence and immunoelectron microscopy to elucidate the subcellular localization of the intracellular Aß accumulation which accumulates in Aß precursor protein mice lacking Atg7. Autophagy deficiency causes accumulation of p62(+) aggregates, but these aggregates do not contain Aß. However, knockdown of Atg7 induced Aß accumulation in the Golgi and a concomitant reduction of Aß in the multivesicular bodies. This indicates that Atg7 influences the transport of Aß possibly derived from Golgi to multivesicular bodies.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Complexo de Golgi/metabolismo , Proteínas Associadas aos Microtúbulos/deficiência , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Proteína 7 Relacionada à Autofagia , Complexo de Golgi/genética , Complexo de Golgi/patologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Fragmentos de Peptídeos/genética
15.
Cell Rep ; 5(1): 61-9, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24095740

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease biochemically characterized by aberrant protein aggregation, including amyloid beta (Aß) peptide accumulation. Protein aggregates in the cell are cleared by autophagy, a mechanism impaired in AD. To investigate the role of autophagy in Aß pathology in vivo, we crossed amyloid precursor protein (APP) transgenic mice with mice lacking autophagy in excitatory forebrain neurons obtained by conditional knockout of autophagy-related protein 7. Remarkably, autophagy deficiency drastically reduced extracellular Aß plaque burden. This reduction of Aß plaque load was due to inhibition of Aß secretion, which led to aberrant intraneuronal Aß accumulation in the perinuclear region. Moreover, autophagy-deficiency-induced neurodegeneration was exacerbated by amyloidosis, which together severely impaired memory. Our results establish a function for autophagy in Aß metabolism: autophagy influences secretion of Aß to the extracellular space and thereby directly affects Aß plaque formation, a pathological hallmark of AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Autofagia/fisiologia , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia
16.
Nanomedicine ; 9(6): 795-805, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23434679

RESUMO

Matrigel, a mouse sarcoma-derived basement membrane protein mixture, is frequently used to facilitate human tumor xenograft growth in rodents. Despite its known effects on tumor growth and metastasis, its impact on tumor pathophysiology and preclinical evaluation of nanomedicines in tumor xenografts has not been reported previously. Herein bilateral MDA435 tumors were established orthotopically with (Mat+) or without (Mat-) co-injection of Matrigel. Tumor perfusion, morphology and nanoparticle retention were evaluated. As compared to Mat- tumors, Mat+tumors exhibited enhanced vascular perfusion and lymphatic flow, greater blood vessel and lymphatic growth within the tumor core, and more deformation and collapse of lymphatics in tumor-associated lymph nodes. These changes were accompanied by reduced nanoparticle retention in Mat+tumors. The results suggest that Matrigel is not a passive medium for tumor growth, but rather significantly alters long-term tumor architecture. These findings have significant implications for the evaluation of therapeutic nanomedicine in xenograft mouse models. FROM THE CLINICAL EDITOR: Matrigel is utilized in facilitating human tumor xenograft growth in rodents. The authors demonstrate that Matrigel is not a passive medium for tumor growth; instead it significantly alters long-term tumor architecture, with major implications in the evaluation of therapeutic nanomedicine in xenograft mouse models.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias da Mama/fisiopatologia , Colágeno/administração & dosagem , Xenoenxertos/fisiopatologia , Laminina/administração & dosagem , Proteoglicanas/administração & dosagem , Animais , Linhagem Celular Tumoral , Colágeno/metabolismo , Combinação de Medicamentos , Feminino , Humanos , Laminina/metabolismo , Camundongos , Nanomedicina , Nanopartículas/administração & dosagem , Nanopartículas/química , Proteoglicanas/metabolismo
17.
J Cell Mol Med ; 14(3): 671-86, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19243469

RESUMO

The immunosuppressive agents cyclosporin A (CsA) and FK-506 have previously been shown to exhibit neurotrophic and neuroprotective properties in vivo. Given that significant clinical expertise exists for both drugs, they represent an attractive starting point for treatment of acute neural injuries. One putative mechanism for neuroprotection by these drugs relates to inhibition of calcineurin activity. However each drug-immunophilin complex can potentially influence additional signal transduction pathways. Furthermore, several non-immunosuppressive immunophilin ligands have been described as possessing neuroprotective properties, suggesting that neuroprotection may be separable from calcineurin inhibition. In the present study, we examined the mechanism of this neuroprotection in facial motor neurons following axotomy-induced injury. Similar to previous studies in rats, CsA and FK-506 enhanced motor neuron survival in mice following acute injury. To examine the mechanism responsible for neuroprotection by these agents, pharmacologic inhibitors of several potential alternate signalling pathways (17-(allylamino)-17-demethoxygeldanamycin, rapamycin, cypermethrin) were evaluated with respect to neuroprotection. Of these, only cypermethrin, a direct calcineurin inhibitor not previously associated with neuronal survival properties, was observed to significantly enhance motor neuron survival following injury. The results demonstrate for the first time that direct inhibition of calcineurin is neuroprotective in vivo. These data support a model in which calcineurin inhibition promotes neuronal survival, distinct from effects upon neurite outgrowth.


Assuntos
Inibidores de Calcineurina , Neurônios Motores/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Piretrinas/farmacologia , Animais , Animais Recém-Nascidos , Axotomia , Calcineurina/genética , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Imuno-Histoquímica , Imunossupressores/farmacologia , Inseticidas/farmacologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Endogâmicos , Camundongos Knockout , Microscopia de Fluorescência , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Transdução de Sinais/efeitos dos fármacos , Tacrolimo/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...