Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nucleic Acids Res ; 52(12): 6733-6747, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38828787

RESUMO

Adenosine Deaminases Acting on RNA (ADARs) are enzymes that catalyze the conversion of adenosine to inosine in RNA duplexes. These enzymes can be harnessed to correct disease-causing G-to-A mutations in the transcriptome because inosine is translated as guanosine. Guide RNAs (gRNAs) can be used to direct the ADAR reaction to specific sites. Chemical modification of ADAR guide strands is required to facilitate delivery, increase metabolic stability, and increase the efficiency and selectivity of the editing reaction. Here, we show the ADAR reaction is highly sensitive to ribose modifications (e.g. 4'-C-methylation and Locked Nucleic Acid (LNA) substitution) at specific positions within the guide strand. Our studies were enabled by the synthesis of RNA containing a new, ribose-modified nucleoside analog (4'-C-methyladenosine). Importantly, the ADAR reaction is potently inhibited by LNA or 4'-C-methylation at different positions in the ADAR guide. While LNA at guide strand positions -1 and -2 block the ADAR reaction, 4'-C-methylation only inhibits at the -2 position. These effects are rationalized using high-resolution structures of ADAR-RNA complexes. This work sheds additional light on the mechanism of ADAR deamination and aids in the design of highly selective ADAR guide strands for therapeutic editing using chemically modified RNA.


Assuntos
Adenosina Desaminase , Edição de RNA , Ribose , Adenosina Desaminase/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/química , Ribose/química , Ribose/metabolismo , Humanos , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/química , Metilação , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/química , Nucleosídeos/química , Nucleosídeos/metabolismo , RNA/metabolismo , RNA/química , Inosina/metabolismo , Inosina/química
2.
Front Genome Ed ; 6: 1304110, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38510848

RESUMO

Epigenetics refers to the molecules and mechanisms that modify gene expression states without changing the nucleotide context. These modifications are what encode the cell state during differentiation or epigenetic memory in mitosis. Epigenetic modifications can alter gene expression by changing the chromatin architecture by altering the affinity for DNA to wrap around histone octamers, forming nucleosomes. The higher affinity the DNA has for the histones, the tighter it will wrap and therefore induce a heterochromatin state, silencing gene expression. Several groups have shown the ability to harness the cell's natural epigenetic modification pathways to engineer proteins that can induce changes in epigenetics and consequently regulate gene expression. Therefore, epigenetic modification can be used to target and treat disorders through the modification of endogenous gene expression. The use of epigenetic modifications may prove an effective path towards regulating gene expression to potentially correct or cure genetic disorders.

4.
Mol Ther ; 31(4): 1088-1105, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-36641623

RESUMO

Angelman syndrome (AS) is a neurogenetic disorder caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. The UBE3A gene is paternally imprinted in brain neurons. Clinical features of AS are primarily due to the loss of maternally expressed UBE3A in the brain. A healthy copy of paternal UBE3A is present in the brain but is silenced by a long non-coding antisense transcript (UBE3A-ATS). Here, we demonstrate that an artificial transcription factor (ATF-S1K) can silence Ube3a-ATS in an adult mouse model of Angelman syndrome (AS) and restore endogenous physiological expression of paternal Ube3a. A single injection of adeno-associated virus (AAV) expressing ATF-S1K (AAV-S1K) into the tail vein enabled whole-brain transduction and restored UBE3A protein in neurons to ∼25% of wild-type protein. The ATF-S1K treatment was highly specific to the target site with no detectable inflammatory response 5 weeks after AAV-S1K administration. AAV-S1K treatment of AS mice showed behavioral rescue in exploratory locomotion, a task involving gross and fine motor abilities, similar to low ambulation and velocity in AS patients. The specificity and tolerability of a single injection of AAV-S1K therapy for AS demonstrate the use of ATFs as a promising translational approach for AS.


Assuntos
Síndrome de Angelman , Animais , Camundongos , Síndrome de Angelman/genética , Síndrome de Angelman/terapia , Síndrome de Angelman/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição/genética , Fenótipo , Ubiquitina-Proteína Ligases/genética
5.
Diagnostics (Basel) ; 12(4)2022 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-35453845

RESUMO

Background: Novel, non-invasive diagnostic biomarkers that facilitate early intervention in head and neck cancer are urgently needed. Polyamine metabolites have been observed to be elevated in numerous cancer types and correlated with poor prognosis. The aim of this study was to assess the concentration of polyamines in the saliva and urine from head and neck cancer (HNC) patients, compared to healthy controls. Methods: Targeted metabolomic analysis was performed on saliva and urine from 39 HNC patient samples and compared to 89 healthy controls using a quantitative, targeted liquid chromatography mass spectrometry approach. Results: The metabolites N1-acetylspermine (ASP), N8-acetylspermidine (ASD) and N1,N12-diacetylspermine (DAS) were detected at significantly different concentrations in the urine of HNC patients as compared to healthy controls. Only ASP was detected at elevated levels in HNC saliva as compared to healthy controls. Conclusion: These data suggest that assessment of polyamine-based metabolite biomarkers within the saliva and urine warrants further investigation as a potential diagnostic in HNC patients.

6.
Hum Mol Genet ; 31(18): 3032-3050, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-35445702

RESUMO

Many neurodevelopmental disorders (NDDs) are the result of mutations on the X chromosome. One severe NDD resulting from mutations on the X chromosome is CDKL5 deficiency disorder (CDD). CDD is an epigenetic, X-linked NDD characterized by intellectual disability (ID), pervasive seizures and severe sleep disruption, including recurring hospitalizations. CDD occurs at a 4:1 ratio, with a female bias. CDD is driven by the loss of cyclin-dependent kinase-like 5 (CDKL5), a serine/threonine kinase that is essential for typical brain development, synapse formation and signal transmission. Previous studies focused on male subjects from animal models, likely to avoid the complexity of X mosaicism. For the first time, we report translationally relevant behavioral phenotypes in young adult (8-20 weeks) females and males with robust signal size, including impairments in learning and memory, substantial hyperactivity and increased susceptibility to seizures/reduced seizure thresholds, in both sexes, and in two models of CDD preclinical mice, one with a general loss-of-function mutation and one that is a patient-derived mutation.


Assuntos
Quinases Ciclina-Dependentes , Animais , Cognição , Quinases Ciclina-Dependentes/deficiência , Síndromes Epilépticas , Feminino , Humanos , Masculino , Camundongos , Proteínas Serina-Treonina Quinases/genética , Convulsões/genética , Serina
7.
Neurotherapeutics ; 18(3): 1535-1547, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34528170

RESUMO

Angelman syndrome (AS) is a rare (~1:15,000) neurodevelopmental disorder characterized by severe developmental delay and intellectual disability, impaired communication skills, and a high prevalence of seizures, sleep disturbances, ataxia, motor deficits, and microcephaly. AS is caused by loss-of-function of the maternally inherited UBE3A gene. UBE3A is located on chromosome 15q11-13 and is biallelically expressed throughout the body but only maternally expressed in the brain due to an RNA antisense transcript that silences the paternal copy. There is currently no cure for AS, but advancements in small molecule drugs and gene therapies offer a promising approach for the treatment of the disorder. Here, we review AS and how loss-of-function of the maternal UBE3A contributes to the disorder. We also discuss the strengths and limitations of current animal models of AS. Furthermore, we examine potential small molecule drug and gene therapies for the treatment of AS and associated challenges faced by the therapeutic design. Finally, gene therapy offers the opportunity for precision medicine in AS and advancements in the treatment of this disorder can serve as a foundation for other single-gene neurodevelopmental disorders.


Assuntos
Síndrome de Angelman/genética , Síndrome de Angelman/terapia , Anticonvulsivantes/uso terapêutico , Terapia Genética/métodos , Animais , Produtos Biológicos/uso terapêutico , Humanos , Isoxazóis/uso terapêutico , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/terapia , RNA Antissenso/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Ubiquitina-Proteína Ligases/genética
8.
Curr Neuropharmacol ; 19(12): 2125-2140, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33998992

RESUMO

Advancements in programmable DNA-Binding Proteins (DBDs) that target the genome, such as zinc fingers, transcription activator-like effectors, and Cas9, have broadened drug target design beyond traditional protein substrates. Effective delivery methodologies remain a major barrier in targeting the central nervous system. Currently, adeno-associated virus is the most wellvalidated delivery system for the delivery of DBDs towards the central nervous with multiple, ongoing clinical trials. While effective in transducing neuronal cells, viral delivery systems for DBDs remain problematic due to inherent viral packaging limits or immune responses that hinder translational potential. Direct administration of DBDs or encapsulation in lipid nanoparticles may provide alternative means towards delivering gene therapies into the central nervous system. This review will evaluate the strengths and limitations of current DBD delivery strategies in vivo. Furthermore, this review will discuss the use of adult stem cells as a putative delivery vehicle for DBDs and the potential advantages that these systems have over previous methodologies.


Assuntos
Edição de Genes , Nanopartículas , Sistema Nervoso Central , DNA , Lipossomos
9.
Hum Mol Genet ; 30(12): 1067-1083, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33856035

RESUMO

Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by impaired communication skills, ataxia, motor and balance deficits, intellectual disabilities, and seizures. The genetic cause of AS is the neuronal loss of UBE3A expression in the brain. A novel approach, described here, is a stem cell gene therapy which uses lentivector-transduced hematopoietic stem and progenitor cells to deliver functional UBE3A to affected cells. We have demonstrated both the prevention and reversal of AS phenotypes upon transplantation and engraftment of human CD34+ cells transduced with a Ube3a lentivector in a novel immunodeficient Ube3amat-/pat+ IL2rg-/y mouse model of AS. A significant improvement in motor and cognitive behavioral assays as well as normalized delta power measured by electroencephalogram was observed in neonates and adults transplanted with the gene modified cells. Human hematopoietic profiles observed in the lymphoid organs by detection of human immune cells were normal. Expression of UBE3A was detected in the brains of the adult treatment group following immunohistochemical staining illustrating engraftment of the gene-modified cells expressing UBE3A in the brain. As demonstrated with our data, this stem cell gene therapy approach offers a promising treatment strategy for AS, not requiring a critical treatment window.


Assuntos
Síndrome de Angelman/terapia , Terapia Genética , Deficiência Intelectual/terapia , Convulsões/terapia , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/genética , Síndrome de Angelman/patologia , Animais , Antígenos CD34/genética , Ataxia/genética , Ataxia/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/terapia , Modelos Animais de Doenças , Eletroencefalografia , Regulação da Expressão Gênica/genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Humanos , Deficiência Intelectual/genética , Interleucina-2/genética , Lentivirus/genética , Camundongos , Transtornos das Habilidades Motoras/genética , Transtornos das Habilidades Motoras/patologia , Transtornos das Habilidades Motoras/terapia , Convulsões/genética
10.
Stem Cells Transl Med ; 10(7): 1033-1043, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33710799

RESUMO

Huntington's disease (HD) is a fatal autosomal-dominant neurodegenerative disease caused by a trinucleotide CAG repeat expansion of the huntingtin gene (HTT) that affects 1 in every 10 000 individuals in the United States. Our lab developed a novel immune deficient HD mouse strain, the YACNSG, from a commonly used line, the YAC128 mouse, to enable transplantation studies using engineered human cells in addition to studying the impact of the immune system on disease progression. The primary goal of this project was to characterize this novel immune deQficient HD mouse model, using behavioral assays and histology to compare this new model to the immune competent YAC128 and immune deficient mice that had engraftment of a human immune system. Flow cytometry was used to confirm that the YACNSG strain lacked immune cells, and in vivo imaging was used to assess human mesenchymal stem/stromal cell (MSC) retention compared with a commonly used immune deficient line, the NSG mouse. We found that YACNSG were able to retain human MSCs longer than the immune competent YAC128 mice. We performed behavioral assessments starting at 4 months of age and continued testing monthly until 12 months on the accelerod and in the open field. At 12 months, brains were isolated and evaluated using immunohistochemistry for striatal volume. Results from these studies suggest that the novel immune deficient YACNSG strain of mice could provide a good model for human stem-cell based therapies and that the immune system appears to play an important role in the pathology of HD.


Assuntos
Modelos Animais de Doenças , Doença de Huntington , Transplante de Células-Tronco Mesenquimais , Doenças Neuroinflamatórias , Animais , Progressão da Doença , Humanos , Doença de Huntington/fisiopatologia , Doença de Huntington/terapia , Camundongos , Camundongos Transgênicos , Doenças Neuroinflamatórias/fisiopatologia , Doenças Neuroinflamatórias/terapia
11.
Front Mol Neurosci ; 14: 789913, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35153670

RESUMO

Zinc finger (ZF), transcription activator-like effectors (TALE), and CRISPR/Cas9 therapies to regulate gene expression are becoming viable strategies to treat genetic disorders, although effective in vivo delivery systems for these proteins remain a major translational hurdle. We describe the use of a mesenchymal stem/stromal cell (MSC)-based delivery system for the secretion of a ZF protein (ZF-MSC) in transgenic mouse models and young rhesus monkeys. Secreted ZF protein from mouse ZF-MSC was detectable within the hippocampus 1 week following intracranial or cisterna magna (CM) injection. Secreted ZF activated the imprinted paternal Ube3a in a transgenic reporter mouse and ameliorated motor deficits in a Ube3a deletion Angelman Syndrome (AS) mouse. Intrathecally administered autologous rhesus MSCs were well-tolerated for 3 weeks following administration and secreted ZF protein was detectable within the cerebrospinal fluid (CSF), midbrain, and spinal cord. This approach is less invasive when compared to direct intracranial injection which requires a surgical procedure.

12.
Nucleic Acids Res ; 48(5): 2372-2387, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-31925439

RESUMO

A significant number of X-linked genes escape from X chromosome inactivation and are associated with a distinct epigenetic signature. One epigenetic modification that strongly correlates with X-escape is reduced DNA methylation in promoter regions. Here, we created an artificial escape by editing DNA methylation on the promoter of CDKL5, a gene causative for an infantile epilepsy, from the silenced X-chromosomal allele in human neuronal-like cells. We identify that a fusion of the catalytic domain of TET1 to dCas9 targeted to the CDKL5 promoter using three guide RNAs causes significant reactivation of the inactive allele in combination with removal of methyl groups from CpG dinucleotides. Strikingly, we demonstrate that co-expression of TET1 and a VP64 transactivator have a synergistic effect on the reactivation of the inactive allele to levels >60% of the active allele. We further used a multi-omics assessment to determine potential off-targets on the transcriptome and methylome. We find that synergistic delivery of dCas9 effectors is highly selective for the target site. Our findings further elucidate a causal role for reduced DNA methylation associated with escape from X chromosome inactivation. Understanding the epigenetics associated with escape from X chromosome inactivation has potential for those suffering from X-linked disorders.


Assuntos
Cromossomos Humanos X/química , Epigênese Genética , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Inativação do Cromossomo X , Alelos , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Domínio Catalítico , Linhagem Celular Tumoral , Cromossomos Humanos X/metabolismo , Ilhas de CpG , Edição de Genes , Inativação Gênica , Humanos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transativadores/genética , Transativadores/metabolismo
13.
Front Genome Ed ; 2: 7, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34713216

RESUMO

Understanding of cell-type specific transcription factors has promoted progress in methods for cellular reprogramming, such as directly reprogramming somatic cells to induced neurons (iN). Methods for direct reprogramming require neuronal-fate determining gene activation via neuron-specific microRNAs, chemical modulation of key neuronal signaling pathways or overexpression via viral vectors, with some reprogramming strategies requiring a combination of these methods to induce the neuronal-cell fate. These methods have been employed in a multitude of cell types, including fibroblasts, hepatocytes, peripheral blood mononuclear, and T cells. The ability to create iN from skin biopsies and blood samples coupled with recent advancements in artificially inducing age- and disease-associated phenotypes are accelerating the development of disease models for late-onset neurodegenerative disorders. Here, we review how activation of the neuronal transcriptome alters the epigenetic landscape of the donor cell to facilitate reprogramming to neurons. We also discuss the advantages of using DNA binding domains such as CRISPR/dCas9 to overcome epigenetic barriers to induce neuronal-cell fate by activating endogenous neuronal cell-fate determining genes.

14.
Epigenetics Chromatin ; 12(1): 26, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-31053162

RESUMO

BACKGROUND: Rewriting of the epigenome has risen as a promising alternative to gene editing for precision medicine. In nature, epigenetic silencing can result in complete attenuation of target gene expression over multiple mitotic divisions. However, persistent repression has been difficult to achieve in a predictable manner using targeted systems. RESULTS: Here, we report that persistent epigenetic memory required both a DNA methyltransferase (DNMT3A-dCas9) and a histone methyltransferase (Ezh2-dCas9 or KRAB-dCas9). We demonstrate that the histone methyltransferase requirement can be locus specific. Co-targeting Ezh2-dCas9, but not KRAB-dCas9, with DNMT3A-dCas9 and DNMT3L induced long-term HER2 repression over at least 50 days (approximately 57 cell divisions) and triggered an epigenetic switch to a heterochromatic environment. An increase in H3K27 trimethylation and DNA methylation was stably maintained and accompanied by a sustained loss of H3K27 acetylation. Interestingly, substitution of Ezh2-dCas9 with KRAB-dCas9 enabled long-term repression at some target genes (e.g., SNURF) but not at HER2, at which H3K9me3 and DNA methylation were transiently acquired and subsequently lost. Off-target DNA hypermethylation occurred at many individual CpG sites but rarely at multiple CpGs in a single promoter, consistent with no detectable effect on transcription at the off-target loci tested. Conversely, robust hypermethylation was observed at HER2. We further demonstrated that Ezh2-dCas9 required full-length DNMT3L for maximal activity and that co-targeting DNMT3L was sufficient for persistent repression by Ezh2-dCas9 or KRAB-dCas9. CONCLUSIONS: These data demonstrate that targeting different combinations of histone and DNA methyltransferases is required to achieve maximal repression at different loci. Fine-tuning of targeting tools is a necessity to engineer epigenetic memory at any given locus in any given cell type.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteínas Repressoras/genética , Animais , Sistemas CRISPR-Cas , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Edição de Genes , Engenharia Genética/métodos , Células HCT116 , Histona Metiltransferases/genética , Histona Metiltransferases/metabolismo , Histonas/metabolismo , Humanos , Camundongos , Regiões Promotoras Genéticas , RNA Guia de Cinetoplastídeos/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo
15.
Biochem Biophys Res Commun ; 512(4): 729-735, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-30926165

RESUMO

Mesenchymal stem cell (MSC) based therapies are currently being evaluated as a putative therapeutic in numerous human clinical trials. Recent reports have established that exosomes mediate much of the therapeutic properties of MSCs. Exosomes are nanovesicles which mediate intercellular communication, transmitting signals between cells which regulate a diverse range of biological processes. MSC-derived exosomes are packaged with numerous types of proteins and RNAs, however, their metabolomic and lipidomic profiles to date have not been well characterized. We previously reported that MSCs, in response to priming culture conditions that mimic the in vivo microenvironmental niche, substantially modulate cellular signaling and significantly increase the secretion of exosomes. Here we report that MSCs exposed to such priming conditions undergo glycolytic reprogramming, which homogenizes MSCs' metabolomic profile. In addition, we establish that exosomes derive from primed MSCs are packaged with numerous metabolites that have been directly associated with immunomodulation, including M2 macrophage polarization and regulatory T lymphocyte induction.


Assuntos
Exossomos/imunologia , Células-Tronco Mesenquimais/imunologia , Linhagem Celular , Exossomos/metabolismo , Glicólise , Humanos , Imunomodulação , Ativação de Macrófagos , Células-Tronco Mesenquimais/metabolismo , Metaboloma , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
16.
Methods Mol Biol ; 1937: 47-58, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30706389

RESUMO

Transcription activator-like effectors (TALEs) are modular proteins derived from the plant Xanthomonas sp. pathogen that can be designed to target unique DNA sequences following a simple cipher. Customized TALE proteins can be used in a variety of molecular applications that include gene editing and transcriptional modulation. Presently, we provide a brief primer on the design and construction of TALEs. TALE proteins can be fused to a variety of different effector domains that alter the function of the TALE upon binding. This flexibility of TALE design and downstream effect may offer therapeutic applications that are discussed in this section. Finally, we provide a future perspective on TALE technology and what challenges remain for successful translation of gene-editing strategies to the clinic.


Assuntos
Efetores Semelhantes a Ativadores de Transcrição/genética , Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Xanthomonas/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Montagem e Desmontagem da Cromatina , DNA/metabolismo , Engenharia Genética , Humanos , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Efetores Semelhantes a Ativadores de Transcrição/química , Ativação Transcricional , Xanthomonas/genética
17.
Neurosci Biobehav Rev ; 98: 194-207, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30658070

RESUMO

To improve the translational predictability of treatment strategies for Huntington's disease (HD), sensitive and analogous cognitive outcomes are needed across HD animal models and humans. Spatial memory measures are promising candidates because they are based on 'visual' or 'non-verbal' cognition, and are commonly tested in both animals and humans. Here, we consider the suitability of spatial memory for strengthening translational links between animals and humans in HD research and clinical trials. We describe findings of spatial memory impairments in human HD and mouse models, including which aspects of spatial memory are most affected and at which time points in disease progression. We also describe the neural systems that underlie spatial memory and link spatial memory impairments to HD neuropathology, focussing on striatal and hippocampal systems. We provide a critical analysis of the literature in terms of the suitability of spatial memory for bridging the translational gap between species. Finally, we discuss possible neural mechanisms that might explain the spatial memory impairments seen in HD, and their relevance to potential treatments.


Assuntos
Transtornos Cognitivos/patologia , Hipocampo/patologia , Doença de Huntington/patologia , Neurogênese/fisiologia , Memória Espacial/fisiologia , Animais , Modelos Animais de Doenças , Humanos
18.
Stem Cells Dev ; 28(6): 398-409, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30638129

RESUMO

Mesenchymal stem cells (MSCs) facilitate functional recovery in numerous animal models of inflammatory and ischemic tissue-related diseases with a growing body of research suggesting that exosomes mediate many of these therapeutic effects. It remains unclear, however, which types of proteins are packaged into exosomes compared with the cells from which they are derived. In this study, using comprehensive proteomic analysis, we demonstrated that human primed MSCs secrete exosomes (pMEX) that are packaged with markedly higher fractions of specific protein subclasses compared with their cells of origin, indicating regulation of their contents. Notably, we found that pMEX are also packaged with substantially elevated levels of extracellular-associated proteins. Fibronectin was the most abundant protein detected, and data established that fibronectin mediates the mitogenic properties of pMEX. In addition, treatment of SHSY5Y cells with pMEX induced the secretion of growth factors known to possess mitogenic and neurotrophic properties. Taken together, our comprehensive analysis indicates that pMEX are packaged with specific protein subtypes, which may provide a molecular basis for their distinct functional properties.


Assuntos
Exossomos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mitose , Adolescente , Adulto , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade
19.
Orthopedics ; 41(4): e580-e582, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29309717

RESUMO

Bucket-handle meniscal tears are extremely rare injuries in the pediatric population. Given the known sequelae of meniscal tears and the development of osteoarthritis, early diagnosis and treatment of pediatric meniscal tears are essential. The communication barrier between physicians and pediatric patients, however, creates difficulties in making the correct diagnosis of meniscal pathology. Providers should have a low threshold for obtaining further confirmatory advanced imaging if patients present with prolonged knee pain and swelling, with associated locking mechanical symptoms. Good clinical outcomes following meniscal repair have been observed, likely a result of the increased residual vascularity found within pediatric menisci. The youngest reported case of an isolated traumatic bucket-handle medial meniscal tear is examined in this article. A 2-year-old girl presented with right knee pain after a heavy object fell onto the knee several weeks earlier. On examination, the patient walked with a limp but lacked full range of active knee motion. The McMurray test elicited pain without a mechanical or palpable click. After the patient continued to exhibit limitations following conservative management, magnetic resonance imaging of the right knee was obtained, which showed a bucket-handle tear of the right medial meniscus. The patient subsequently underwent meniscal repair and, on recovery, was able to regain normal functioning of the right knee without limitations. [Orthopedics. 2018; 41(4):e580-e582.].


Assuntos
Lesões do Menisco Tibial/diagnóstico por imagem , Lesões do Menisco Tibial/cirurgia , Artroscopia , Pré-Escolar , Tratamento Conservador , Feminino , Humanos , Imageamento por Ressonância Magnética , Dor/etiologia , Lesões do Menisco Tibial/complicações
20.
Spartan Med Res J ; 3(2): 7021, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-33655147

RESUMO

CONTEXT: The Affordable Care Act (ACA) was implemented to make insurance accessible and reduce healthcare costs. The purpose of this study was to examine for changes in the use of lower-acuity types of Emergency Department (ED) services at two suburban Detroit facilities before, and after implementation of the ACA. METHODS: A retrospective chart review of patients presenting to the ED at a two-campus suburban hospital system was performed over two 18-month pre- and post-ACA periods. The authors completed a review of electronic health record data and used ICD-9 code and ED visit billing and collections data. Sample patients included those who had received lower-acuity ED care within the two designated time periods. A total of 16 lower-acuity ICD-9 codes were included to gauge pre and post changes in use of ED services. RESULTS: The authors identified 2,099 patients meeting study criteria during the pre-ACA period and 2,158 patients within the post-ACA period. A subgroup of 166,483 ED patients received care during the pre-ACA time period and 179,879 post-ACA. There was no statistically significant difference between the volume of lower-acuity ED visits during the two analytic periods (i.e., 1.26% seen pre-ACA implementation and 1.20% seen post-ACA). (p = 0.420) Neither did the absolute number of all ED visits significantly change. As could be anticipated, however, the proportion of self-pay patients pre-ACA significantly decreased from 506 (24.1%) to 191 (8.9%) post-ACA. (p < 0.001) Medicaid HMO payments also increased significantly from 824 visits pre-ACA to 1,086 visits post-ACA. (p < 0.001) In addition, Blue Cross coverage increased from 54 visits pre-ACA to 98 visits post-ACA. (p < 0.001). CONCLUSIONS: In summary, our results revealed no significant change in the absolute volume of all ED visits or proportions of lower-acuity ED visits between the pre- and post-ACA periods. As the authors had anticipated, pre and post changes in the number of self-pay patients and those with certain types of insurance coverage were dramatic. The authors conclude that changes in lower-acuity visits to the ED in these study settings had not decreased as envisioned by ACA developers. Future studies with larger longitudinal samples are warranted to more fully investigate the longer-term implications of the ACA on use of ED services.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...