Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
SLAS Discov ; 29(2): 100137, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38128829

RESUMO

Aberrant protein aggregation is a pathological cellular hallmark of many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), where the tau protein is aggregating, forming neurofibrillary tangles (NFTs), and propagating from neuron to neuron. These processes have been linked to disease progression and a decline in cognitive function. Various therapeutic approaches aim at the prevention or reduction of tau aggregates in neurons. Human induced pluripotent stem cells (hiPSCs) are a very valuable tool in neuroscience discovery, as they offer access to potentially unlimited amounts of cell types that are affected in disease, including cortical neurons, for in vitro studies. We have generated an in vitro model for tau aggregation that uses hiPSC - derived neurons expressing an aggregation prone, fluorescently tagged version of the human tau protein after lentiviral transduction. Upon addition of tau seeds in the form of recombinant sonicated paired helical filaments (sPHFs), the neurons show robust, disease-like aggregation of the tau protein. The model was developed as a plate-based high content screening assay coupled with an image analysis algorithm to evaluate the impact of small molecules or genetic perturbations on tau. We show that the assay can be used to evaluate small molecules or screen targeted compound libraries. Using siRNA-based gene knockdown, genes of interest can be evaluated, and we could show that a targeted gene library can be screened, by screening nearly 100 deubiquitinating enzymes (DUBs) in that assay. The assay uses an imaging-based readout, a relatively short timeline, quantifies the extent of tau aggregation, and also allows the assessment of cell viability. Furthermore, it can be easily adapted to different hiPSC lines or neuronal subtypes. Taken together, this complex and highly relevant approach can be routinely applied on a weekly basis in the screening funnels of several projects and generates data with a turnaround time of approximately five weeks.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Proteínas tau/genética , Proteínas tau/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Doença de Alzheimer/metabolismo , Neurônios/metabolismo
2.
Eur J Pharmacol ; 934: 175301, 2022 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-36191631

RESUMO

In this study we aimed to reduce tau pathology, a hallmark of Alzheimer's Disease (AD), by activating mTOR-dependent autophagy in a transgenic mouse model of tauopathy by long-term dosing of animals with mTOR-inhibitors. Rapamycin treatment reduced the burden of hyperphosphorylated and aggregated pathological tau in the cerebral cortex only when applied to young mice, prior to the emergence of pathology. Conversely, PQR530 which exhibits better brain exposure and superior pharmacokinetic properties, reduced tau pathology even when the treatment started after the onset of pathology. Our results show that dosing animals twice per week with PQR530 resulted in intermittent, rather than sustained target engagement. Nevertheless, this pulse-like mTOR inhibition followed by longer intervals of re-activation was sufficient to reduce tau pathology in the cerebral cortex in P301S tau transgenic mice. This suggests that balanced therapeutic dosing of blood-brain-barrier permeable mTOR-inhibitors can result in a disease-modifying effect in AD and at the same time prevents toxic side effects due to prolonged over activation of autophagy.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Proteínas tau/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Camundongos Transgênicos , Encéfalo , Sirolimo/farmacologia , Modelos Animais de Doenças
3.
Cell Rep ; 38(9): 110435, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235798

RESUMO

Apolipoprotein E transports lipids and couples metabolism between astrocytes and neurons. The E4 variant (APOE4) affects these functions and represents a genetic predisposition for Alzheimer's disease, but the molecular mechanisms remain elusive. We show that ApoE produces different types of lipoproteins via distinct lipidation pathways. ApoE forms high-density lipoprotein (HDL)-like, cholesterol-rich particles via the ATP-binding cassette transporter 1 (ABCA1), a mechanism largely unaffected by ApoE polymorphism. Alternatively, ectopic accumulation of fat in astrocytes, a stress-associated condition, redirects ApoE toward the assembly and secretion of triacylglycerol-rich lipoproteins, a process boosted by the APOE4 variant. We demonstrate in vitro that ApoE can detect triacylglycerol in membranes and spontaneously assemble lipoprotein particles (10-20 nm) rich in unsaturated triacylglycerol, and that APOE4 has remarkable properties behaving as a strong triacylglycerol binder. We propose that fatty APOE4 astrocytes have reduced ability to clear toxic fatty acids from the extracellular milieu, because APOE4 reroutes them back to secretion.


Assuntos
Apolipoproteína E4 , Astrócitos , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Isoformas de Proteínas/metabolismo , Triglicerídeos/metabolismo
4.
Sci Rep ; 12(1): 79, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34996966

RESUMO

Autophagic flux can be quantified based on the accumulation of lipidated LC3B in the presence of late-stage autophagy inhibitors. This method has been widely applied to identify novel compounds that activate autophagy. Here we scrutinize this approach and show that bafilomycin A1 (BafA) but not chloroquine is suitable for flux quantification due to the stimulating effect of chloroquine on non-canonical LC3B-lipidation. Significant autophagic flux increase by rapamycin could only be observed when combining it with BafA concentrations not affecting basal flux, a condition which created a bottleneck, rather than fully blocking autophagosome-lysosome fusion, concomitant with autophagy stimulation. When rapamycin was combined with saturating concentrations of BafA, no significant further increase of LC3B lipidation could be detected over the levels induced by the late-stage inhibitor. The large assay window obtained by this approach enables an effective discrimination of autophagy activators based on their cellular potency. To demonstrate the validity of this approach, we show that a novel inhibitor of the acetyltransferase EP300 activates autophagy in a mTORC1-dependent manner. We propose that the creation of a sensitized background rather than a full block of autophagosome progression is required to quantitatively capture changes in autophagic flux.


Assuntos
Autofagossomos/metabolismo , Autofagia , Metabolismo dos Lipídeos , Proteínas Associadas aos Microtúbulos/metabolismo , Autofagossomos/efeitos dos fármacos , Autofagossomos/genética , Autofagia/efeitos dos fármacos , Bioensaio , Biomarcadores/metabolismo , Cloroquina/farmacologia , Relação Dose-Resposta a Droga , Proteína p300 Associada a E1A/metabolismo , Células HeLa , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Macrolídeos/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Associadas aos Microtúbulos/genética
5.
Methods Mol Biol ; 1994: 31-39, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31124102

RESUMO

To better understand and model neurological, in particular neurodegenerative diseases, human induced pluripotent stem cells (hiPSCs) offer a great source for generation of neural cells. We provide a protocol for the differentiation of hiPSc-derived astrocytes in vitro. This protocol not only is chemically defined, that is, it does not use serum, but also allows for the expansion of astrocyte progenitor cells and mature astrocytes. Large batches of hiPSc-derived astrocytes can be stored and used for defined in vitro disease models.


Assuntos
Astrócitos/citologia , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Modelos Biológicos , Células-Tronco Neurais/efeitos dos fármacos , Doenças Neurodegenerativas , Neurogênese/efeitos dos fármacos
6.
J Med Chem ; 62(8): 4120-4130, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-30933499

RESUMO

Apolipoprotein E is a 299-residue lipid carrier protein produced in both the liver and the brain. The protein has three major isoforms denoted apoE2, apoE3, and apoE4 which differ at positions 112 and 158 and which occur at different frequencies in the human population. Genome-wide association studies indicate that the possession of two apoE4 alleles is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). In an attempt to identify a small molecule stabilizer of apoE4 function that may have utility as a therapy for Alzheimer's disease, we carried out an NMR-based fragment screen on the N-terminal domain of apoE4 and identified a benzyl amidine based fragment binder. In addition to NMR, binding was characterized using various other biophysical techniques, and a crystal structure of the bound core was obtained. Core elaboration ultimately yielded a compound that showed activity in an IL-6 and IL-8 cytokine release assay.


Assuntos
Apolipoproteína E4/metabolismo , Bibliotecas de Moléculas Pequenas/química , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amidinas/química , Amidinas/metabolismo , Apolipoproteína E4/química , Apolipoproteína E4/genética , Sítios de Ligação , Cristalografia por Raios X , Descoberta de Drogas , Humanos , Lipossomos/química , Lipossomos/metabolismo , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Domínios Proteicos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/uso terapêutico , Relação Estrutura-Atividade , Temperatura de Transição
7.
Stem Cell Reports ; 11(4): 897-911, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30245212

RESUMO

Reproducibility in molecular and cellular studies is fundamental to scientific discovery. To establish the reproducibility of a well-defined long-term neuronal differentiation protocol, we repeated the cellular and molecular comparison of the same two iPSC lines across five distinct laboratories. Despite uncovering acceptable variability within individual laboratories, we detect poor cross-site reproducibility of the differential gene expression signature between these two lines. Factor analysis identifies the laboratory as the largest source of variation along with several variation-inflating confounders such as passaging effects and progenitor storage. Single-cell transcriptomics shows substantial cellular heterogeneity underlying inter-laboratory variability and being responsible for biases in differential gene expression inference. Factor analysis-based normalization of the combined dataset can remove the nuisance technical effects, enabling the execution of robust hypothesis-generating studies. Our study shows that multi-center collaborations can expose systematic biases and identify critical factors to be standardized when publishing novel protocols, contributing to increased cross-site reproducibility.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Proteômica/métodos , Linhagem Celular , Análise Fatorial , Regulação da Expressão Gênica , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Fenótipo , Reprodutibilidade dos Testes , Transcriptoma/genética
8.
Biochim Biophys Acta ; 1854(8): 979-86, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25882196

RESUMO

Evaluation of drug-target interaction kinetics is becoming increasingly important during the drug-discovery process to investigate selectivity of a drug and predict in vivo target occupancy. To date, it remains challenging to obtain kinetic information for interactions between G-protein-coupled receptors (GPCRs) and small-molecule ligands in a label-free manner. Often GPCRs need to be solubilized or even stabilized by mutations which can be difficult and is time consuming. In addition, it is often unclear if the native conformation of the receptors is sustained. In this study, surface plasmon resonance (SPR) and surface acoustic wave (SAW) technologies have been used to detect ligand binding to the GPCR chemokine (C-X-C motif) receptor 4 (CXCR4) expressed in lipoparticles. We first evaluated different strategies to immobilize CXCR4-expressing lipoparticles. The highest small-molecule binding signal in SPR and SAW was achieved with a matrix-free carboxymethylated sensor chip coated with wheat germ agglutinin for lipoparticle capturing. Next, the binding kinetics of the anti-CXCR4 antibody 12G5 raised against a conformational epitope (k(on)=1.83×10(6)M(-1)s(-1), k(off)=2.79×10(-4) s(-1)) and the small molecule AMD3100 (k(on)=5.46×10(5)M(-1)s(-1), k(off)=1.01×10(-2)s(-1)) were assessed by SAW. Our kinetic and affinity data are consistent with previously published radioligand-binding experiments using cells and label-free experiments with solubilized CXCR4. This is the first study demonstrating label-free kinetic characterization of small-molecule binding to a GPCR in the membrane environment. The presented method holds the potential to greatly facilitate label-free assay development for GPRCs that can be expressed at high levels in lipoparticles.


Assuntos
Membrana Celular/química , Compostos Heterocíclicos/química , Receptores CXCR4/química , Ressonância de Plasmônio de Superfície , Benzilaminas , Membrana Celular/genética , Membrana Celular/metabolismo , Ciclamos , Células HEK293 , Compostos Heterocíclicos/metabolismo , Humanos , Cinética , Ligação Proteica , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
9.
Neuropsychopharmacology ; 37(2): 364-77, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21956444

RESUMO

Administration of the DNA-alkylating agent methylazoxymethanol acetate (MAM) on embryonic day 17 (E17) produces behavioral and anatomical brain abnormalities, which model some aspects of schizophrenia. This has lead to the premise that MAM rats are a neurodevelopmental model for schizophrenia. However, the underlying molecular pathways affected in this model have not been elucidated. In this study, we investigated the molecular phenotype of adult MAM rats by focusing on the frontal cortex and hippocampal areas, as these are known to be affected in schizophrenia. Proteomic and metabonomic analyses showed that the MAM treatment on E17 resulted primarily in deficits in hippocampal glutamatergic neurotransmission, as seen in some schizophrenia patients. Most importantly, these results were consistent with our finding of functional deficits in glutamatergic neurotransmission, as identified using electrophysiological recordings. Thus, this study provides the first molecular evidence, combined with functional validation, that the MAM-E17 rat model reproduces hippocampal deficits relevant to the pathology of schizophrenia.


Assuntos
Hipocampo/metabolismo , Acetato de Metilazoximetanol/farmacologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Esquizofrenia/induzido quimicamente , Esquizofrenia/metabolismo , Transmissão Sináptica/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Humanos , Masculino , Metabolômica/métodos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Proteômica/métodos , Ratos , Esquizofrenia/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos
10.
Neuropharmacology ; 59(6): 367-74, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20493887

RESUMO

Cyclic nucleotide-specific phosphodiesterases (PDEs) play a critical role in signal transduction by regulating the level of adenosine 3',5'-cyclic monophosphate (cAMP) and guanosine 3',5'-cyclic monophosphate (cGMP) in cells. The gene expression pattern of a PDE provides important information regarding its role in physiological and pathological processes. In this study, we have established the mRNA expression profile all PDE isoenzymes (PDE1A/B/C, 2A, 3A/B, 4A/B/C/D, 5A, 6A/B/C, 7A/B, 8A/B, 9A, 10A, 11A) in a human cDNA collection consisting of 10 brain regions (parietal, frontal, temporal cortex, hippocampus, striatum, thalamus, hypothalamus, substantia nigra, nucleus accumbens, cerebellum), spinal cord, dorsal root ganglia and 12 peripheral tissues (skeletal muscle, heart, thyroid, adrenal gland, pancreas, bladder, kidney, liver, lung, small intestine, spleen, and stomach). Using quantitative real-time polymerase chain reaction and parallel analysis of a carefully selected group of reference genes, we have determined the relative expression of each PDE isoenzyme across the 24 selected tissues, and also compared the expression of selected PDEs to each other within a given tissue type. Several PDEs show strikingly selective expression (e.g. PDE10A and PDE1B mRNA levels in the caudate nucleus are 20-fold higher than in most other tissues; PDE1C and PDE3A are highly expressed in the heart and PDE8B is expressed very strongly in the thyroid gland). This comprehensive approach provides a coherent and quantitative view of the mRNA expression of the PDE gene family and enables an integration of data obtained with other non-quantitative methods.


Assuntos
Encéfalo/enzimologia , Diester Fosfórico Hidrolases/metabolismo , RNA Mensageiro/metabolismo , Glândulas Suprarrenais/enzimologia , Sistema Digestório/enzimologia , Expressão Gênica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Rim/enzimologia , Pulmão/enzimologia , Músculo Esquelético/enzimologia , Miocárdio/enzimologia , Especificidade de Órgãos , Diester Fosfórico Hidrolases/genética , RNA Mensageiro/genética , Glândula Tireoide/enzimologia , Bexiga Urinária/enzimologia
11.
Brain Res ; 1260: 65-75, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19401173

RESUMO

The effects of amyloid-beta peptide (Aß) on astrocyte responses to activation of mGlu5 receptors have been investigated using calcium imaging. Pre-incubation with Aß1-40 peptide for up to 72 h produced a time- and concentration-dependent 2-4 fold enhancement in the magnitude of the intracellular calcium mobilization response to the group I metabotropic glutamate receptor agonist (S)-3,5-dihydroxyphenylglycine (DHPG). In contrast, pre-treatment with Aß1-40 did not alter the calcium responses induced by other G protein coupled- or ion channel-receptors. Aß 1-40-enhanced DHPG responses were blocked by the mGlu5 antagonist MPEP but not by inhibitors of voltage dependent calcium channels or by the AMPA/KA receptor antagonist CNQX. Up-regulation of mGlu5 coupled responses was associated with significant increases in astrocyte mGlu5 receptor-mRNA and-protein expression after preincubation with Aß . The changes observed in vitro were consistent with results obtained from human Alzheimer's disease (AD) patients.Immunostaining for mGlu5 receptors was increased on astrocytes which were colocalized with Aß plaques in hippocampal tissue from AD patients compared to age-matched controls. These results suggest that modulation of mGlu5 receptors in astrocytes could be an important mechanism in determining the progression of pathology in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Humanos , Espaço Intracelular/metabolismo , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores de AMPA/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ácido Caínico/metabolismo , Fatores de Tempo , Regulação para Cima
12.
Glia ; 55(8): 844-58, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17405146

RESUMO

The molecular mechanisms used by oligodendrocyte precursor cells (OPCs), oligodendrocytes (OLs), and Schwann cells (SCs) to advance processes for motility in the developing nervous system and to ensheath axons at myelination are currently not well defined. Here we demonstrate that OPCs, OLs, and SCs express the major proteins involved in actin polymerization-driven protrusion; these key proteins including F-actin, the Arp2/3 complex, neural-Wiskott Aldrich Syndrome protein (N-WASP) and WAVE proteins, and the RhoGTPases Rac and Cdc42 are present at the leading edges of processes being extended by OPCs, OLs, and SCs. We reveal by real-time PCR that OLs and SCs have different dominant WAVE isoforms. Inhibition of the WASP/WAVE protein, N-WASP, with wiskostatin that prevents activation of the Arp2/3 complex, blocks process extension by OPCs and SCs. Inhibition of N-WASP also causes OPC and SC process retraction, which is preceded by retraction of filopodia. This implicates filopodia in OPC and SC process stability and also of N-WASP in OPC and SC process dynamics. We also demonstrate that p34 (a component of the Arp2/3 complex), WASP/WAVE proteins, actin, alpha-tubulin, Rac, Cdc42, vinculin, and focal adhesion kinase are detected in water-shocked myelin purified from brain. Inhibition of N-WASP with wiskostatin decreases the number of axons undergoing initial ensheathment in intact optic nerve samples and reduces the Po content of dorsal root ganglia:SC co-cultures. Our findings indicate that OPCs, OLs, and SCs extend processes using actin polymerization-driven protrusion dependent on N-WASP. We hypothesize that inner mesaxons of OLs and SCs use the same mechanism to ensheath axons at myelination.


Assuntos
Fibras Nervosas Mielinizadas/fisiologia , Oligodendroglia/fisiologia , Pseudópodes/metabolismo , Células de Schwann/fisiologia , Células-Tronco/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Animais , Carbazóis/farmacologia , Linhagem Celular , Técnicas de Cocultura , Cortactina/metabolismo , Gânglios Espinais/citologia , Expressão Gênica , Microscopia Eletrônica de Varredura , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/fisiologia , Oligodendroglia/ultraestrutura , Nervo Óptico/citologia , Propanolaminas/farmacologia , Pseudópodes/efeitos dos fármacos , Ratos , Células de Schwann/ultraestrutura , Células-Tronco/ultraestrutura , Proteína Neuronal da Síndrome de Wiskott-Aldrich/antagonistas & inibidores , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética
14.
Methods Mol Biol ; 329: 353-69, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16846004

RESUMO

The process of bringing a new pharmacologically active drug to market is laborious, time consuming, and costly. From drug discovery to safety assessment, new methods are constantly sought to develop faster and more efficient procedures to eliminate drugs from further investigation because of their limited effectiveness or high toxicity. Because in vitro cell assays are an important arm of this discovery process, it is therefore somewhat unsurprising that there is an emerging contribution of embryonic stem (ES) cell technology to this area. This technology utilizes the in vitro differentiation of ES cells into somatic cell target populations that, when coupled to the use of "lineage selection" protocols, allows for the production of infinite numbers of pure populations of the desired cells for both bioactivity and toxicological screens. Unlike the use of transformed cell lines, ES-derived cells remain karyotypically normal and therefore better reflect the potential responses of cells in vivo, and when selected are more homogeneous than those obtained using primary cultures. In this chapter we discuss the use of ES cell-derived somatic cells in pharmacological screens, with particular emphasis on neural cells, and describe the methods and protocols associated with the development of ES cell-derived neural cell assays.


Assuntos
Embrião de Mamíferos/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Meios de Cultura , Proteínas de Ligação a DNA/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Marcação de Genes , Genes Reporter , Vetores Genéticos , Proteínas de Grupo de Alta Mobilidade/genética , Camundongos , Plasmídeos/genética , Fatores de Transcrição SOXB1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA