Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther Oncolytics ; 24: 417-428, 2022 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-35141398

RESUMO

Adoptive T cell therapy (ACT) with expanded tumor-infiltrating lymphocytes (TIL) can induce durable responses in cancer patients from multiple histologies, with response rates of up to 50%. Antibodies blocking the engagement of the inhibitory receptor programmed cell death protein 1 (PD-1) have been successful across a variety of cancer diagnoses. We hypothesized that these approaches could be combined by using CRISPR-Cas9 gene editing to knock out PD-1 in TILs from metastatic melanoma and head-and-neck, thyroid, and colorectal cancer. Non-viral, non-plasmid-based PD-1 knockout was carried out immediately prior to the traditional 14-day TIL-based ACT rapid-expansion protocol. A median 87.53% reduction in cell surface PD-1 expression was observed post-expansion and confirmed at the genomic level. No off-target editing was detected, and PD-1 knockout had no effect on final fold expansion. Edited cells exhibited few phenotypic differences and matched control functionality. Pre-clinical-scale results were confirmed at a clinical scale by generating a PD-1-deficient TIL product using the good manufacturing practice facilities, equipment, procedures, and starting material used for standard patient treatment. Our results demonstrate that simple, non-viral, non-plasmid-based CRISPR-Cas9 methods can be feasibly adopted into a TIL-based ACT protocol to produce treatment products deficient in molecules such as PD-1, without any evident negative effects.

2.
FEBS Open Bio ; 11(6): 1638-1644, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33838073

RESUMO

Dysregulation of interleukin-33 (IL-33) has been implicated in the pathogenesis of several autoimmune and inflammatory diseases, but few studies have examined transcriptional regulation of the IL33 gene. In the intestines, gene regulation is controlled by a transcription factor network of which the intestinal-specific transcription factor CDX2 is a key component. In this study, we investigated whether CDX2 regulates IL33 mRNA expression. We examined IL33 mRNA expression in primary colonic epithelial cells from healthy humans and epithelial cell lines, revealing high expression levels in primary colonic and LS174T cells. Combining genomics data (ChIP-seq, RNA-seq) and IL33 promoter analyses in LS174T cells revealed intronic enhancer activity in the IL33 gene that is dependent on CDX2 expression. Western blotting and qRT-PCR confirmed that IL33 expression is upregulated in a CDX2 concentration-dependent manner, thereby providing the first evidence that CDX2 regulates the expression of IL33.


Assuntos
Fator de Transcrição CDX2/metabolismo , Células Epiteliais/metabolismo , Interleucina-33/genética , Intestinos/metabolismo , Fator de Transcrição CDX2/genética , Humanos , Interleucina-33/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
3.
BMC Biotechnol ; 19(1): 36, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31208390

RESUMO

BACKGROUND: CRISPR/Cas9 is widely used for precise genetic editing in various organisms. CRISPR/Cas9 editing may in many plants be hampered by the presence of complex and high ploidy genomes and inefficient or poorly controlled delivery of the CRISPR/Cas9 components to gamete cells or cells with regenerative potential. Optimized strategies and methods to overcome these challenges are therefore in demand. RESULTS: In this study we investigated the feasibility of improving CRISPR/Cas9 editing efficiency by Fluorescence Activated Cell Sorting (FACS) of protoplasts. We used Agrobacterium infiltration in leaves of Nicotiana benthamiana for delivery of viral replicons for high level expression of gRNAs designed to target two loci in the genome, NbPDS and NbRRA, together with the Cas9 nuclease in fusion with the 2A self-splicing sequence and GFP (Cas9-2A-GFP). Protoplasts isolated from the infiltrated leaves were then subjected to FACS for selection of GFP enriched protoplast populations. This procedure resulted in a 3-5 fold (from 20 to 30% in unsorted to more than 80% in sorted) increase in mutation frequencies as evidenced by restriction enzyme analysis and the Indel Detection by Amplicon Analysis, which allows for high throughput profiling and quantification of the generated mutations. CONCLUSIONS: FACS of protoplasts expressing GFP tagged CRISPR/Cas9, delivered through A. tumefaciens leaf infiltration, facilitated clear CRISPR/Cas9 mediated mutation enrichment in selected protoplast populations.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Proteínas de Fluorescência Verde/metabolismo , Nicotiana/metabolismo , Folhas de Planta/metabolismo , Protoplastos/metabolismo , Citometria de Fluxo , Fluorescência , Proteínas de Fluorescência Verde/genética , Microscopia de Fluorescência , Mutação , Folhas de Planta/citologia , Folhas de Planta/genética , Plantas Geneticamente Modificadas , Protoplastos/citologia , Nicotiana/citologia , Nicotiana/genética
4.
Nat Commun ; 10(1): 1785, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31040271

RESUMO

Lysosomal replacement enzymes are essential therapeutic options for rare congenital lysosomal enzyme deficiencies, but enzymes in clinical use are only partially effective due to short circulatory half-life and inefficient biodistribution. Replacement enzymes are primarily taken up by cell surface glycan receptors, and glycan structures influence uptake, biodistribution, and circulation time. It has not been possible to design and systematically study effects of different glycan features. Here we present a comprehensive gene engineering screen in Chinese hamster ovary cells that enables production of lysosomal enzymes with N-glycans custom designed to affect key glycan features guiding cellular uptake and circulation. We demonstrate distinct circulation time and organ distribution of selected glycoforms of α-galactosidase A in a Fabry disease mouse model, and find that an α2-3 sialylated glycoform designed to eliminate uptake by the mannose 6-phosphate and mannose receptors exhibits improved circulation time and targeting to hard-to-reach organs such as heart. The developed design matrix and engineered CHO cell lines enables systematic studies towards improving enzyme replacement therapeutics.


Assuntos
Lisossomos/enzimologia , Animais , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Doença de Fabry/tratamento farmacológico , Doença de Fabry/enzimologia , Doença de Fabry/metabolismo , Glicosilação , Masculino , Camundongos , Camundongos Knockout , Proteínas Recombinantes/uso terapêutico , alfa-Galactosidase/uso terapêutico
5.
Int J Mol Sci ; 19(10)2018 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-30262754

RESUMO

We question whether the expression of GalNAc-T3, the only known O-GalNAc-transferase present in germ cells, is correlated with qualitative and functional parameters of spermatozoa. We investigated the expression of GalNAc-T3 in ejaculated spermatozoa with immunocytochemistry in swim-up purified and acrosome-reacted spermatozoa from quality-control semen donors and in semen samples from 206 randomly selected men representing a broad spectrum of semen quality. Using donor ejaculates and immunofluorescence detection we found that expression of GalNAc-T3 and the presence of the immature O-glycans Tn and T localized to the equatorial segment of spermatozoa. The proportion of GalNAc-T3-positive spermatozoa in the ejaculate increased after swim-up and appeared unaffected by induction of acrosomal exocytosis. The fraction of spermatozoa with equatorial expression of GalNAc-T3 correlated with classical semen parameters (concentration p = 9 × 10-6, morphology p = 7 × 10-8, and motility p = 1.8 × 10-5) and was significantly lower in men with oligoteratoasthenozoospermia (p = 0.0048). In conclusion, GalNAc-T3 was highly expressed by motile spermatozoa and the expression correlated positively with the classical semen parameters. Therefore, GalNAc-T3 expression seems related to the quality of the spermatozoa, and we propose that reduced expression of GalNAc-T3 may lead to impaired O-glycosylation of proteins and thereby abnormal maturation and reduced functionality of the spermatozoa.


Assuntos
Astenozoospermia/metabolismo , N-Acetilgalactosaminiltransferases/metabolismo , Motilidade dos Espermatozoides , Espermatozoides/metabolismo , Adulto , Astenozoospermia/genética , Humanos , Masculino , N-Acetilgalactosaminiltransferases/genética , Espermatozoides/citologia , Espermatozoides/fisiologia , Polipeptídeo N-Acetilgalactosaminiltransferase
6.
Sci Rep ; 8(1): 11813, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087389

RESUMO

The type II membrane-anchored serine protease, matriptase, encoded by suppression of tumorgenicity-14 (ST14) regulates the integrity of the intestinal epithelial barrier in concert with its inhibitor, HAI-1 encoded by serine peptidase inhibitor, Kunitz type -1 (SPINT1). The balance of the protease/inhibitor gene expression ratio is vital in preventing the oncogenic potential of matriptase. The intestinal cell lineage is regulated by a transcriptional regulatory network where the tumor suppressor, Caudal homeobox 2 (CDX2) is considered to be an intestinal master transcription factor. In this study, we show that CDX2 has a dual function in regulating both ST14 and SPINT1, gene expression in intestinal cells. We find that CDX2 is not required for the basal ST14 and SPINT1 gene expression; however changes in CDX2 expression affects the ST14/SPINT1 mRNA ratio. Exploring CDX2 ChIP-seq data from intestinal cell lines, we identified genomic CDX2-enriched enhancer elements for both ST14 and SPINT1, which regulate their corresponding gene promoter activity. We show that CDX2 displays both repressive and enhancing regulatory abilities in a cell specific manner. Together, these data reveal new insight into transcriptional mechanisms controlling the intestinal matriptase/inhibitor balance.


Assuntos
Fator de Transcrição CDX2/metabolismo , Proteínas de Ciclo Celular/biossíntese , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Fosfoproteínas/biossíntese , Serina Endopeptidases/biossíntese , Fator de Transcrição CDX2/genética , Células CACO-2 , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Mucosa Intestinal/citologia , Proteínas Associadas aos Microtúbulos/genética , Fosfoproteínas/genética , Serina Endopeptidases/genética
7.
PLoS One ; 13(7): e0200215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29975781

RESUMO

Sequencing of primary colorectal tumors has identified a gene fusion in approximately 3% of colorectal cancer patients of the VTI1A and TCF7L2 genes, encoding a VTI1A-TCF4 fusion protein containing a truncated TCF4. As dysregulation of the Wnt signaling pathway is associated with colorectal cancer development and progression, the functional properties and transcriptional regulation of the VTI1A-TCF4 fusion protein may also play a role in these processes. Functional characteristics of the VTI1A-TCF4 fusion protein in Wnt signaling were analyzed in NCI-H508 and LS174T colon cancer cell lines. The NCI-H508 cell line, containing the VTI1A-TCF7L2 fusion gene, showed no active Wnt signaling, and overexpression of the VTI1A-TCF4 fusion protein in LS174T cells along with a Wnt signaling luciferase reporter plasmid showed inhibition of activity. The transcriptional regulation of the VTI1A-TCF4 fusion gene was investigated in LS174T cells where the activity of the VTI1A promoter was compared to that of the TCF7L2 promoter, and the transcription factor CDX2 was analyzed for gene regulatory activity of the VTI1A promoter through luciferase reporter gene assay using colon cancer cell lines as a model. Transfection of LS174T cells showed that the VTI1A promoter is highly active compared to the TCF7L2 promoter, and that CDX2 activates transcription of VTI1A. These results suggest that the VTI1A-TCF4 fusion protein is a dominant negative regulator of the Wnt signaling pathway, and that transcription of VTI1A is activated by CDX2.


Assuntos
Fator de Transcrição CDX2/genética , Neoplasias do Colo/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Qb-SNARE/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Via de Sinalização Wnt , Sítios de Ligação , Fator de Transcrição CDX2/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Humanos , Intestinos/patologia , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas , Proteínas Qb-SNARE/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , beta Catenina/metabolismo
8.
Glycobiology ; 26(4): 360-76, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26610890

RESUMO

A large family of UDP-GalNAc:polypeptide GalNAc transferases (ppGalNAc-Ts) initiates and defines sites of mucin-type Ser/Thr-O-GalNAc glycosylation. Family members have been classified into peptide- and glycopeptide-preferring subfamilies, although both families possess variable activities against glycopeptide substrates. All but one isoform contains a C-terminal carbohydrate-binding lectin domain whose roles in modulating glycopeptide specificity is just being understood. We have previously shown for several peptide-preferring isoforms that the presence of a remote Thr-O-GalNAc, 6-17 residues from a Ser/Thr acceptor site, may enhance overall catalytic activity in an N- or C-terminal direction. This enhancement varies with isoform and is attributed to Thr-O-GalNAc interactions at the lectin domain. We now report on the glycopeptide substrate utilization of a series of glycopeptide (human-ppGalNAc-T4, T7, T10, T12 and fly PGANT7) and peptide-preferring transferases (T2, T3 and T5) by exploiting a series of random glycopeptide substrates designed to probe the functions of their catalytic and lectin domains. Glycosylation was observed at the -3, -1 and +1 residues relative to a neighboring Thr-O-GalNAc, depending on isoform, which we attribute to specific Thr-O-GalNAc binding at the catalytic domain. Additionally, these glycopeptide-preferring isoforms show remote lectin domain-assisted Thr-O-GalNAc enhancements that vary from modest to none. We conclude that the glycopeptide specificity of the glycopeptide-preferring isoforms predominantly resides in their catalytic domain but may be further modulated by remote lectin domain interactions. These studies further demonstrate that both domains of the ppGalNAc-Ts have specialized and unique functions that work in concert to control and order mucin-type O-glycosylation.


Assuntos
Glicopeptídeos/química , Lectinas/química , Mucinas/química , Sialiltransferases/química , Sequência de Aminoácidos/genética , Sítios de Ligação , Carboidratos/química , Carboidratos/genética , Domínio Catalítico , Fucose/análogos & derivados , Fucose/química , Glicopeptídeos/biossíntese , Glicopeptídeos/genética , Glicosilação , Humanos , Lectinas/genética , Mucinas/biossíntese , Mucinas/genética , Filogenia , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Sialiltransferases/genética , Especificidade por Substrato
9.
Glycobiology ; 25(1): 55-65, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25155433

RESUMO

N-acetylgalactosaminyltransferase (GalNAc)-type (mucin-type) O-glycosylation is an abundant and highly diverse modification of proteins. This type of O-glycosylation is initiated in the Golgi by a large family of up to 20 homologous polypeptide GalNAc-T isoenzymes that transfer GalNAc to Ser, Thr and possibly Tyr residues. These GalNAc residues are then further elongated by a large set of glycosyltransferases to build a variety of complex O-glycan structures. What determines O-glycan site occupancy is still poorly understood, although it is clear that the substrate specificities of individual isoenzymes and the repertoire of GalNAc-Ts in cells are key parameters. The GalNAc-T isoenzymes are differentially expressed in cells and tissues in principle allowing cells to produce unique O-glycoproteomes dependent on the specific subset of isoforms present. In vitro analysis of acceptor peptide substrate specificities using recombinant expressed GalNAc-Ts has been the method of choice for probing activities of individual isoforms, but these studies have been hampered by biological validation of actual O-glycosylation sites in proteins and number of substrate testable. Here, we present a systematic analysis of the activity of 10 human GalNAc-T isoenzymes with 195 peptide substrates covering known O-glycosylation sites and provide a comprehensive dataset for evaluating isoform-specific contributions to the O-glycoproteome.


Assuntos
N-Acetilgalactosaminiltransferases/química , Peptídeos/química , Polissacarídeos/química , Sequência de Carboidratos , Ensaios Enzimáticos , Regulação da Expressão Gênica , Glicômica , Glicosilação , Complexo de Golgi/química , Complexo de Golgi/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Dados de Sequência Molecular , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Peptídeos/síntese química , Polissacarídeos/metabolismo , Proteômica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Polipeptídeo N-Acetilgalactosaminiltransferase
10.
J Biol Chem ; 289(25): 17312-24, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24798328

RESUMO

The low density lipoprotein receptor (LDLR) is crucial for cholesterol homeostasis and deficiency in LDLR functions cause hypercholesterolemia. LDLR is a type I transmembrane protein that requires O-glycosylation for stable expression at the cell surface. It has previously been suggested that LDLR O-glycosylation is found N-terminal to the juxtamembrane region. Recently we identified O-glycosylation sites in the linker regions between the characteristic LDLR class A repeats in several LDLR-related receptors using the "SimpleCell" O-glycoproteome shotgun strategy. Herein, we have systematically characterized O-glycosylation sites on recombinant LDLR shed from HEK293 SimpleCells and CHO wild-type cells. We find that the short linker regions between LDLR class A repeats contain an evolutionarily conserved O-glycosylation site at position -1 of the first cysteine residue of most repeats, which in wild-type CHO cells is glycosylated with the typical sialylated core 1 structure. The glycosites in linker regions of LDLR class A repeats are conserved in LDLR from man to Xenopus and found in other homologous receptors. O-Glycosylation is controlled by a large family of polypeptide GalNAc transferases. Probing into which isoform(s) contributed to glycosylation of the linker regions of the LDLR class A repeats by in vitro enzyme assays suggested a major role of GalNAc-T11. This was supported by expression of LDLR in HEK293 cells, where knock-out of the GalNAc-T11 isoform resulted in the loss of glycosylation of three of four linker regions.


Assuntos
Receptores de LDL/metabolismo , Motivos de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Glicosilação , Células HEK293 , Humanos , Oócitos , Estrutura Terciária de Proteína , Receptores de LDL/genética , Sequências Repetitivas de Aminoácidos , Sialiltransferases/genética , Sialiltransferases/metabolismo , Xenopus laevis
11.
EMBO J ; 32(10): 1478-88, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23584533

RESUMO

Glycosylation is the most abundant and diverse posttranslational modification of proteins. While several types of glycosylation can be predicted by the protein sequence context, and substantial knowledge of these glycoproteomes is available, our knowledge of the GalNAc-type O-glycosylation is highly limited. This type of glycosylation is unique in being regulated by 20 polypeptide GalNAc-transferases attaching the initiating GalNAc monosaccharides to Ser and Thr (and likely some Tyr) residues. We have developed a genetic engineering approach using human cell lines to simplify O-glycosylation (SimpleCells) that enables proteome-wide discovery of O-glycan sites using 'bottom-up' ETD-based mass spectrometric analysis. We implemented this on 12 human cell lines from different organs, and present a first map of the human O-glycoproteome with almost 3000 glycosites in over 600 O-glycoproteins as well as an improved NetOGlyc4.0 model for prediction of O-glycosylation. The finding of unique subsets of O-glycoproteins in each cell line provides evidence that the O-glycoproteome is differentially regulated and dynamic. The greatly expanded view of the O-glycoproteome should facilitate the exploration of how site-specific O-glycosylation regulates protein function.


Assuntos
Glicoproteínas/análise , N-Acetilgalactosaminiltransferases/metabolismo , Proteômica/métodos , Algoritmos , Motivos de Aminoácidos , Linhagem Celular Tumoral , Engenharia Genética/métodos , Glicoproteínas/metabolismo , Glicosilação , Humanos , N-Acetilgalactosaminiltransferases/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
12.
J Biol Chem ; 285(47): 36293-303, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20837471

RESUMO

The angiopoietin-like protein 3 (ANGPTL3) is an important inhibitor of the endothelial and lipoprotein lipases and a promising drug target. ANGPTL3 undergoes proprotein convertase processing (RAPR(224)↓TT) for activation, and the processing site contains two potential GalNAc O-glycosylation sites immediately C-terminal (TT(226)). We developed an in vivo model system in CHO ldlD cells that was used to show that O-glycosylation in the processing site blocked processing of ANGPTL3. Genome-wide SNP association studies have identified the polypeptide GalNAc-transferase gene, GALNT2, as a candidate gene for low HDL and high triglyceride blood levels. We hypothesized that the GalNAc-T2 transferase performed critical O-glycosylation of proteins involved in lipid metabolism. Screening of a panel of proteins known to affect lipid metabolism for potential sites glycosylated by GalNAc-T2 led to identification of Thr(226) adjacent to the proprotein convertase processing site in ANGPTL3. We demonstrated that GalNAc-T2 glycosylation of Thr(226) in a peptide with the RAPR(224)↓TT processing site blocks in vitro furin cleavage. The study demonstrates that ANGPTL3 activation is modulated by O-glycosylation and that this step is probably controlled by GalNAc-T2.


Assuntos
Angiopoietinas/metabolismo , Glicopeptídeos/metabolismo , Lipídeos/sangue , N-Acetilgalactosaminiltransferases/metabolismo , Pró-Proteína Convertases/metabolismo , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Animais , Western Blotting , Células CHO , Cricetinae , Cricetulus , Furina/metabolismo , Glicosilação , Humanos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Polipeptídeo N-Acetilgalactosaminiltransferase
13.
J Biol Chem ; 281(27): 18370-7, 2006 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-16638743

RESUMO

Mutations in the gene encoding the glycosyltransferase polypeptide GalNAc-T3, which is involved in initiation of O-glycosylation, were recently identified as a cause of the rare autosomal recessive metabolic disorder familial tumoral calcinosis (OMIM 211900). Familial tumoral calcinosis is associated with hyperphosphatemia and massive ectopic calcifications. Here, we demonstrate that the secretion of the phosphaturic factor fibroblast growth factor 23 (FGF23) requires O-glycosylation, and that GalNAc-T3 selectively directs O-glycosylation in a subtilisin-like proprotein convertase recognition sequence motif, which blocks processing of FGF23. The study suggests a novel posttranslational regulatory model of FGF23 involving competing O-glycosylation and protease processing to produce intact FGF23.


Assuntos
Calcinose/genética , Fatores de Crescimento de Fibroblastos/metabolismo , N-Acetilgalactosaminiltransferases/metabolismo , Proteínas de Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Motivos de Aminoácidos , Animais , Ligação Competitiva , Células CHO , Calcinose/metabolismo , Cricetinae , Fator de Crescimento de Fibroblastos 23 , Glicosilação , Humanos , Doenças Metabólicas/genética , Dados de Sequência Molecular , N-Acetilgalactosaminiltransferases/genética , Proteínas de Neoplasias/genética , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Especificidade por Substrato , Polipeptídeo N-Acetilgalactosaminiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA