Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 134(2): 143-161, 2024 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-38156445

RESUMO

BACKGROUND: Single-nucleotide polymorphisms linked with the rs1474868 T allele (MFN2 [mitofusin-2] T/T) in the human mitochondrial fusion protein MFN2 gene are associated with reduced platelet MFN2 RNA expression and platelet counts. This study investigates the impact of MFN2 on megakaryocyte and platelet biology. METHODS: Mice with megakaryocyte/platelet deletion of Mfn2 (Mfn2-/- [Mfn2 conditional knockout]) were generated using Pf4-Cre crossed with floxed Mfn2 mice. Human megakaryocytes were generated from cord blood and platelets isolated from healthy subjects genotyped for rs1474868. Ex vivo approaches assessed mitochondrial morphology, function, and platelet activation responses. In vivo measurements included endogenous/transfused platelet life span, tail bleed time, transient middle cerebral artery occlusion, and pulmonary vascular permeability/hemorrhage following lipopolysaccharide-induced acute lung injury. RESULTS: Mitochondria was more fragmented in megakaryocytes derived from Mfn2-/- mice and from human cord blood with MFN2 T/T genotype compared with control megakaryocytes. Human resting platelets of MFN2 T/T genotype had reduced MFN2 protein, diminished mitochondrial membrane potential, and an increased rate of phosphatidylserine exposure during ex vivo culture. Platelet counts and platelet life span were reduced in Mfn2-/- mice accompanied by an increased rate of phosphatidylserine exposure in resting platelets, especially aged platelets, during ex vivo culture. Mfn2-/- also decreased platelet mitochondrial membrane potential (basal) and activated mitochondrial oxygen consumption rate, reactive oxygen species generation, calcium flux, platelet-neutrophil aggregate formation, and phosphatidylserine exposure following dual agonist activation. Ultimately, Mfn2-/- mice showed prolonged tail bleed times, decreased ischemic stroke infarct size after cerebral ischemia-reperfusion, and exacerbated pulmonary inflammatory hemorrhage following lipopolysaccharide-induced acute lung injury. Analysis of MFN2 SNPs in the iSPAAR study (Identification of SNPs Predisposing to Altered ALI Risk) identified a significant association between MFN2 and 28-day mortality in patients with acute respiratory distress syndrome. CONCLUSIONS: Mfn2 preserves mitochondrial phenotypes in megakaryocytes and platelets and influences platelet life span, function, and outcomes of stroke and lung injury.


Assuntos
Lesão Pulmonar Aguda , Lipopolissacarídeos , Idoso , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/metabolismo , Plaquetas/metabolismo , Hemorragia/metabolismo , Mitocôndrias/metabolismo , Fosfatidilserinas/metabolismo
3.
Blood Cells Mol Dis ; 92: 102624, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34775219

RESUMO

The purpose of this research was to assess the effects of a microRNA (miRNA) cluster on platelet production. Human chromosome 19q13.41 harbors an evolutionarily conserved cluster of three miRNA genes (MIR99B, MIRLET7E, MIR125A) within 727 base-pairs. We now report that levels of miR-99b-5p, miR-let7e-5p and miR-125a-5p are strongly correlated in human platelets, and all are positively associated with platelet count, but not white blood count or hemoglobin level. Although the cluster regulates hematopoietic stem cell proliferation, the function of this genomic locus in megakaryocyte (MK) differentiation and platelet production is unknown. Furthermore, studies of individual miRNAs do not represent broader effects in the context of a cluster. To address this possibility, MK/platelet lineage-specific Mir-99b/let7e/125a knockout mice were generated. Compared to wild type littermates, cluster knockout mice had significantly lower platelet counts and reduced MK proplatelet formation, but no differences in MK numbers, ploidy, maturation or ultra-structural morphology, and no differences in platelet function. Compared to wild type littermates, knockout mice showed similar survival after pulmonary embolism. The major conclusions are that the effect of the Mir-99b/let7e/125a cluster is confined to a late stage of thrombopoiesis, and this effect on platelet number is uncoupled from platelet function.


Assuntos
Plaquetas/metabolismo , Megacariócitos/metabolismo , MicroRNAs/genética , Animais , Plaquetas/citologia , Deleção de Genes , Humanos , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Família Multigênica , Contagem de Plaquetas , Testes de Função Plaquetária , Trombocitopenia/genética , Trombopoese
4.
Blood Adv ; 5(9): 2362-2374, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33944898

RESUMO

Human anucleate platelets cannot be directly modified using traditional genetic approaches. Instead, studies of platelet gene function depend on alternative models. Megakaryocytes (the nucleated precursor to platelets) are the nearest cell to platelets in origin, structure, and function. However, achieving consistent genetic modifications in primary megakaryocytes has been challenging, and the functional effects of induced gene deletions on human megakaryocytes for even well-characterized platelet genes (eg, ITGA2B) are unknown. Here we present a rapid and systematic approach to screen genes for platelet functions in CD34+ cell-derived megakaryocytes called CRIMSON (CRISPR-edited megakaryocytes for rapid screening of platelet gene functions). By using CRISPR/Cas9, we achieved efficient nonviral gene editing of a panel of platelet genes in megakaryocytes without compromising megakaryopoiesis. Gene editing induced loss of protein in up to 95% of cells for platelet function genes GP6, RASGRP2, and ITGA2B; for the immune receptor component B2M; and for COMMD7, which was previously associated with cardiovascular disease and platelet function. Gene deletions affected several select responses to platelet agonists in megakaryocytes in a manner largely consistent with those expected for platelets. Deletion of B2M did not significantly affect platelet-like responses, whereas deletion of ITGA2B abolished agonist-induced integrin activation and spreading on fibrinogen without affecting the translocation of P-selectin. Deletion of GP6 abrogated responses to collagen receptor agonists but not thrombin. Deletion of RASGRP2 impaired functional responses to adenosine 5'-diphosphate (ADP), thrombin, and collagen receptor agonists. Deletion of COMMD7 significantly impaired multiple responses to platelet agonists. Together, our data recommend CRIMSON for rapid evaluation of platelet gene phenotype associations.


Assuntos
Plaquetas , Megacariócitos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Fatores de Troca do Nucleotídeo Guanina , Humanos , Fenótipo , Trombopoese
5.
J Thromb Haemost ; 19(5): 1319-1330, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33587773

RESUMO

BACKGROUND: Heparanase (HPSE) is the only known mammalian enzyme that can degrade heparan sulfate. Heparan sulfate proteoglycans are essential components of the glycocalyx, and maintain physiological barriers between the blood and endothelial cells. HPSE increases during sepsis, which contributes to injurious glyocalyx degradation, loss of endothelial barrier function, and mortality. OBJECTIVES: As platelets are one of the most abundant cellular sources of HPSE, we sought to determine whether HPSE expression and activity increases in human platelets during clinical sepsis. We also examined associations between platelet HPSE expression and clinical outcomes. PATIENTS/METHODS: Expression and activity of HPSE was determined in platelets isolated from septic patients (n = 59) and, for comparison, sex-matched healthy donors (n = 46) using complementary transcriptomic, proteomic, and functional enzymatic assays. Septic patients were followed for the primary outcome of mortality, and clinical data were captured prospectively for septic patients. RESULTS: The mRNA expression of HPSE was significantly increased in platelets isolated from septic patients. Ribosomal footprint profiling, followed by [S35] methionine labeling assays, demonstrated that HPSE mRNA translation and HPSE protein synthesis were significantly upregulated in platelets during sepsis. While both the pro- and active forms of HPSE protein increased in platelets during sepsis, only the active form of HPSE protein significantly correlated with sepsis-associated mortality. Consistent with transcriptomic and proteomic upregulation, HPSE enzymatic activity was also increased in platelets during sepsis. CONCLUSIONS: During clinical sepsis HPSE, translation, and enzymatic activity are increased in platelets. Increased expression of the active form of HPSE protein is associated with sepsis-associated mortality.


Assuntos
Plaquetas/enzimologia , Glucuronidase/metabolismo , Sepse , Células Endoteliais , Glucuronidase/genética , Humanos , Proteômica
6.
Blood ; 136(15): 1760-1772, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32844999

RESUMO

There is heritability to interindividual variation in platelet count, and better understanding of the regulating genetic factors may provide insights for thrombopoiesis. MicroRNAs (miRs) regulate gene expression in health and disease, and megakaryocytes (MKs) deficient in miRs have lower platelet counts, but information about the role of miRs in normal human MK and platelet production is limited. Using genome-wide miR profiling, we observed strong correlations among human bone marrow MKs, platelets, and differentiating cord blood-derived MK cultures, and identified MK miR-125a-5p as associated with human platelet number but not leukocyte or hemoglobin levels. Overexpression and knockdown studies showed that miR-125a-5p positively regulated human MK proplatelet (PP) formation in vitro. Inhibition of miR-125a-5p in vivo lowered murine platelet counts. Analyses of MK and platelet transcriptomes identified LCP1 as a miR-125a-5p target. LCP1 encodes the actin-bundling protein, L-plastin, not previously studied in MKs. We show that miR-125a-5p directly targets and reduces expression of MK L-plastin. Overexpression and knockdown studies show that L-plastin promotes MK progenitor migration, but negatively correlates with human platelet count and inhibits MK PP formation (PPF). This work provides the first evidence for the actin-bundling protein, L-plastin, as a regulator of human MK PPF via inhibition of the late-stage MK invagination system, podosome and PPF, and PP branching. We also provide resources of primary and differentiating MK transcriptomes and miRs associated with platelet counts. miR-125a-5p and L-plastin may be relevant targets for increasing in vitro platelet manufacturing and for managing quantitative platelet disorders.


Assuntos
Plaquetas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Megacariócitos/citologia , Megacariócitos/metabolismo , Glicoproteínas de Membrana/genética , MicroRNAs/genética , Proteínas dos Microfilamentos/genética , Trombopoese/genética , Actinas/metabolismo , Biomarcadores , Técnicas de Silenciamento de Genes , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Interferência de RNA
7.
Blood ; 134(12): 911-923, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31366617

RESUMO

There is increasing recognition that platelets have a functional role in the pathophysiology of sepsis, though this role has not been precisely defined. Whether sepsis alters the human platelet transcriptome and translational landscape has never been established. We used parallel techniques of RNA sequencing and ribosome footprint profiling to interrogate the platelet transcriptome and translatome in septic patients and healthy donors. We identified 1806 significantly differentially expressed (false discovery rate <0.05) transcripts in platelets from septic patients. Platelet translational events during sepsis were also upregulated. To explore the relevance of a murine model of sepsis, cecal ligation and puncture (CLP), we compared sepsis-induced changes in platelet gene expression between septic patients and mice subjected to CLP. Platelet transcriptional (ρ = 0.42, P = 3.2 × 10-285) and translational (ρ = 0.65, P = 1.09 × 10-56) changes were significantly correlated between septic patients and mice. We focused on ITGA2B, tracking and validating the expression, regulation, and functional impact of changes in ITGA2B during sepsis. Increased ITGA2B was identified in bone marrow megakaryocytes within 24 hours of sepsis onset. Subsequent increases in ITGA2B were seen in circulating platelets, suggesting dynamic trafficking of the messenger RNA. Transcriptional changes in ITGA2B were accompanied by de novo protein synthesis of αIIb and integrin αIIbß3 activation. Increased αIIb was associated with mortality in humans and mice. These findings provide previously unrecognized evidence that human and murine sepsis similarly alters the platelet transcriptional and translational landscape. Moreover, ITGA2B is upregulated and functional in sepsis due to trafficking from megakaryocytes and de novo synthesis in platelets and is associated with increased mortality.


Assuntos
Plaquetas/metabolismo , Sepse/genética , Sepse/metabolismo , Animais , Plaquetas/patologia , Proteínas Sanguíneas/análise , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Biossíntese de Proteínas , Proteoma/análise , Proteômica , Sepse/sangue , Sepse/patologia , Índice de Gravidade de Doença , Transcrição Gênica , Transcriptoma
8.
Pediatr Res ; 86(1): 55-62, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30965356

RESUMO

BACKGROUND: Dysregulated inflammation leads to morbidity and mortality in neonates. Neutrophil-mediated inflammation can cause inflammatory tissue damage. The mammalian target of rapamycin (mTOR) pathway governs IL-6Rα protein expression in human neutrophils. Shed IL-6Rα then participates in trans-signaling of IL-6/IL-6Rα to cells not otherwise sensitive to IL-6. Signaling to endothelial cells triggers efferocytosis where macrophages limit persistent inflammation by phagocytizing neutrophils. We hypothesized that preterm neonatal PMNs fail to synthesize IL-6Rα due to alterations in mTOR signaling. METHODS: We studied IL-6Rα expression, PAF receptor expression, and mTOR signaling in plasma and PAF-stimulated PMNs isolated from newborn infants and healthy adults using ELISA, real-time RT-PCR, western blotting, flow cytometry, and immunocytochemistry with phospho-specific antibodies. RESULTS: Compared to healthy adults, plasma from neonates contains significantly less soluble IL-6Rα. IL-6Rα mRNA expression in PAF-stimulated PMNs does not differ between neonates and adults, but IL-6Rα protein expression is decreased in preterm neonatal PMNs. Rapamycin, an mTOR inhibitor, blocks IL-6Rα protein expression. mTOR signaling following PAF stimulation is decreased in preterm neonatal PMNs. CONCLUSIONS: Preterm neonatal PMNs exhibit decreased mTOR pathway signaling leading to decreased IL-6Rα synthesis. Decreased synthesis of IL-6Rα by neonatal PMNs may result in decreased IL-6/IL-6Rα trans-signaling with prolonged inflammatory response and increased morbidity.


Assuntos
Regulação da Expressão Gênica , Recém-Nascido Prematuro , Interleucina-6/sangue , Neutrófilos/metabolismo , Receptores de Interleucina-6/sangue , Serina-Treonina Quinases TOR/sangue , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Adulto , Proteínas de Ciclo Celular/metabolismo , Células Endoteliais/metabolismo , Sangue Fetal/metabolismo , Humanos , Recém-Nascido , Inflamação , Macrófagos/metabolismo , Pessoa de Meia-Idade , Fagocitose , Fosforilação , Transdução de Sinais , Adulto Jovem
9.
J Thromb Haemost ; 17(3): 511-524, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30632265

RESUMO

Essentials The action of microRNAs (miRs) in human megakaryocyte signaling is largely unknown. Cord blood-derived human megakaryocytes (MKs) were used to test the function of candidate miRs. miR-15a-5p negatively regulated MK GPVI-mediated αIIbß3 activation and α-granule release. miR-15a-5p acts as a potential "master-miR" regulating genes in the MK GPVI signaling pathway. SUMMARY: Background Megakaryocytes (MKs) invest their progeny platelets with proteins and RNAs. MicroRNAs (miRs), which inhibit mRNA translation into protein, are abundantly expressed in MKs and platelets. Although platelet miRs have been associated with platelet reactivity and disease, there is a paucity of information on the function of miRs in human MKs. Objective To identify MK miRs that regulate the GPVI signaling pathway in the MK-platelet lineage. Methods Candidate miRs associated with GPVI-mediated platelet aggregation were tested for functionality in cultured MKs derived from cord blood. Results An unbiased, transcriptome-wide screen in 154 healthy donors identified platelet miR-15a-5p as significantly negatively associated with CRP-induced platelet aggregation. Platelet agonist dose-response curves demonstrated activation of αIIbß3 in suspensions of cord blood-derived cultured MKs. Overexpression and knockdown of miR-15a-5p in these MKs reduced and enhanced, respectively, CRP-induced αIIbß3 activation but did not alter thrombin or ADP stimulation. FYN, SRGN, FCER1G, MYLK. and PRKCQ, genes involved in GPVI signaling, were identified as miR-15a-5p targets and were inhibited or de-repressed in MKs with miR-15a-5p overexpression or inhibition, respectively. Lentiviral overexpression of miR-15a-5p also inhibited GPVI-FcRγ-mediated phosphorylation of Syk and PLCγ2, GPVI downstream signaling molecules, but effects of miR-15a-5p on αIIbß3 activation did not extend to other ITAM-signaling receptors (FcγRIIa and CLEC-2). Conclusion Cord blood-derived MKs are a useful human system for studying the functional effects of candidate platelet genes. miR-15a-5p is a potential "master-miR" for specifically regulating GPVI-mediated MK-platelet signaling. Targeting miR-15a-5p may have therapeutic potential in hemostasis and thrombosis.


Assuntos
Plaquetas/metabolismo , Megacariócitos/metabolismo , MicroRNAs/metabolismo , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Transdução de Sinais , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Sangue Fetal/citologia , Regulação da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , MicroRNAs/genética , Ativação Plaquetária/genética , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Transdução de Sinais/genética
10.
J Immunol ; 200(1): 295-304, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29167233

RESUMO

Dysregulated inflammation is implicated in the pathobiology of aging, yet platelet-leukocyte interactions and downstream cytokine synthesis in aging remains poorly understood. Platelets and monocytes were isolated from healthy younger (age <45, n = 37) and older (age ≥65, n = 30) adults and incubated together under autologous and nonautologous conditions. Synthesis of inflammatory cytokines by monocytes, alone or in the presence of platelets, was examined. Next-generation RNA-sequencing allowed for unbiased profiling of the platelet transcriptome in aging. Basal IL-8 and MCP-1 synthesis by monocytes alone did not differ between older and younger adults. However, in the presence of autologous platelets, monocytes from older adults synthesized greater IL-8 (41 ± 5 versus 9 ± 2 ng/ml, p < 0.0001) and MCP-1 (867 ± 150 versus 216 ± 36 ng/ml, p < 0.0001) than younger adults. Platelets from older adults were sufficient for upregulating the synthesis of inflammatory cytokines by monocytes. Using RNA-sequencing of platelets followed by validation via RT-PCR and immunoblot, we discovered that granzyme A (GrmA), a serine protease not previously identified in human platelets, increases with aging (∼9-fold versus younger adults, p < 0.05) and governs increased IL-8 and MCP-1 synthesis through TLR4 and caspase-1. Inhibiting GrmA reduced excessive IL-8 and MCP-1 synthesis in aging to levels similar to younger adults. In summary, human aging is associated with changes in the platelet transcriptome and proteome. GrmA is present and bioactive in human platelets, is higher in older adults, and controls the synthesis of inflammatory cytokines by monocytes. Alterations in the platelet molecular signature and signaling to monocytes may contribute to dysregulated inflammatory syndromes in older adults.


Assuntos
Envelhecimento/imunologia , Plaquetas/fisiologia , Quimiocina CCL2/metabolismo , Granzimas/metabolismo , Inflamação/imunologia , Interleucina-8/metabolismo , Monócitos/imunologia , Idoso , Células Cultivadas , Quimiocina CCL2/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Granzimas/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-8/genética , Masculino , Pessoa de Meia-Idade , Receptor 4 Toll-Like/metabolismo
11.
Platelets ; 28(2): 155-164, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27848259

RESUMO

Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.


Assuntos
Plaquetas/metabolismo , Doenças Transmissíveis/metabolismo , Grânulos Citoplasmáticos/metabolismo , Inflamação/metabolismo , Vesículas Secretórias/metabolismo , Animais , Doenças Transmissíveis/etiologia , Progressão da Doença , Endocitose , Exocitose , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Inflamação/etiologia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas SNARE/metabolismo
12.
Biochem J ; 468(3): 459-73, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25849538

RESUMO

Platelets are activated by a range of stimuli that share little or no resemblance in structure to each other or to recognized ligands, including diesel exhaust particles (DEP), small peptides [4N1-1, Champs (computed helical anti-membrane proteins), LSARLAF (Leu-Ser-Ala-Arg-Leu-Ala-Phe)], proteins (histones) and large polysaccharides (fucoidan, dextran sulfate). This miscellaneous group stimulate aggregation of human and mouse platelets through the glycoprotein VI (GPVI)-FcR γ-chain complex and/or C-type lectin-like receptor-2 (CLEC-2) as shown using platelets from mice deficient in either or both of these receptors. In addition, all of these ligands stimulate tyrosine phosphorylation in GPVI/CLEC-2-double-deficient platelets, indicating that they bind to additional surface receptors, although only in the case of dextran sulfate does this lead to activation. DEP, fucoidan and dextran sulfate, but not the other agonists, activate GPVI and CLEC-2 in transfected cell lines as shown using a sensitive reporter assay confirming a direct interaction with the two receptors. We conclude that this miscellaneous group of ligands bind to multiple proteins on the cell surface including GPVI and/or CLEC-2, inducing activation. These results have pathophysiological significance in a variety of conditions that involve exposure to activating charged/hydrophobic agents.


Assuntos
Poluentes Atmosféricos/toxicidade , Antígenos CD36/química , Coagulantes/farmacologia , Lectinas Tipo C/agonistas , Lectinas Tipo C/química , Glicoproteínas de Membrana/agonistas , Ativação Plaquetária/efeitos dos fármacos , Emissões de Veículos/toxicidade , Poluentes Atmosféricos/química , Poluentes Atmosféricos/metabolismo , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Linhagem Celular , Galinhas , Coagulantes/antagonistas & inibidores , Coagulantes/química , Coagulantes/metabolismo , Cruzamentos Genéticos , Genes Reporter/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células Jurkat , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ligantes , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Transgênicos , Conformação Molecular , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Engenharia de Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...