Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nutr ; 153(1): 279-292, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36913463

RESUMEN

BACKGROUND: Obesity leads to insulin resistance, altered lipoprotein metabolism, dyslipidemia, and cardiovascular disease. The relationship between long-term intake of n-3 polyunsaturated fatty acids (n-3 PUFAs) and prevention of cardiometabolic disease remains unresolved. OBJECTIVES: The aim of this study was to explore direct and indirect pathways between adiposity and dyslipidemia, and the degree to which n-3 PUFAs moderate adiposity-induced dyslipidemia in a population with highly variable n-3 PUFA intake from marine foods. METHODS: In total, 571 Yup'ik Alaska Native adults (18-87 y) were enrolled in this cross-sectional study. The red blood cell (RBC) nitrogen isotope ratio (15N/14N, or NIR) was used as a validated objective measure of n-3 PUFA intake. EPA and DHA were measured in RBCs. Insulin sensitivity and resistance were estimated by the HOMA2 method. Mediation analysis was conducted to evaluate the contribution of the indirect causal path between adiposity and dyslipidemia mediated through insulin resistance. Moderation analysis was used to assess the influence of dietary n-3 PUFAs on the direct and indirect paths between adiposity and dyslipidemia. Outcomes of primary interest included plasma total cholesterol (TC), LDL-cholesterol (LDL-C), HDL-cholesterol (HDL-C), non-HDL-C, and triglycerides (TG). RESULTS: In this Yup'ik study population, we found that up to 21.6% of the total effects of adiposity on plasma TG, HDL-C, and non-HDL-C are mediated through measures of insulin resistance or sensitivity. Moreover, RBC DHA and EPA moderated the positive association between waist circumference (WC) and TC or non-HDL-C, whereas only DHA moderated the positive association between WC and TG. However, the indirect path between WC and plasma lipids was not significantly moderated by dietary n-3 PUFAs. CONCLUSIONS: Intake of n-3 PUFAs may independently reduce dyslipidemia through the direct path resulting from excess adiposity in Yup'ik adults. NIR moderation effects suggest that additional nutrients contained in n-3 PUFA-rich foods may also reduce dyslipidemia.


Asunto(s)
Ácidos Grasos Omega-3 , Resistencia a la Insulina , Adulto , Humanos , Estudios Transversales , Obesidad , Ácidos Grasos Insaturados , Ácidos Grasos Omega-3/farmacología , Triglicéridos , HDL-Colesterol
2.
Eur J Haematol ; 110(2): 123-130, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36336470

RESUMEN

Iron deficiency is the most common nutrient deficiency in the world, affecting over 20% of premenopausal women worldwide. Oral iron supplementation is often the first-line treatment for the acute and chronic management of iron deficiency due to its ease and accessibility. However, there is no consensus on the optimal formulation or dosing strategy, or which patients should be preferentially treated with intravenous iron. Management of iron deficiency is complicated by the hepcidin-ferroportin iron regulatory pathway, which has evolved to prevent iron overload and thereby creates an inherent limit on gastrointestinal iron uptake and efficacy of oral iron. Unabsorbed iron propagates many of the side effects that complicate oral iron use including dyspepsia and constipation, all of which can thus be exacerbated by excessive oral iron doses. Daily low dose and every other day dosing protocols have attempted to bypass this physiologic bottleneck to allow for effective absorption and limit side effects; however, this approach has still resulted in low fractional iron absorption. In the following manuscript, we review the pathophysiology of iron absorption and current evidence for various preparations of oral iron. Lastly, we highlight opportunities for further study to advance the care of individuals affected by iron deficiency.


Asunto(s)
Anemia Ferropénica , Deficiencias de Hierro , Sobrecarga de Hierro , Humanos , Adulto , Femenino , Hierro/metabolismo , Sobrecarga de Hierro/tratamiento farmacológico , Administración Intravenosa , Administración Oral , Anemia Ferropénica/diagnóstico , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/etiología
3.
Platelets ; 34(1): 2217932, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37246523

RESUMEN

Proteomics tools provide a powerful means to identify, detect, and quantify protein-related details in studies of platelet phenotype and function. Here, we consider how historical and recent advances in proteomics approaches have informed our understanding of platelet biology, and, how proteomics tools can be used going forward to advance studies of platelets. It is now apparent that the platelet proteome is comprised of thousands of different proteins, where specific changes in platelet protein systems can accompany alterations in platelet function in health and disease. Going forward, many challenges remain in how to best carry out, validate and interpret platelet proteomics experiments. Future studies of platelet protein post-translational modifications such as glycosylation, or studies that take advantage of single cell proteomics and top-down proteomics methods all represent areas of interest to profiling and more richly understanding platelets in human wellness and disease.


What is the context?Platelets are well known as cellular mediators of hemostasis and drivers of thrombosis and inflammation. Thousands of different proteins come together to make up the molecular structure of platelets and support their functions in health and disease.What is new?"Proteomics" refers to laboratory approaches that study many proteins simultaneously to provide details on proteomes and the biology of a given system of interest. Many different high-throughput biochemical methods can be considered as proteomics, including mass spectrometry analysis.What is the impact?Proteomics has already identified more than 5,000 individual proteins in human platelets and provided important insights into how platelets may serve as biomarkers or therapeutic targets in human health and disease. In coming years, advances in proteomics technologies will likely help to characterize additional proteins and molecular systems in platelets to shed light on platelet-mediated mechanisms of physiology and pathology that have been largely unapproachable with other methods.


Asunto(s)
Plaquetas , Proteómica , Humanos , Plaquetas/metabolismo , Proteómica/métodos , Fenotipo , Proteoma/metabolismo
4.
Am J Physiol Cell Physiol ; 323(4): C1231-C1250, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35938677

RESUMEN

Tyrosine kinase inhibitors (TKIs) have emerged as a promising class of target-directed, small molecule inhibitors used to treat hematologic malignancies, inflammatory diseases, and autoimmune disorders. Recently, TKIs have also gained interest as potential antiplatelet-directed therapeutics that could be leveraged to reduce pathologic thrombus formation and atherothrombotic complications, while minimally affecting platelet hemostatic function. This review provides a mechanistic overview and summarizes the known effects of tyrosine kinase inhibitors on platelet signaling and function, detailing prominent platelet signaling pathways downstream of the glycoprotein VI (GPVI) receptor, integrin αIIbß3, and G protein-coupled receptors (GPCRs). This review focuses on mechanistic as well as clinically relevant and emerging TKIs targeting major families of tyrosine kinases including but not limited to Bruton's tyrosine kinase (BTK), spleen tyrosine kinase (Syk), Src family kinases (SFKs), Janus kinases (JAK), and signal transducers and activators of transcription (STAT) and evaluates their effects on platelet aggregation and adhesion, granule secretion, receptor expression and activation, and protein phosphorylation events. In summation, this review highlights current advances and knowledge on the effects of select TKIs on platelet biology and furthers insight on signaling pathways that may represent novel druggable targets coupled to specific platelet functional responses.


Asunto(s)
Hemostáticos , Activación Plaquetaria , Agammaglobulinemia Tirosina Quinasa/metabolismo , Plaquetas/metabolismo , Hemostáticos/metabolismo , Hemostáticos/farmacología , Quinasas Janus/metabolismo , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasa Syk/metabolismo , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
5.
Am J Physiol Cell Physiol ; 322(3): C370-C381, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35080922

RESUMEN

Cannabis usage has steadily increased as acceptance is growing for both medical and recreational reasons. Medical cannabis is administered for treatment of chronic pain based on the premise that the endocannabinoid system signals desensitize pain sensor neurons and produce anti-inflammatory effects. The major psychoactive ingredient of cannabis is Δ9-tetrahydrocannabinol (THC) that signals mainly through cannabinoid receptor-1 (CBr), which is also present on nonneuron cells including blood platelets of the circulatory system. In vitro, CBr-mediated signaling has been shown to acutely inhibit platelet activation downstream of the platelet collagen receptor glycoprotein (GP)VI. The systemic effects of chronic THC administration on platelet activity and function remain unclear. This study investigates the effects of chronic THC administration on platelet function using a nonhuman primate (NHP) model. Our results show that female and male NHPs consuming a daily THC edible had reduced platelet adhesion, aggregation, and granule secretion in response to select platelet agonists. Furthermore, a change in bioactive lipids (oxylipins) was observed in the female cohort after THC administration. These results indicate that chronic THC edible administration desensitized platelet activity and function in response to GPVI- and G-protein coupled receptor-based activation by interfering with primary and secondary feedback signaling pathways. These observations may have important clinical implications for patients who use medical marijuana and for providers caring for these patients.


Asunto(s)
Plaquetas/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/administración & dosificación , Dronabinol/administración & dosificación , Marihuana Medicinal/administración & dosificación , Administración Oral , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Femenino , Macaca mulatta , Masculino , Oxilipinas/sangre , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Transducción de Señal , Tromboxanos/sangre , Factores de Tiempo
6.
Blood ; 136(20): 2346-2358, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32640021

RESUMEN

Platelets engage cues of pending vascular injury through coordinated adhesion, secretion, and aggregation responses. These rapid, progressive changes in platelet form and function are orchestrated downstream of specific receptors on the platelet surface and through intracellular signaling mechanisms that remain systematically undefined. This study brings together cell physiological and phosphoproteomics methods to profile signaling mechanisms downstream of the immunotyrosine activation motif (ITAM) platelet collagen receptor GPVI. Peptide tandem mass tag (TMT) labeling, sample multiplexing, synchronous precursor selection (SPS), and triple stage tandem mass spectrometry (MS3) detected >3000 significant (false discovery rate < 0.05) phosphorylation events on >1300 proteins over conditions initiating and progressing GPVI-mediated platelet activation. With literature-guided causal inference tools, >300 site-specific signaling relations were mapped from phosphoproteomics data among key and emerging GPVI effectors (ie, FcRγ, Syk, PLCγ2, PKCδ, DAPP1). Through signaling validation studies and functional screening, other less-characterized targets were also considered within the context of GPVI/ITAM pathways, including Ras/MAPK axis proteins (ie, KSR1, SOS1, STAT1, Hsp27). Highly regulated GPVI/ITAM targets out of context of curated knowledge were also illuminated, including a system of >40 Rab GTPases and associated regulatory proteins, where GPVI-mediated Rab7 S72 phosphorylation and endolysosomal maturation were blocked by TAK1 inhibition. In addition to serving as a model for generating and testing hypotheses from omics datasets, this study puts forth a means to identify hemostatic effectors, biomarkers, and therapeutic targets relevant to thrombosis, vascular inflammation, and other platelet-associated disease states.


Asunto(s)
Algoritmos , Activación Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteómica/métodos , Animales , Humanos , Transducción de Señal/fisiología
7.
J Nutr ; 152(3): 844-855, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34871429

RESUMEN

BACKGROUND: The relationship between dietary n-3 PUFAs and the prevention of cardiometabolic diseases, including type 2 diabetes, is unresolved. Examination of the association between n-3 PUFAs and chronic low-grade inflammation in a population where many individuals have had an extremely high intake of marine mammals and fish throughout their lifespan may provide important clues regarding the impact of n-3 PUFAs on health. OBJECTIVES: The aim of this study was to explore associations between concentrations of n-3 PUFAs resulting from habitual intake of natural food sources high in fish and marine mammals with immune biomarkers of metabolic inflammation and parameters of glucose regulation. METHODS: A total of 569 Yup'ik Alaska Native adults (18-87 years old) were enrolled in this cross-sectional study between December 2016 and November 2019. The RBC nitrogen isotope ratio (NIR; 15N/14N) was used as a validated measure of n-3 PUFA intake to select 165 participant samples from the first and fourth quartiles of n-3 PUFA intakes. Outcomes included 38 pro- and anti-inflammatory cytokines and 8 measures of glucose homeostasis associated with type 2 diabetes risks. These outcomes were evaluated for their associations with direct measurements of EPA, DHA, and arachidonic acid in RBCs. ANALYSIS: Linear regression was used to detect significant relationships with cytokines and n-3 PUFAs, adiposity, and glucose-related variables. RESULTS: The DHA concentration in RBC membranes was inversely associated with IL-6 (ß = -0.0066; P < 0.001); EPA was inversely associated with TNFα (ß = -0.4925; P < 0.001); and the NIR was inversely associated with Monocyte chemoattractant protein-1 (MCP-1) (ß = -0.8345; P < 0.001) and IL-10 (ß = -1.2868; P < 0.001). CONCLUSIONS: Habitual intake of marine mammals and fish rich in n-3 PUFAs in this study population of Yup'ik Alaska Native adults is associated with reduced systemic inflammation, which may contribute to the low prevalence of diseases in which inflammation plays an important role.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Ácidos Grasos Omega-3 , Animales , Estudios Transversales , Citocinas , Diabetes Mellitus Tipo 2/prevención & control , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-3/farmacología , Peces/metabolismo , Glucosa , Humanos , Inflamación , Mamíferos
8.
Eur J Haematol ; 109(6): 633-642, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36153674

RESUMEN

Iron deficiency and/or iron deficiency anemia (IDA) complicate nearly 50% of pregnancies globally, negatively impacting both maternal and fetal outcomes. Iron deficiency can cause a range of symptoms that range from aggravating to debilitating including fatigue, poor quality of life, pagophagia, and restless leg syndrome. Iron deficiency and IDA are also associated with maternal complications including preterm labor, increased rates of cesarean delivery, postpartum hemorrhage, and maternal death. Fetal complications include increased rates of low birth weight and small for gestational age newborns. Prenatal maternal anemia has also been associated with autism spectrum disorders in the neonate, although causation is not established. Deficiency in the newborn is associated with compromised memory, processing, and bonding, with some of these deficits persisting into adulthood. Despite the prevalence and consequences associated with iron deficiency in pregnancy, data show that it is routinely undertreated. Due to the physiologic changes of pregnancy, all pregnant individuals should receive oral iron supplementation. However, the bioavailability of oral iron is poor and it is often ineffective at preventing and treating iron deficiency. Likewise, it frequently causes gastrointestinal symptoms that can worsen the quality of life in pregnancy. Intravenous iron formulations administered in a single or multiple dose series are now available. There is increasing data suggesting that newer intravenous formulations are safe and effective in the second and third trimesters and should be strongly considered in pregnant individuals without optimal response to oral iron repletion.


Asunto(s)
Anemia Ferropénica , Anemia , Deficiencias de Hierro , Complicaciones Hematológicas del Embarazo , Embarazo , Femenino , Recién Nacido , Humanos , Adulto , Incidencia , Calidad de Vida , Anemia Ferropénica/diagnóstico , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/epidemiología , Hierro , Anemia/tratamiento farmacológico , Complicaciones Hematológicas del Embarazo/tratamiento farmacológico , Complicaciones Hematológicas del Embarazo/epidemiología , Complicaciones Hematológicas del Embarazo/etiología
9.
Arterioscler Thromb Vasc Biol ; 41(3): 999-1011, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33441027

RESUMEN

Platelets rapidly undergo responsive transitions in form and function to repair vascular endothelium and mediate hemostasis. In contrast, heterogeneous platelet subpopulations with a range of primed or refractory phenotypes gradually arise in chronic inflammatory and other conditions in a manner that may indicate or support disease. Qualitatively distinguishable platelet phenotypes are increasingly associated with a variety of physiological and pathological circumstances; however, the origins and significance of platelet phenotypic variation remain unclear and conceptually vague. As changes in platelet function in disease exhibit many similarities to platelets following the activation of platelet agonist receptors, the intracellular responses of platelets common to hemostasis and inflammation may provide insights to the molecular basis of platelet phenotype. Here, we review concepts around how protein-level relations-from platelet receptors through intracellular signaling events-may help to define platelet phenotypes in inflammation, immune responses, aging, and other conditions. We further discuss how representing systems-wide platelet proteomics data profiles as circuit-like networks of causally related intracellular events, or, pathway maps, may inform molecular definitions of platelet phenotype. In addition to offering insights into platelets as druggable targets, maps of causally arranged intracellular relations underlying platelet function can also advance precision and interceptive medicine efforts by leveraging platelets as accessible, dynamic, endogenous, circulating biomarkers of vascular wellness and disease. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Plaquetas/fisiología , Proteoma/fisiología , Enfermedades Vasculares/sangre , Animales , Biomarcadores/sangre , Endotelio Vascular/fisiología , Hemostasis/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/sangre , Péptidos y Proteínas de Señalización Intracelular/fisiología , Modelos Cardiovasculares , Fenotipo , Proteómica , Transducción de Señal , Enfermedades Vasculares/fisiopatología
10.
J Pept Sci ; 28(5): e3382, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34859535

RESUMEN

Disintegrins comprise a family of small proteins that bind to and alter the physiological function of integrins, especially integrins that mediate platelet aggregation in blood. Here, we report a lysine-glycine-aspartic acid (KGD) disintegrin-like motif present in a 15-amino acid residue peptide identified in a cDNA library of the amphibian Hypsiboas punctatus skin. The original peptide sequence was used as a template from which five new analogs were designed, chemically synthesized by solid phase, and tested for disintegrin activity and tridimensional structural studies using NMR spectroscopy. The original amphibian peptide had no effect on integrin-mediated responses. Nevertheless, derived peptide analogs inhibited integrin-mediated platelet function, including platelet spreading on fibrinogen.


Asunto(s)
Desintegrinas , Péptidos , Anfibios/genética , Anfibios/metabolismo , Animales , ADN Complementario/genética , Desintegrinas/química , Desintegrinas/genética , Desintegrinas/farmacología , Péptidos/química , Péptidos/genética , Péptidos/farmacología , Agregación Plaquetaria/fisiología
11.
Platelets ; 33(3): 404-415, 2022 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-34097573

RESUMEN

Several Janus kinase (JAK) inhibitors (jakinibs) have recently been approved to treat inflammatory, autoimmune and hematological conditions. Despite emerging roles for JAKs and downstream signal transducer and activator of transcription (STAT) proteins in platelets, it remains unknown whether jakinibs affect platelet function. Here, we profile platelet biochemical and physiological responses in vitro in the presence of five different clinically relevant jakinibs, including ruxolitinib, upadacitinib, oclacitinib, baricitinib and tofacitinib. Flow cytometry, microscopy and other assays found that potent JAK1/2 inhibitors baricitinib and ruxolitinib reduced platelet adhesion to collagen, as well as platelet aggregation, secretion and integrin αIIbß3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP-XL). Western blot analysis demonstrated that jakinibs reduced Akt phosphorylation and activation following GPVI activation, where ruxolitinib and baricitinib prevented DAPP1 phosphorylation. In contrast, jakinibs had no effects on platelet responses to thrombin. Inhibitors of GPVI and JAK signaling also abrogated platelet STAT5 phosphorylation following CRP-XL stimulation. Additional pharmacologic experiments supported roles for STAT5 in platelet secretion, integrin activation and cytoskeletal responses. Together, our results demonstrate that ruxolitinib and baricitinib have inhibitory effects on platelet function in vitro and support roles for JAK/STAT5 pathways in GPVI/ITAM mediated platelet function.


Asunto(s)
Azetidinas/uso terapéutico , Plaquetas/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Nitrilos/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/efectos de los fármacos , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Azetidinas/farmacología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Nitrilos/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Sulfonamidas/farmacología
12.
Am J Physiol Cell Physiol ; 320(5): C902-C915, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33689480

RESUMEN

Spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (BTK) play critical roles in platelet physiology, facilitating intracellular immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling downstream of platelet glycoprotein VI (GPVI) and GPIIb/IIIa receptors. Small molecule tyrosine kinase inhibitors (TKIs) targeting Syk and BTK have been developed as antineoplastic and anti-inflammatory therapeutics and have also gained interest as antiplatelet agents. Here, we investigate the effects of 12 different Syk and BTK inhibitors on GPVI-mediated platelet signaling and function. These inhibitors include four Syk inhibitors, Bay 61-3606, R406 (fostamatinib), entospletinib, TAK-659; four irreversible BTK inhibitors, ibrutinib, acalabrutinib, ONO-4059 (tirabrutinib), AVL-292 (spebrutinib); and four reversible BTK inhibitors, CG-806, BMS-935177, BMS-986195, and fenebrutinib. In vitro, TKIs targeting Syk or BTK reduced platelet adhesion to collagen, dense granule secretion, and alpha granule secretion in response to the GPVI agonist cross-linked collagen-related peptide (CRP-XL). Similarly, these TKIs reduced the percentage of activated integrin αIIbß3 on the platelet surface in response to CRP-XL, as determined by PAC-1 binding. Although all TKIs tested inhibited phospholipase C γ2 (PLCγ2) phosphorylation following GPVI-mediated activation, other downstream signaling events proximal to phosphoinositide 3-kinase (PI3K) and PKC were differentially affected. In addition, reversible BTK inhibitors had less pronounced effects on GPIIb/IIIa-mediated platelet spreading on fibrinogen and differentially altered the organization of PI3K around microtubules during platelets spreading on fibrinogen. Select TKIs also inhibited platelet aggregate formation on collagen under physiological flow conditions. Together, our results suggest that TKIs targeting Syk or BTK inhibit central platelet functional responses but may differentially affect protein activities and organization in critical systems downstream of Syk and BTK in platelets.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Plaquetas/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinasa Syk/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Plaquetas/enzimología , Femenino , Humanos , Masculino , Terapia Molecular Dirigida , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal , Quinasa Syk/metabolismo
13.
Physiol Genomics ; 53(11): 473-485, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34677084

RESUMEN

Hibernating mammals undergo a dramatic drop in temperature and blood flow during torpor, yet avoid stasis blood clotting through mechanisms that remain unspecified. The effects of hibernation on hemostasis are especially complex, as cold temperatures generally activate platelets, resulting in platelet clearance and cold storage lesions in the context of blood transfusion. With a hibernating body temperature of 4°C-8°C, 13-lined ground squirrels (Ictidomys tridecemlineatus) provide a model to study hemostasis as well as platelet cold storage lesion resistance during hibernation. Here, we quantified and systematically compared proteomes of platelets collected from ground squirrels at summer (active), fall (entrance), and winter (topor) to elucidate how molecular-level changes in platelets may support hemostatic adaptations in torpor. Platelets were isolated from a total of 11 squirrels in June, October, and January. Platelet lysates from each animal were digested with trypsin prior to 11-plex tandem mass tag (TMT) labeling, followed by LC-MS/MS analysis for relative protein quantification. We measured >700 proteins with significant variations in abundance in platelets over the course of entrance, torpor, and activity-including systems of proteins regulating translation, secretion, metabolism, complement, and coagulation cascades. We also noted species-specific differences in levels of hemostatic, secretory, and inflammatory regulators in ground squirrel platelets relative to human platelets. Altogether, we provide the first ever proteomic characterization of platelets from hibernating animals, where systematic changes in metabolic, hemostatic, and other proteins may account for physiological adaptations in torpor and also inform translational effort to improve cold storage of human platelets for transfusion.


Asunto(s)
Plaquetas/química , Hibernación/fisiología , Proteoma/química , Sciuridae/sangre , Estaciones del Año , Animales , Cromatografía Liquida/métodos , Femenino , Humanos , Masculino , Proteómica/métodos , Especificidad de la Especie , Espectrometría de Masas en Tándem/métodos , Temperatura
14.
Platelets ; 32(3): 424-428, 2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32233694

RESUMEN

The core structure of the extracellular basement membrane is made up of self-assembling networks of collagen and laminin which associate with each other through the bridging adapter proteins including the sulfated monomeric glycoprotein nidogen. While collagen and laminin are known to support platelet adhesion and activation via ß1 integrins and glycoprotein (GP) VI, respectively, whether nidogen contributes to platelet activation and hemostasis is unknown. In this study, we demonstrate that recombinant human nidogen-1 supports platelet adhesion and stimulates platelet activation in a phospholipase-C γ-2 (PLCγ2), Src and Syk kinase-dependent manner downstream. Platetet adhesion to nidogen-1 was inhibited by blocking the platelet receptors GPVI and ß1 integrins. Platelet adhesion to nidogen-1 activated the IκB kinase (IKK) complex, while pharmacological inhibition of IKK blocked platelet spreading on nidogen. Taken together our results suggest that nidogen may play a redundant role in hemostasis by activating platelets downstream of GPVI.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Humanos
15.
Proteomics ; 19(11): e1900001, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30977292

RESUMEN

Protein posttranslational modifications critically regulate a range of physiological and disease processes. In addition to tyrosine, serine, and threonine phosphorylation, reversible N-ε acylation and alkylation of protein lysine residues also modulate diverse aspects of cellular function. Studies of lysine acyl and alkyl modifications have focused on nuclear proteins in epigenetic regulation; however, lysine modifications are also prevalent on cytosolic proteins to serve increasingly apparent, although less understood roles in cell regulation. Here, the methyl-lysine (meK) proteome of anucleate blood platelets is characterized. With high-resolution, multiplex MS methods, 190 mono-, di-, and tri-meK modifications are identified on 150 different platelet proteins-including 28 meK modifications quantified by tandem mass tag (TMT) labeling. In addition to identifying meK modifications on calmodulin (CaM), GRP78 (HSPA5, BiP), and EF1A1 that have been previously characterized in other cell types, more novel modifications are also uncovered on cofilin, drebin-like protein (DBNL, Hip-55), DOCK8, TRIM25, and numerous other cytoplasmic proteins. Together, the results and analyses support roles for lysine methylation in mediating cytoskeletal, translational, secretory, and other cellular processes. MS data for this study have been deposited into the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD012217.


Asunto(s)
Plaquetas/química , Lisina/análisis , Procesamiento Proteico-Postraduccional , Proteoma/química , Plaquetas/citología , Chaperón BiP del Retículo Endoplásmico , Humanos , Metilación , Proteómica , Espectrometría de Masas en Tándem
16.
J Cell Sci ; 130(11): 1865-1876, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28476937

RESUMEN

Vertebrate proteins that fulfill multiple and seemingly disparate functions are increasingly recognized as vital solutions to maintaining homeostasis in the face of the complex cell and tissue physiology of higher metazoans. However, the molecular adaptations that underpin this increased functionality remain elusive. In this Commentary, we review the PACS proteins - which first appeared in lower metazoans as protein traffic modulators and evolved in vertebrates to integrate cytoplasmic protein traffic and interorganellar communication with nuclear gene expression - as examples of protein adaptation 'caught in the act'. Vertebrate PACS-1 and PACS-2 increased their functional density and roles as metabolic switches by acquiring phosphorylation sites and nuclear trafficking signals within disordered regions of the proteins. These findings illustrate one mechanism by which vertebrates accommodate their complex cell physiology with a limited set of proteins. We will also highlight how pathogenic viruses exploit the PACS sorting pathways as well as recent studies on PACS genes with mutations or altered expression that result in diverse diseases. These discoveries suggest that investigation of the evolving PACS protein family provides a rich opportunity for insight into vertebrate cell and organ homeostasis.


Asunto(s)
Homeostasis/genética , Neoplasias/genética , Enfermedades Neurodegenerativas/genética , Obesidad/genética , Proteínas de Transporte Vesicular/genética , Adaptación Biológica , Animales , Apoptosis , Transporte Biológico , Señalización del Calcio , Secuencia Conservada , Regulación de la Expresión Génica , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Obesidad/metabolismo , Obesidad/patología , Filogenia , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas de Transporte Vesicular/metabolismo
17.
Platelets ; 30(1): 17-22, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29799302

RESUMEN

Rho GTPases are master orchestrators of cytoskeletal dynamics and serve critical roles in platelet physiology to promote hemostasis or pathology in thrombotic, inflammatory and other disease states. Over the past 25 years, specific platelet cell biological outputs have been linked to the activities of Rho GTPases, including RhoA, Rac1, Cdc42, and RhoG in shape change and secretion as well as cytoskeletal assembly events underlying platelet aggregation and thrombus stability. Rho GTPases have also more recently been noted to serve more specialized roles in platelet function and to cooperate with one another in mediating essential platelet responses. The evolving molecular mechanisms regulating platelet Rho GTPase functions are increasingly complex, involving an interdependent array of signal transduction molecules, including several protein kinases as well as numerous Rho GEFs, GAPs, and GDI proteins such as LARG, ARHGEF6 (Cool-2, α-Pix), ARHGEF10, GIT1, ARHGAP17 (Nadrin, Rich1), OPHN1, and Ly-GDI. In this review, we provide an update of recent work and developing hypotheses further establishing more specialized as well as cooperative roles for Rho GTPases in platelet physiology and emerging regulatory and downstream effector mechanisms whereby Rho GTPases participate in platelet activation programs in physiology and an expanding set of platelet-associated disease states.


Asunto(s)
Plaquetas/fisiología , Susceptibilidad a Enfermedades , Proteínas de Unión al GTP rho/metabolismo , Animales , Biomarcadores , Humanos , Espacio Intracelular/metabolismo , Activación Plaquetaria , Agregación Plaquetaria
18.
Platelets ; 30(1): 126-135, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30560697

RESUMEN

Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.


Asunto(s)
Plaquetas/metabolismo , Comunicación Celular , Gránulos Citoplasmáticos/metabolismo , Leucocitos/metabolismo , Receptores de Trombina/metabolismo , Animales , Biomarcadores , Citometría de Flujo , Humanos , Masculino , Papio , Activación Plaquetaria , Agregación Plaquetaria
19.
Am J Physiol Cell Physiol ; 314(5): C603-C615, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29412690

RESUMEN

Upon encountering physiological cues associated with damaged or inflamed endothelium, blood platelets set forth intracellular responses to ultimately support hemostatic plug formation and vascular repair. To gain insights into the molecular events underlying platelet function, we used a combination of interactome, pathway analysis, and other systems biology tools to analyze associations among proteins functionally modified by reversible phosphorylation upon platelet activation. While an interaction analysis mapped out a relative organization of intracellular mediators in platelet signaling, pathway analysis revealed directional signaling relations around protein kinase C (PKC) isoforms and mitogen-activated protein kinases (MAPKs) associated with platelet cytoskeletal dynamics, inflammatory responses, and hemostatic function. Pathway and causality analysis further suggested that platelets activate a specific p38-MK2 axis to phosphorylate RTN4 (reticulon-4, also known as Nogo), a Bcl-xl sequestration protein and critical regulator of endoplasmic reticulum (ER) physiology. In vitro, we find that platelets drive a p38-MK2-RTN4-Bcl-xl pathway associated with the regulation of the ER and platelet phosphatidylserine exposure. Together, our results support the use of pathway tools in the analysis of omics data sets as a means to help generate novel, mechanistic, and testable hypotheses for platelet studies while uncovering RTN4 as a putative regulator of platelet cell physiological responses.


Asunto(s)
Coagulación Sanguínea , Plaquetas/enzimología , Retículo Endoplásmico/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nogo/metabolismo , Activación Plaquetaria , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Biología Computacional , Bases de Datos de Proteínas , Activación Enzimática , Humanos , Fenotipo , Fosfatidilserinas/metabolismo , Fosforilación , Transducción de Señal , Proteína bcl-X/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA