Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Infect Immun ; 91(3): e0055622, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36853027

RESUMO

Platelets are known for essential activities in hemostasis and for their important contribution to protection against infectious pathogens. Klebsiella pneumoniae is an opportunistic pathogen widely known to cause nosocomial infections. Recently, hypervirulent strains of K. pneumoniae have been emerging, which can cause severe infections in immunocompetent individuals. Combined with the increase in antibiotic resistance, it is important to understand how K. pneumoniae affects components of the immune system. We studied the interactions of human platelets with several K. pneumoniae strains (the wild type encapsulated strain, and a nonencapsulated mutant). Thrombin-stimulated whole human and mouse blood significantly inhibited bacterial growth compared to unstimulated whole blood. Furthermore, we investigated the effect of K. pneumoniae on platelet activation. Both strains induced significant increase in activation of both unstimulated and thrombin-stimulated human platelets. Additionally, only the nonencapsulated mutant increased aggregation of platelets in response to ADP. K. pneumoniae killing assays were then performed with washed platelets in the presence or absence of thrombin. Surprisingly, washed platelets failed to exhibit any effects on the growth of K. pneumoniae. We further explored the impact of platelets on monocyte-mediated killing of K. pneumoniae. Importantly, we found that activated platelets significantly enhanced monocyte-mediated killing of K. pneumoniae. This effect was likely due to the formation of platelet-monocyte aggregates in blood upon thrombin stimulation. Overall, this study highlights the role of platelets in mediating a protective response against K. pneumoniae and reinforces the importance of platelets in modulating leukocyte behavior.


Assuntos
Plaquetas , Infecções por Klebsiella , Animais , Camundongos , Humanos , Klebsiella pneumoniae , Monócitos , Trombina/farmacologia , Ativação Plaquetária , Infecções por Klebsiella/microbiologia , Antibacterianos
2.
Am J Pathol ; 190(10): 2039-2055, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32650005

RESUMO

This study investigated intercellular adhesion molecule-1 (ICAM-1), a membrane protein that mediates cell-to-cell adhesion and communication, as a mechanism through which the inflammatory response facilitates muscle regeneration after injury. Toxin-induced muscle injury to tibialis anterior muscles of wild-type mice caused ICAM-1 to be expressed by a population of satellite cells/myoblasts and myofibers. Myogenic cell expression of ICAM-1 contributed to the restoration of muscle structure after injury, as regenerating myofibers were more abundant and myofiber size was larger for wild-type compared with Icam1-/- mice during 28 days of recovery. Contrastingly, restoration of muscle function after injury was similar between the genotypes. ICAM-1 facilitated the restoration of muscle structure after injury through mechanisms involving the regulation of myofiber branching, protein synthesis, and the organization of nuclei within myofibers after myogenic cell fusion. These findings provide support for a paradigm in which ICAM-1 expressed by myogenic cells after muscle injury augments their adhesive and fusogenic properties, which, in turn, facilitates regenerative and hypertrophic processes that restore structure to injured muscle.


Assuntos
Adesão Celular/fisiologia , Molécula 1 de Adesão Intercelular/metabolismo , Desenvolvimento Muscular/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Animais , Comunicação Celular/fisiologia , Feminino , Hipertrofia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Regeneração/genética
3.
Int J Mol Sci ; 22(3)2021 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-33498864

RESUMO

The paracrine and autocrine processes of the host response play an integral role in the success of scaffold-based tissue regeneration. Recently, the immunomodulatory scaffolds have received huge attention for modulating inflammation around the host tissue through releasing anti-inflammatory cytokine. However, controlling the inflammation and providing a sustained release of anti-inflammatory cytokine from the scaffold in the digestive inflammatory environment are predicated upon a comprehensive understanding of three fundamental questions. (1) How does the release rate of cytokine from the scaffold change in the digestive inflammatory environment? (2) Can we prevent the premature scaffold degradation and burst release of the loaded cytokine in the digestive inflammatory environment? (3) How does the scaffold degradation prevention technique affect the immunomodulatory capacity of the scaffold? This study investigated the impacts of the digestive inflammatory environment on scaffold degradation and how pre-mature degradation can be prevented using genipin crosslinking and how genipin crosslinking affects the interleukin-4 (IL-4) release from the scaffold and differentiation of naïve macrophages (M0). Our results demonstrated that the digestive inflammatory environment (DIE) attenuates protein retention within the scaffold. Over 14 days, the encapsulated protein released 46% more in DIE than in phosphate buffer saline (PBS), which was improved through genipin crosslinking. We have identified the 0.5 (w/v) genipin concentration as an optimal concentration for improved IL-4 released from the scaffold, cell viability, mechanical strength, and scaffold porosity, and immunomodulation studies. The IL-4 released from the injectable scaffold could differentiate naïve macrophages to an anti-inflammatory (M2) lineage; however, upon genipin crosslinking, the immunomodulatory capacity of the scaffold diminished significantly, and pro-inflammatory markers were expressed dominantly.


Assuntos
Regeneração Tecidual Guiada/métodos , Imunomodulação , Iridoides/farmacologia , Macrófagos/efeitos dos fármacos , Alicerces Teciduais/química , Animais , Diferenciação Celular , Células Cultivadas , Colágeno , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Injeções , Interleucina-4/imunologia , Interleucina-4/metabolismo , Iridoides/uso terapêutico , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Sistema Musculoesquelético/imunologia , Porosidade
4.
Infect Immun ; 88(4)2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-31932331

RESUMO

Candida albicans is a pervasive commensal fungus that is the most common pathogen responsible for invasive fungal infection (IFI). With incidence of IFI on the rise due to increasing susceptible populations, it is imperative that we investigate how Candida albicans interacts with blood components. When stimulating either human or mouse whole blood with thrombin, we saw a significant decrease in C. albicans survival. We then repeated Candida killing assays with thrombin-stimulated or unstimulated washed platelets and saw a similar decrease in CFU. To investigate whether killing was mediated through surface components or releasable products, platelets were pretreated with an inhibitor of actin polymerization (cytochalasin D [CytoD]). CytoD was able to abrogate C. albicans killing. Moreover, dilution of releasates from thrombin-stimulated platelets showed that the toxicity of the releasates on C. albicans is concentration dependent. We then investigated C. albicans actions on platelet activation, granule release, and aggregation. While C. albicans does not appear to affect alpha or dense granule release, C. albicans exerts a significant attenuation of platelet aggregation to multiple agonists. These results illustrate for the first time that platelets can directly kill C. albicans through release of their granular contents. Additionally, C. albicans can also exert inhibitory effects on platelet aggregation.


Assuntos
Antifúngicos/metabolismo , Plaquetas/metabolismo , Plaquetas/microbiologia , Candida albicans/imunologia , Fatores Imunológicos/metabolismo , Animais , Candida albicans/fisiologia , Contagem de Colônia Microbiana , Humanos , Camundongos , Viabilidade Microbiana/efeitos dos fármacos
5.
J Immunol ; 198(1): 344-351, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27895175

RESUMO

Platelets are the chief effector cells in hemostasis. However, recent evidence suggests they have multiple roles in host defense against infection. Reports by us and others showed that platelets functionally contribute to protection against Staphylococcus aureus infection. In the current study, the capacity of mouse platelets to participate in host defense against S. aureus infection was determined by assessing two possibilities. First, we determined the ability of platelets to kill S. aureus directly; and, second, we tested the possibility that platelets enhance macrophage phagocytosis and intracellular killing of S. aureus In this study we report evidence in support of both mechanisms. Platelets effectively killed two different strains of S. aureus. A clinical isolate of methicillin-resistant S. aureus was killed by platelets (>40% killing in 2 h) in a thrombin-dependent manner whereas a methicillin-sensitive strain was killed to equal extent but did not require thrombin. Interestingly, thrombin-stimulated platelets also significantly enhanced peritoneal macrophage phagocytosis of both methicillin-resistant S. aureus and methicillin-sensitive S. aureus by >70%, and restricted intracellular growth by >40%. Enhancement of macrophage anti-S. aureus activities is independent of contact with platelets but is mediated through releasable products, namely IL-1ß. These data confirm our hypothesis that platelets participate in host defense against S. aureus both through direct killing of S. aureus and enhancing the antimicrobial function of macrophages in protection against S. aureus infection.


Assuntos
Plaquetas/imunologia , Citotoxicidade Imunológica/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Infecções Estafilocócicas/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Staphylococcus aureus/imunologia
6.
Infect Immun ; 86(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29914928

RESUMO

Staphylococcus aureus is a major human pathogen that can cause mild to severe life-threatening infections in many tissues and organs. Platelets are known to participate in protection against S. aureus by direct killing and by enhancing the activities of neutrophils and macrophages in clearing S. aureus infection. Platelets have also been shown to induce monocyte differentiation into dendritic cells and to enhance activation of dendritic cells. Therefore, in the present study, we explored the role of platelets in enhancing bone marrow-derived dendritic cell (BMDC) function against S. aureus We observed a significant increase in dendritic cell phagocytosis and intracellular killing of a methicillin-resistant Staphylococcus aureus (MRSA) strain (USA300) by thrombin-activated platelets or their releasates. Enhancement of bacterial uptake and killing by DCs is mediated by platelet-derived CD40L. Coculture of USA300 and BMDCs in the presence of thrombin-activated platelet releasates invokes upregulation of the maturation marker CD80 on DCs and enhanced production of the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 12 (IL-12), and IL-6. Overall, these observations support our hypothesis that platelets play a critical role in the host defense against S. aureus infection. Platelets stimulate DCs, leading to direct killing of S. aureus and enhanced DC maturation, potentially leading to adaptive immune responses against S. aureus.


Assuntos
Plaquetas/imunologia , Ligante de CD40/imunologia , Citotoxicidade Imunológica/fisiologia , Células Dendríticas/imunologia , Ativação Plaquetária/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Antígeno B7-1/metabolismo , Citocinas/metabolismo , Humanos , Ativação de Macrófagos/fisiologia , Macrófagos/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Fagocitose/imunologia
7.
J Immunol ; 190(12): 6457-67, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23677468

RESUMO

Elevated numbers of activated platelets circulate in patients with chronic inflammatory diseases, including atherosclerosis and coronary disease. Activated platelets can activate the complement system. Although complement activation is essential for immune responses and removal of spent cells from circulation, it also contributes to inflammation and thrombosis, especially in patients with defective complement regulation. Proinflammatory activated leukocytes, which interact directly with platelets in response to vascular injury, are among the main sources of properdin, a positive regulator of the alternative pathway. The role of properdin in complement activation on stimulated platelets is unknown. Our data show that physiological forms of human properdin bind directly to human platelets after activation by strong agonists in the absence of C3, and bind nonproportionally to surface CD62P expression. Activation of the alternative pathway on activated platelets occurs when properdin is on the surface and recruits C3b or C3(H2O) to form C3b,Bb or a novel cell-bound C3 convertase [C3(H2O),Bb], which normally is present only in the fluid phase. Alternatively, properdin can be recruited by C3(H2O) on the platelet surface, promoting complement activation. Inhibition of factor H-mediated cell surface complement regulation significantly increases complement deposition on activated platelets with surface properdin. Finally, properdin released by activated neutrophils binds to activated platelets. Altogether, these data suggest novel molecular mechanisms for alternative pathway activation on stimulated platelets that may contribute to localization of inflammation at sites of vascular injury and thrombosis.


Assuntos
Complemento C3/imunologia , Via Alternativa do Complemento/fisiologia , Ativação Plaquetária/fisiologia , Properdina/imunologia , Plaquetas/imunologia , Plaquetas/metabolismo , Complemento C3/metabolismo , Humanos , Properdina/metabolismo
8.
Am J Pathol ; 182(1): 244-54, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23141924

RESUMO

Platelets are known for their important role in hemostasis, however their significance in other functions, including inflammation and infection, are becoming more apparent. Patients with systemic lupus erythematosus (SLE) are known to have circulating IgG complexes in their blood and are highly susceptible to thrombotic events. Because platelets express a single receptor for IgG, we tested the hypothesis that ligation of this receptor (FcγRIIa) induces platelet hypersensitivity to thrombotic stimuli. Platelets from SLE patients were considerably more sensitive to thrombin compared to healthy volunteers, and this correlated with elevated levels of surface IgG on SLE platelets. To test whether FcγRIIa ligation stimulated thrombin hypersensitivity, platelets from healthy volunteers were incubated with buffer or heat-aggregated IgG, then stimulated with increasing concentrations of thrombin. Interestingly, heat-aggregated IgG-stimulated platelets, but not buffer-treated platelets, were hypersensitive to thrombin, and hypersensitivity was blocked by an anti-FcγRIIa monoclonal antibody (mAb). Thrombin hypersensitivity was not due to changes in thrombin receptor expression (GPIbα or PAR1) but is dependent on activation of shared signaling molecules. These observations suggest that ligation of platelet FcγRIIa by IgG complexes induces a hypersensitive state whereby small changes in thrombotic stimuli may result in platelet activation and subsequent vascular complications such as transient ischemic attacks or stroke.


Assuntos
Lúpus Eritematoso Sistêmico/complicações , Ativação Plaquetária/fisiologia , Receptores de IgG/fisiologia , Trombose/etiologia , Adulto , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Células Cultivadas , Feminino , Temperatura Alta , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade , Ativação Plaquetária/efeitos dos fármacos , Desnaturação Proteica , Receptores de IgG/sangue , Trombina/farmacologia , Trombose/sangue
9.
Mucosal Immunol ; 17(2): 182-200, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38246240

RESUMO

The fungus Candida albicans can cause mucosal infections including oropharyngeal candidiasis (OPC) in immunocompromised patients. In humans, an increased risk of fungal infections correlates with thrombocytopenia. However, our understanding of platelets and megakaryocytes (Mks) in mucosal fungal infections is almost entirely unknown. When megakaryocyte- and platelet-depleted mice were infected with OPC, the tongue showed higher fungal burden, due to decreased neutrophil accumulation. Protection depended on a distinct population of oral-resident Mks. Interleukin-17, important in antifungal immunity, was required since mice lacking the IL-17 receptor had decreased circulating platelets and their oral Mks did not expand during OPC. The secretion of the peptide toxin candidalysin activated human Mks to release platelets with antifungal capacity. Infection with a candidalysin-deficient strain resulted in decreased expansion of tongue Mks during OPC. This is the first time that a distinct megakaryocyte population was identified in the oral mucosa which is critical for immunity against fungal infection.


Assuntos
Candidíase Bucal , Doenças Transmissíveis , Proteínas Fúngicas , Micoses , Humanos , Camundongos , Animais , Candida albicans , Megacariócitos , Interleucina-17 , Antifúngicos , Candidíase Bucal/microbiologia , Mucosa Bucal
10.
Inflamm Res ; 62(1): 37-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22945762

RESUMO

OBJECTIVE: To determine if receptor localization into lipid rafts, or the lipid rafts themselves, are important for FcγRIIa effector functions. MATERIAL: Wild-type FcγRIIa or mutant FcγRIIa(C208A) that does not translocate to lipid rafts were transfected into Chinese hamster ovary (CHO) cells which have been shown to be reliable cells for studying FcγR function. TREATMENT: Cells were treated with buffer or methyl-ß-cyclodextrin (MßCD) to deplete cholesterol and dissolve the structure of lipid rafts. METHODS: To evaluate lipid raft association, transfected CHO cells were lysed and centrifuged over a sucrose gradient. Fractions were run on SDS-PAGE and blotted for FcγRIIa or sphingolipid GM1 to illustrate the lipid raft fractions. Lateral mobility of GFP-tagged wild-type or mutant FcγRIIa was assessed using fluorescence recovery after photobleaching (FRAP) microscopy. Internalization of IgG-opsonized erythrocytes was assessed by fluorescence microscopy and uptake of heat-aggregated IgG (haIgG) was measured using flow cytometry. RESULTS: We observed that FcγRIIa(C208A) did not localize into lipid rafts. However, the mutant FcγRIIa retained lateral mobility and effector function similar to wild-type FcγRIIa. However, mutant FcγRIIa function was abolished upon treatment with MßCD. CONCLUSIONS: Lipid rafts provide an essential component required for effector activities independent of receptor localization.


Assuntos
Microdomínios da Membrana/fisiologia , Receptores de IgG/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Difusão , Humanos , beta-Ciclodextrinas/farmacologia
11.
Neuropsychopharmacol Rep ; 41(3): 325-335, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34254465

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) have anti-inflammatory properties that may have clinical utility in treating severe pulmonary manifestations of COVID-19. SSRIs exert anti-inflammatory effects at three mechanistic levels: (a) inhibition of proinflammatory transcription factor activity, including NF-κB and STAT3; (b) downregulation of lung tissue damage and proinflammatory cell recruitment via inhibition of cytokines, including IL-6, IL-8, TNF-α, and IL-1ß; and (c) direct suppression inflammatory cells, including T cells, macrophages, and platelets. These pathways are implicated in the pathogenesis of COVID-19. In this review, we will compare the pathogenesis of lung inflammation in pulmonary diseases including COVID-19, ARDS, and chronic obstructive pulmonary disease (COPD), describe the anti-inflammatory properties of SSRIs, and discuss the applications of SSRIS in treating COVID-19-associated inflammatory lung disease.


Assuntos
Anti-Inflamatórios/uso terapêutico , COVID-19/complicações , Pneumonia/tratamento farmacológico , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Humanos , Pneumonia/virologia , SARS-CoV-2
12.
Cell Immunol ; 263(1): 129-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20378102

RESUMO

Platelets are a crucial element in maintenance of hemostasis. Other functions attributable to platelets are now being appreciated such as their role in inflammatory reactions and vascular remodeling. Platelets have been reported to bind immunological stimuli like IgG-complexes and the understanding that platelets may participate in immunological reactions has been speculated for nearly 50years. In previous observations, we demonstrated that platelets could bind and internalize aggregated IgG-complexes without inducing platelet aggregation or granule release. To characterize this observation further, we tested the hypothesis that aggregated IgG-complexes do not activate platelets. To this end, platelets were stimulated with IgG-complexes or thrombin as a positive control and evaluated for activation by aggregation, expression of surface markers and production of cytokines. Activation with thrombin resulted in aggregation, expression of high levels of CD62P (P-selectin) expression and activation of the fibrinogen receptor, alpha(IIb)beta(3). Furthermore, stimulation with thrombin resulted in significant amounts of sCD40L (CD154) and RANTES (CCL5). However, platelets stimulated with IgG-complexes resulted in no aggregation and low levels of CD62P expression. Surprisingly, platelets stimulated with aggregated IgG-complexes released similar amounts of sCD40L and RANTES as platelets activated by thrombin. These data suggest that platelets are capable of secreting inflammatory molecules in response to IgG-complexes.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Plaquetas/metabolismo , Ligante de CD40/biossíntese , Quimiocina CCL5/biossíntese , Ativação Plaquetária/imunologia , Complexo Antígeno-Anticorpo/imunologia , Biomarcadores/metabolismo , Plaquetas/imunologia , Plaquetas/patologia , Ligante de CD40/genética , Ligante de CD40/imunologia , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Citocinas/metabolismo , Humanos , Inflamação , Selectina-P/biossíntese , Selectina-P/genética , Selectina-P/imunologia , Receptores de Fibrinogênio/biossíntese , Receptores de Fibrinogênio/genética , Receptores de Fibrinogênio/imunologia , Trombina/imunologia , Trombina/metabolismo
13.
Cell Immunol ; 265(2): 111-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20728077

RESUMO

Immunoglobulin G (IgG) dependent activities are important in host defense and autoimmune diseases. Various cell types including macrophages and neutrophils contribute to pathogen destruction and tissue damage through binding of IgG to Fcγ receptors (FcγR). One member of this family, FcγRIIA, is a transmembrane glycoprotein known to mediate binding and internalization of IgG-containing targets. FcγRIIA has been observed to translocate into lipids rafts upon binding IgG-containing targets. We hypothesize that lipid rafts participate to different extents in binding and internalizing targets of different sizes. We demonstrate that disruption of lipid rafts with 8mM methyl-ß-cyclodextrin (MßCD) nearly abolishes binding (91% reduction) and phagocytosis (60% reduction) of large IgG-coated targets. Conversely, binding and internalization of small IgG-complexes is less dependent on lipid rafts (49% and 17% inhibition at 8mM MßCD, respectively). These observations suggest that differences between phagocytosis and endocytosis may arise as early as the initial stages of ligand recognition.


Assuntos
Microdomínios da Membrana/imunologia , Fagocitose/imunologia , Receptores de IgG/imunologia , Animais , Células CHO , Cricetinae , Cricetulus , Endocitose/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Ligantes , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Ligação Proteica , Receptores de IgG/metabolismo , beta-Ciclodextrinas/farmacologia
14.
PLoS One ; 15(10): e0240667, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33057410

RESUMO

Medical student wellness is of great concern in the health care field. A growing number of studies point to increases in suicide, depression, anxiety, mood disorders, and burnout related to physician lifestyles. Mental health issues commencing in medical school have been suggested to have a significant impact on future physician lifestyle and burnout. Tracking the mental health of medical students at the University of Toledo College of Medicine and Life Sciences (UTCOMLS) with standardized indices will help elucidate triggers of poor mental health. Anonymous surveys were developed and distributed to preclinical medical students at five strategic time points throughout the 2018 2019 academic year. Surveys collected basic demographic information as well as inventories measuring perceived stress, burnout, resilience, and mindfulness. 172 M1s (83 males and 89 females) were included in the study and average response rate for the first 4 (out of 5) surveys averaged 74.8%. M1 males and females had on average increased personal burnout over time with females consistently scoring higher. Both males and females had an increase in stress from August to each subsequent month (p<0.05). Females reported a higher level of perceived stress than males in the beginning and middle of the academic year (p<0.05). Both males and females report a gradual decrease in resiliency throughout the academic year. These surveys demonstrated over half of males and females in medical school reported higher perceived stress scores than their gender-matched peers in the general United States population. Our study strengthens documented trends in resiliency, perceived stress, and burnout amongst medical students. More study in designing targeted approaches to ameliorate these findings in the medical student population is warranted.


Assuntos
Esgotamento Profissional/psicologia , Resiliência Psicológica , Estresse Psicológico/psicologia , Estudantes de Medicina/psicologia , Feminino , Humanos , Masculino , Adulto Jovem
15.
Biology (Basel) ; 9(10)2020 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-33092021

RESUMO

Platelets have long been recognized for their role in maintaining the balance between hemostasis and thrombosis. While their contributions to blood clotting have been well established, it has been increasingly evident that their roles extend to both innate and adaptive immune functions during infection and inflammation. In this comprehensive review, we describe the various ways in which platelets interact with different microbes and elicit immune responses either directly, or through modulation of leukocyte behaviors.

16.
PLoS One ; 15(8): e0236966, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32776968

RESUMO

Platelet-leukocyte aggregates (PLAs) are associated with increased thrombosis risk. The influence of PLA formation is especially important for cancer patients, since thrombosis accounts for approximately 10% of cancer-associated deaths. Our objective was to characterize and quantify PLAs in whole blood samples from lung cancer patients compared to healthy volunteers with the intent to analyze PLA formation in the context of lung cancer-associated thrombosis. Consenting lung cancer patients (57) and healthy volunteers (56) were enrolled at the Dana Cancer Center at the University of Toledo Health Science Campus. Peripheral blood samples were analyzed by flow cytometry. Patient medical history was reviewed through electronic medical records. Most importantly, we found lung cancer patients to have higher percentages of platelet-T cell aggregates (PTCAs) than healthy volunteers among both CD4+ T lymphocyte and CD8+ T lymphocyte populations. Our findings demonstrate that characterization of PTCAs may have clinical utility in differentiating lung cancer patients from healthy volunteers and stratifying lung cancer patients by history of thrombosis.


Assuntos
Plaquetas/patologia , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/complicações , Linfócitos T/patologia , Trombose/sangue , Trombose/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Estudos de Casos e Controles , Agregação Celular , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Adulto Jovem
17.
Res Pract Thromb Haemost ; 3(4): 704-712, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31624790

RESUMO

BACKGROUND: Platelets are widely recognized for their role in maintaining hemostasis. Recently, platelets have become appreciated for their varying roles in immunity, neuroprotection, and other physiological processes. While there are currently excellent methods to transiently deplete platelets and models of thrombocytopenia, studying the roles of platelets in chronic processes can be challenging. OBJECTIVE: Phenotypic characterization of the PF4-DTR mouse model of conditional platelet depletion compared to antibody depletion. METHODS: We describe the ability of the PF4-DTR mouse to maintain chronic platelet depletion, along with examining the bleeding phenotype compared to antibody-mediated platelet depletion. RESULTS: Systemic administration of diphtheria toxin resulted in >99% platelet depletion that can be maintained for >2 weeks. When compared to an antibody depletion model, PF4-DTR mice showed similar phenotypes when challenged with tail bleed and saphenous vein measurements of hemostasis. Mice depleted with diphtheria toxin were also able to undergo adoptive transfer of platelets. If the frequency and amount of diphtheria toxin is reduced, mice can be maintained at >40% depletion for >28 days, showing that this model is tunable. CONCLUSIONS: When compared to the gold standard of antibody-mediated depletion, PF4-DTR mice showed similar phenotypes and should be considered an important tool for examining the impact of thrombocytopenia over longer periods of time.

18.
J Leukoc Biol ; 80(6): 1553-62, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16921024

RESUMO

Fc gamma receptors (FcgammaRs) contribute to the internalization of large and small immune complexes through phagocytosis and endocytosis, respectively. The molecular processes underlying these internalization mechanisms differ dramatically and have distinct outcomes in immune clearance and modulation of cell function. However, it is unclear how the same receptors (FcgammaR) binding to identical ligands (IgG) can elicit such distinct responses. We and others have shown that Syk kinase, Src-related tyrosine kinases (SRTKs) and phosphatidyl inositol 3-kinases (PI3K) play important roles in FcgammaR phagocytosis. Herein, we demonstrate that these kinases are not required for FcgammaR endocytosis. Endocytosis of heat-aggregated IgG (HA-IgG) by COS-1 cells stably transfected with FcgammaRIIA or chimeric FcgammaRI-gamma-gamma (EC-TM-CYT) was not significantly altered by PP2, piceatannol, or wortmannin. In contrast, phagocytosis of large opsonized particles (IgG-sensitized sheep erythrocytes, EA) was markedly reduced by these inhibitors. These results were confirmed in primary mouse bone marrow-derived macrophages and freshly isolated human monocytes. Levels of receptor phosphorylation were similar when FcgammaRIIA was cross-linked using HA-IgG or EA. However, inhibition of FcgammaR phosphorylation prevented only FcgammaR phagocytosis. Finally, biochemical analyses of PI3K(p85)-Syk binding indicated that direct interactions between native Syk and PI3K proteins are differentially regulated during FcgammaR phagocytosis and endocytosis. Overall, our results indicate that FcgammaR endocytosis and phagocytosis differ dramatically in their requirement for Syk, SRTKs, and PI3K, pointing to striking differences in their signal transduction mechanisms. We propose a competitive inhibition-based model in which PI3K and c-Cbl play contrasting roles in the induction of phagocytosis or endocytosis signaling cascades.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Tirosina Quinases/imunologia , Receptores de IgG/imunologia , Transdução de Sinais/imunologia , Animais , Células da Medula Óssea/imunologia , Células COS , Chlorocebus aethiops , Eritrócitos/imunologia , Humanos , Imunoglobulina G/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Modelos Imunológicos , Fagocitose/genética , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-cbl/imunologia , Receptores de IgG/genética , Ovinos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Quinase Syk
19.
Exp Hematol ; 34(11): 1490-5, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17046568

RESUMO

OBJECTIVE: The physiologic role of platelet FcgammaRIIA, the only Fc receptor for IgG on human platelets, is largely unknown. FcgammaRIIA is also expressed on phagocytes such as monocytes and neutrophils, where it mediates the binding and internalization of both soluble IgG-containing complexes and IgG-coated cells. We previously reported the creation and characterization of a transgenic mouse that expresses human FcgammaRIIA on platelets and macrophages at levels comparable to that seen in humans. Using the transgenic mouse model, we observed that FcgammaRIIA mediates the clearance of IgG-coated cells. With the hypothesis that FcgammaRIIA on platelets may serve to remove IgG complexes from the circulation, we studied the capacity of human platelet FcgammaRIIA to bind and internalize such complexes. METHODS: We demonstrated by flow cytometry and electron microscopy that human platelets at 37 degrees C can bind and endocytose IgG complexes. We also utilized platelets from FcgammaRIIA transgenic mice to study endocytosis of IgG complexes by platelet FcgammaRIIA. RESULTS: Wild-type mouse platelets do not express Fcgamma receptors. While platelets from wild-type mice did not bind or endocytose IgG complexes, the presence of transgenic FcgammaRIIA on mouse platelets allowed the platelets to bind and endocytose IgG complexes. CONCLUSION: Our data indicate that platelet FcgammaRIIA binds and internalizes IgG complexes and suggest that human platelets may function to clear soluble IgG complexes from the circulation.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Antígenos CD/fisiologia , Plaquetas/metabolismo , Imunoglobulina G/fisiologia , Receptores de IgG/fisiologia , Animais , Antígenos CD/sangue , Endocitose/fisiologia , Citometria de Fluxo , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Ligação Proteica , Receptores de IgG/sangue
20.
Front Cell Dev Biol ; 4: 147, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28105409

RESUMO

Platelets are critical to hemostatic and immunological function, and are key players in cancer progression, metastasis, and cancer-related thrombosis. Platelets interact with immune cells to stimulate anti-tumor responses and can be activated by immune cells and tumor cells. Platelet activation can lead to complex interactions between platelets and tumor cells. Platelets facilitate cancer progression and metastasis by: (1) forming aggregates with tumor cells; (2) inducing tumor growth, epithelial-mesenchymal transition, and invasion; (3) shielding circulating tumor cells from immune surveillance and killing; (4) facilitating tethering and arrest of circulating tumor cells; and (5) promoting angiogenesis and tumor cell establishment at distant sites. Tumor cell-activated platelets also predispose cancer patients to thrombotic events. Tumor cells and tumor-derived microparticles lead to thrombosis by secreting procoagulant factors, resulting in platelet activation and clotting. Platelets play a critical role in cancer progression and thrombosis, and markers of platelet-tumor cell interaction are candidates as biomarkers for cancer progression and thrombosis risk.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA