Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Immunity ; 51(1): 131-140.e5, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31315031

RESUMEN

Macrophages play an important role in structural cardiac remodeling and the transition to heart failure following myocardial infarction (MI). Previous research has focused on the impact of blood-derived monocytes on cardiac repair. Here we examined the contribution of resident cavity macrophages located in the pericardial space adjacent to the site of injury. We found that disruption of the pericardial cavity accelerated maladaptive post-MI cardiac remodeling. Gata6+ macrophages in mouse pericardial fluid contributed to the reparative immune response. Following experimental MI, these macrophages invaded the epicardium and lost Gata6 expression but continued to perform anti-fibrotic functions. Loss of this specialized macrophage population enhanced interstitial fibrosis after ischemic injury. Gata6+ macrophages were present in human pericardial fluid, supporting the notion that this reparative function is relevant in human disease. Our findings uncover an immune cardioprotective role for the pericardial tissue compartment and argue for the reevaluation of surgical procedures that remove the pericardium.


Asunto(s)
Fibrosis/prevención & control , Factor de Transcripción GATA6/metabolismo , Corazón/fisiología , Macrófagos/inmunología , Infarto del Miocardio/inmunología , Miocardio/patología , Pericardio/inmunología , Animales , Movimiento Celular , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Remodelación Ventricular
2.
Curr Opin Hematol ; 31(1): 6-15, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37905750

RESUMEN

PURPOSE OF REVIEW: Activated or aged platelets are removed from circulation under (patho)physiologic conditions, the exact mechanism of platelet clearance under such conditions remains unclear and are currently being investigated. This review focuses on recent findings and controversies regarding platelet clearance and the disruption of platelet life cycle. RECENT FINDINGS: The platelet life span is determined by glycosylation of platelet surface receptors with sialic acid. Recently, it was shown that platelet activation and granule release leads to desialylation of glycans and accelerated clearance of platelets under pathological conditions. This phenomenon was demonstrated to be a main reason for thrombocytopenia being a complication in several infections and immune disorders. SUMMARY: Although we have recently gained some insight into how aged platelets are cleared from circulation, we are still not seeing the full picture. Further investigations of the platelet clearance pathways under pathophysiologic conditions are needed as well as studies to unravel the connection between platelet clearance and platelet production.


Asunto(s)
Plaquetas , Senescencia Celular , Citofagocitosis , Anciano , Humanos , Plaquetas/metabolismo , Plaquetas/fisiología , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos , Trombocitopenia/etiología , Trombocitopenia/metabolismo , Trombocitopenia/fisiopatología , Senescencia Celular/fisiología , Citofagocitosis/fisiología
3.
Blood ; 137(10): 1392-1405, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32932519

RESUMEN

Polyphosphate is a procoagulant inorganic polymer of linear-linked orthophosphate residues. Multiple investigations have established the importance of platelet polyphosphate in blood coagulation; however, the mechanistic details of polyphosphate homeostasis in mammalian species remain largely undefined. In this study, xenotropic and polytropic retrovirus receptor 1 (XPR1) regulated polyphosphate in platelets and was implicated in thrombosis in vivo. We used bioinformatic analyses of omics data to identify XPR1 as a major phosphate transporter in platelets. XPR1 messenger RNA and protein expression inversely correlated with intracellular polyphosphate content and release. Pharmacological interference with XPR1 activity increased polyphosphate stores, led to enhanced platelet-driven coagulation, and amplified thrombus formation under flow via the polyphosphate/factor XII pathway. Conditional gene deletion of Xpr1 in platelets resulted in polyphosphate accumulation, accelerated arterial thrombosis, and augmented activated platelet-driven pulmonary embolism without increasing bleeding in mice. These data identify platelet XPR1 as an integral regulator of platelet polyphosphate metabolism and reveal a fundamental role for phosphate homeostasis in thrombosis.


Asunto(s)
Plaquetas/metabolismo , Polifosfatos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virales/metabolismo , Trombosis/metabolismo , Animales , Transporte Biológico , Coagulación Sanguínea , Factor XII/metabolismo , Femenino , Masculino , Ratones , Trombosis/sangre , Receptor de Retrovirus Xenotrópico y Politrópico
4.
Hepatology ; 73(5): 1967-1984, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32761929

RESUMEN

BACKGROUND AND AIMS: Kupffer cells (KCs) are the resident intravascular phagocyte population of the liver and critical to the capture and killing of bacteria. Calcineurin/nuclear factor of activated T cells (NFAT) inhibitors (CNIs) such as tacrolimus are used to prevent rejection in solid organ transplant recipients. Although their effect on lymphocytes has been studied extensively, there are limited experimental data about if and how CNIs shape innate immunity, and whether this contributes to the higher rates of infection observed in patients taking CNIs. APPROACH AND RESULTS: Here, we investigated the impact of tacrolimus treatment on innate immunity and, more specifically, on the capability of Kupffer cells (KCs) to fight infections. Retrospective analysis of data of >2,700 liver transplant recipients showed that taking calcineurin inhibitors such as tacrolimus significantly increased the likelihood of Staphylococcus aureus infection. Using a mouse model of acute methicillin-resistant S. aureus (MRSA) bacteremia, most bacteria were sequestered in the liver and we found that bacteria were more likely to disseminate and kill the host in tacrolimus-treated mice. Using imaging, we unveiled the mechanism underlying this observation: the reduced capability of KCs to capture, phagocytose, and destroy bacteria in tacrolimus-treated animals. Furthermore, in a gene expression analysis of infected KCs, the triggering receptor expressed on myeloid cells 1 (TREM1) pathway was the one with the most significant down-regulation after tacrolimus treatment. TREM1 inhibition likewise inhibited KC bacteria capture. TREM1 levels on neutrophils as well as the overall neutrophil response after infection were unaffected by tacrolimus treatment. CONCLUSIONS: Our results indicate that tacrolimus treatment has a significant impact directly on KCs and on TREM1, thereby compromising their capacity to fend off infections.


Asunto(s)
Bacteriemia/etiología , Inmunosupresores/efectos adversos , Macrófagos del Hígado/efectos de los fármacos , Trasplante de Órganos/efectos adversos , Infecciones Estafilocócicas/etiología , Tacrolimus/efectos adversos , Animales , Femenino , Citometría de Flujo , Humanos , Inmunosupresores/uso terapéutico , Macrófagos del Hígado/fisiología , Masculino , Staphylococcus aureus Resistente a Meticilina , Ratones , Persona de Mediana Edad , Trasplante de Órganos/métodos , Fagocitosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Estudios Retrospectivos , Tacrolimus/uso terapéutico
5.
Int J Mol Sci ; 23(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35409226

RESUMEN

There is accumulating evidence that platelets play roles beyond their traditional functions in thrombosis and hemostasis, e.g., in inflammatory processes, infection and cancer, and that they interact, stimulate and regulate cells of the innate immune system such as neutrophils, monocytes and macrophages. In this review, we will focus on platelet activation in hemostatic and inflammatory processes, as well as platelet interactions with neutrophils and monocytes/macrophages. We take a closer look at the contributions of major platelet receptors GPIb, αIIbß3, TLT-1, CLEC-2 and Toll-like receptors (TLRs) as well as secretions from platelet granules on platelet-neutrophil aggregate and neutrophil extracellular trap (NET) formation in atherosclerosis, transfusion-related acute lung injury (TRALI) and COVID-19. Further, we will address platelet-monocyte and macrophage interactions during cancer metastasis, infection, sepsis and platelet clearance.


Asunto(s)
COVID-19 , Trombosis , Plaquetas/patología , Hemostasis , Humanos , Inmunidad Innata , Inflamación/patología , Neutrófilos/patología , Activación Plaquetaria , Tromboinflamación , Trombosis/patología
6.
Platelets ; 32(3): 314-324, 2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32896192

RESUMEN

Platelets are increasingly being recognized for playing roles beyond thrombosis and hemostasis. Today we know that they mediate inflammation by direct interactions with innate immune cells or secretion of cytokines/chemokines. Here we review their interactions with neutrophils and monocytes/macrophages in infection and sepsis, stroke, myocardial infarction and venous thromboembolism. We discuss new roles for platelet surface receptors like GPVI or GPIb and also look at platelet contributions to the formation of neutrophil extracellular traps (NETs) as well as to deep vein thrombosis during infection, e.g. in COVID-19 patients.


Asunto(s)
Plaquetas/inmunología , COVID-19/inmunología , Infarto del Miocardio/inmunología , Neutrófilos/inmunología , Sepsis/inmunología , Accidente Cerebrovascular/inmunología , Tromboembolia Venosa/inmunología , Plaquetas/patología , COVID-19/genética , COVID-19/patología , COVID-19/virología , Comunicación Celular/genética , Comunicación Celular/inmunología , Citocinas/genética , Citocinas/inmunología , Trampas Extracelulares/genética , Trampas Extracelulares/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación , Macrófagos/inmunología , Macrófagos/patología , Monocitos/inmunología , Monocitos/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Neutrófilos/patología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/inmunología , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/inmunología , Sepsis/genética , Sepsis/patología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Tromboembolia Venosa/genética , Tromboembolia Venosa/patología
7.
Semin Immunol ; 28(6): 536-545, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27769639

RESUMEN

The primary function of platelets is to patrol the vasculature and seal vessel breaches to limit blood loss. However, it is becoming increasingly clear that they also contribute to pathophysiological conditions like thrombosis, atherosclerosis, stroke and infection. Severe sepsis is a devastating disease that claims hundreds of thousands of lives every year in North America and is a major burden to the public health system. Platelet surface receptors like GPIb, αIIbß3, TLR2 and TLR4 are involved in direct platelet-bacteria interactions. Plasma proteins like fibrinogen and vWF enable indirect interactions. Furthermore, platelet granules contain a plethora of proteins that modulate the immune response as well as microbicidal agents which can directly lyse bacteria. Bacterial toxins are potent platelet activators and can cause intravascular platelet aggregation. Platelets contribute to the antibacterial response of the host involving Kupffer cells, neutrophils and the complement system. In this review we summarize the current knowledge about platelet-bacteria interactions and highlight recent advances in the field.


Asunto(s)
Plaquetas/inmunología , Plaquetas/metabolismo , Infecciones/etiología , Infecciones/metabolismo , Animales , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Comunicación Celular , Gránulos Citoplasmáticos/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Activación Plaquetaria/inmunología , Glicoproteínas de Membrana Plaquetaria/inmunología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Sepsis/etiología , Sepsis/metabolismo , Transducción de Señal
8.
Blood ; 129(12): 1702-1706, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28077416

RESUMEN

Platelets maintain hemostasis after injury, but also during inflammation. Recent studies have shown that platelets prevent inflammatory bleeding through (hem) immunoreceptor tyrosine-based activation motif-dependent mechanisms irrespective of aggregation during skin and lung inflammation. Although the exact mechanisms underlying this process remain unknown, it was speculated that mediators released from platelet granules might be involved. Maintaining cerebral hemostasis during stroke treatment is of high clinical relevance because hemorrhage may aggravate the disease state and increase mortality. Although it was shown that platelets help maintain hemostasis in the ischemic brain, their exact contribution remains ill defined. Here we show that Unc13d-/- /Nbeal2-/- mice, which lack platelet α- and dense-granule secretion, show no signs of hemorrhage in models of skin or lung inflammation. In stark contrast, lack of platelet granule release resulted in impaired hemostasis in the ischemic brain after transient middle cerebral artery occlusion leading to increased intracranial hemorrhage and mortality. Our results reveal for the first time that platelet granule constituents are essential for maintenance of hemostasis during thrombo-inflammatory brain infarction but not experimental inflammation of the skin or lung, thereby uncovering vascular bed-specific differences in the prevention of inflammatory bleeding.


Asunto(s)
Plaquetas/metabolismo , Isquemia Encefálica/patología , Hemorragia Cerebral/prevención & control , Animales , Isquemia Encefálica/sangre , Hemorragia Cerebral/sangre , Hemorragia/patología , Hemostasis , Infarto de la Arteria Cerebral Media , Inflamación/sangre , Inflamación/patología , Pulmón/patología , Ratones , Vesículas Secretoras/fisiología , Piel/patología
9.
J Immunol ; 199(8): 2948-2957, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28887433

RESUMEN

The neurobeachin-like 2 protein (Nbeal2) belongs to the family of beige and Chediak-Higashi (BEACH) domain proteins. Loss-of-function mutations in the human NBEAL2 gene or Nbeal2 deficiency in mice cause gray platelet syndrome, a bleeding disorder characterized by macrothrombocytopenia, splenomegaly, and paucity of α-granules in megakaryocytes and platelets. We found that in mast cells, Nbeal2 regulates the activation of the Shp1-STAT5 signaling axis and the composition of the c-Kit/STAT signalosome. Furthermore, Nbeal2 mediates granule formation and restricts the expression of the transcription factors, IRF8, GATA2, and MITF as well as of the cell-cycle inhibitor p27, which are essential for mast cell differentiation, proliferation, and cytokine production. These data demonstrate the relevance of Nbeal2 in mast cells above and beyond granule biosynthesis.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Síndrome de Plaquetas Grises/genética , Mastocitos/fisiología , Megacariocitos/fisiología , Animales , Proteínas Sanguíneas/genética , Ciclo Celular , Células Cultivadas , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Hemorragia , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ratones , Ratones Noqueados , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Esplenomegalia , Trombocitopenia
10.
Platelets ; 29(6): 549-555, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29446689

RESUMEN

Platelets patrol the vasculature and adhere at sites of vascular damage after trauma to limit blood loss. In recent years, however, it has become clear that platelets also contribute to pathophysiologic processes such as thrombosis, atherosclerosis, stroke, sepsis and many more. An exciting new role for them is in non-classical hemostasis to prevent bleeding in the inflamed vasculature. Recent studies suggest that GPVI, CLEC-2, integrin αIIbß3 (GPIIb/IIIa), and the content of platelet α- and dense granules are important players in this process. This review summarizes the current knowledge about how platelets prevent vascular integrity during inflammation in the skin, lung, and the ischemic brain and their organ-specific role.


Asunto(s)
Plaquetas/inmunología , Hemorragia/etiología , Hemorragia/patología , Humanos , Transducción de Señal
11.
Blood ; 125(24): 3769-77, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25908104

RESUMEN

Mice with a constitutive or platelet-specific deletion of the C-type-lectin-like receptor (CLEC-2) exhibit hemorrhaging in the brain at mid-gestation. We sought to investigate the basis of this defect, hypothesizing that it is mediated by the loss of CLEC-2 activation by its endogenous ligand, podoplanin, which is expressed on the developing neural tube. To induce deletion of podoplanin at the 2-cell stage, we generated a podoplanin(fl/fl) mouse crossed to a PGK-Cre mouse. Using 3-dimensional light-sheet microscopy, we observed cerebral vessels were tortuous and aberrantly patterned at embryonic (E) day 10.5 in podoplanin- and CLEC-2-deficient mice, preceding the formation of large hemorrhages throughout the fore-, mid-, and hindbrain by E11.5. Immunofluorescence and electron microscopy revealed defective pericyte recruitment and misconnections between the endothelium of developing blood vessels and surrounding pericytes and neuro-epithelial cells. Nestin-Cre-driven deletion of podoplanin on neural progenitors also caused widespread cerebral hemorrhaging. Hemorrhaging was also seen in the ventricles of embryos deficient in the platelet integrin subunit glycoprotein IIb or in embryos in which platelet α-granule and dense granule secretion is abolished. We propose a novel role for podoplanin on the neuro-epithelium, which interacts with CLEC-2 on platelets, mediating platelet adhesion, aggregation, and secretion to guide the maturation and integrity of the developing vasculature and prevent hemorrhage.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/embriología , Circulación Cerebrovascular , Lectinas Tipo C/genética , Glicoproteínas de Membrana/genética , Animales , Plaquetas/metabolismo , Tipificación del Cuerpo , Encéfalo/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Hemorragias Intracraneales/genética , Hemorragias Intracraneales/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Activación Plaquetaria , Agregación Plaquetaria , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 36(4): 629-35, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26916731

RESUMEN

OBJECTIVE: The objective of this study was to investigate the effects of platelet inhibition on myocardial ischemia-reperfusion (IR) injury. APPROACH AND RESULTS: Timely restoration of coronary blood flow after myocardial infarction is indispensable but leads to additional damage to the heart (myocardial IR injury). Microvascular dysfunction contributes to myocardial IR injury. We hypothesized that platelet activation during IR determines microvascular perfusion and thereby the infarct size in the reperfused myocardium. The 3 phases of thrombus formation were analyzed by targeting individual key platelet-surface molecules with monoclonal antibody derivatives: (1) adhesion (anti-glycoprotein [GP]-Ib), (2) activation (anti-GPVI), and (3) aggregation (anti-GPIIbIIIa) in a murine in vivo model of left coronary artery ligation (30 minutes of ischemia followed by 24 hours of reperfusion). Infarct sizes were determined by Evans Blue/2,3,5-triphenyltetrazolium chloride staining, infiltrating neutrophils by immunohistology. Anti-GPVI treatment significantly reduced infarct size versus control, whereas anti-GPIb or anti-GPIIbIIIa antibody fragments showed no significant differences. Mechanistically, anti-GPVI antibody-mediated reduction of infarct size was not because of impaired Ca(2+) signaling or platelet degranulation because mice deficient in store-operated calcium channels (stromal interaction molecule 1, ORAI1), α-granules (Nbeal2(-/-)), and dense granule release (Unc13d(-/-)) had similar infarct sizes as control animals. Protective effects of anti-GPVI treatment were accompanied by improved microperfusion. Leukocyte infiltration was reduced in both anti-GPVI and anti-GPIb-treated IR mice. CONCLUSIONS: Inhibition of platelet activation by an anti-GPVI antibody, but not inhibition of platelet adhesion or aggregation by an anti-GPIb or anti-GPIIbIIIa antibody significantly reduces infarct size. The reduction of the infarct size is primarily based on an improved microperfusion after anti-GPVI antibody treatment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Plaquetas/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Animales , Plaquetas/inmunología , Plaquetas/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/efectos de los fármacos , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Glicoproteínas de Membrana Plaquetaria/inmunología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Factores de Tiempo
13.
Res Pract Thromb Haemost ; 8(4): 102432, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38854821

RESUMEN

Here, we present a series of illustrated capsules from the State of the Art (SOA) speakers at the 2024 International Society on Thrombosis and Haemostasis Congress in Bangkok, Thailand. This year's Congress marks the first time that the International Society on Thrombosis and Haemostasis has held its flagship scientific meeting in Southeast Asia and is the first to be organized by an international Planning Committee. The Bangkok program will feature innovative science and clinical updates from around the world, reflecting the diversity and multidisciplinary growth of our field. In these illustrated SOA capsules, you will find an exploration of novel models of thrombosis and bleeding and biomaterial discoveries that can trigger or block coagulation. Thromboinflammation is now understood to drive many disease states, and the SOA speakers cover cellular and coagulation responses to COVID-19 and other infections. The theme of crosstalk between coagulation and inflammation expands with capsules on protein S signaling, complement, and fibrinolytic inhibitors. Novel agents for hemophilia and thrombosis prevention are introduced. Challenging clinical conditions are also covered, such as inherited platelet disorders and antiphospholipid antibody syndrome. The scientific program in Bangkok will also showcase the work of clinicians and scientists from all parts of the world and chronicle real-world challenges. For example, 2 SOA capsules address the diagnosis and management of von Willebrand disease in low-income settings. Take some time to browse through these short illustrated reviews; we're sure that you'll be entertained, educated, and inspired to further explore the world of thrombosis and hemostasis.

14.
Stroke ; 44(11): 3202-10, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029635

RESUMEN

BACKGROUND AND PURPOSE: Lymphocytes are important players in the pathophysiology of acute ischemic stroke. The interaction of lymphocytes with endothelial cells and platelets, termed thrombo-inflammation, fosters microvascular dysfunction and secondary infarct growth. FTY720, a sphingosine-1-phosphate receptor modulator, blocks the egress of lymphocytes from lymphoid organs and has been shown to reduce ischemic neurodegeneration; however, the underlying mechanisms are unclear. We investigated the mode of FTY720 action in models of cerebral ischemia. METHODS: Transient middle cerebral artery occlusion (tMCAO) was induced in wild-type and lymphocyte-deficient Rag1(-/-) mice treated with FTY720 (1 mg/kg) or vehicle immediately before reperfusion. Stroke outcome was assessed 24 hours later. Immune cells in the blood and brain were counted by flow cytometry. The integrity of the blood-brain barrier was analyzed using Evans Blue dye. Thrombus formation was determined by immunohistochemistry and Western blot, and was correlated with cerebral perfusion. RESULTS: FTY720 significantly reduced stroke size and improved functional outcome in wild-type mice on day 1 and day 3 after transient middle cerebral artery occlusion. This protective effect was lost in lymphocyte-deficient Rag1(-/-) mice and in cultured neurons subjected to hypoxia. Less lymphocytes were present in the cerebral vasculature of FTY720-treated wild-type mice, which in turn reduced thrombosis and increased cerebral perfusion. In contrast, FTY720 was unable to prevent blood-brain barrier breakdown and transendothelial immune cell trafficking after transient middle cerebral artery occlusion. CONCLUSIONS: Induction of lymphocytopenia and concomitant reduction of microvascular thrombosis are key modes of FTY720 action in stroke. In contrast, our findings in Rag1(-/-) mice and cultured neurons argue against direct neuroprotective effects of FTY720.


Asunto(s)
Isquemia Encefálica/terapia , Inmunosupresores/farmacología , Inflamación/terapia , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Accidente Cerebrovascular/terapia , Trombosis/terapia , Animales , Inhibidores Enzimáticos/farmacología , Clorhidrato de Fingolimod , Proteínas de Homeodominio/genética , Hipoxia , Linfocitos/efectos de los fármacos , Linfopenia/patología , Masculino , Ratones , Ratones Transgénicos , Arteria Cerebral Media/patología , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Esfingosina/farmacología
15.
Front Immunol ; 14: 1277808, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38116017

RESUMEN

During fibrosis, (myo)fibroblasts deposit large amounts of extracellular matrix proteins, thereby replacing healthy functional tissue. In liver fibrosis, this leads to the loss of hepatocyte function, portal hypertension, variceal bleeding, and increased susceptibility to infection. At an early stage, liver fibrosis is a dynamic and reversible process, however, from the cirrhotic stage, there is significant progression to hepatocellular carcinoma. Both liver-resident macrophages (Kupffer cells) and monocyte-derived macrophages are important drivers of fibrosis progression, but can also induce its regression once triggers of chronic inflammation are eliminated. In liver cancer, they are attracted to the tumor site to become tumor-associated macrophages (TAMs) polarized towards a M2- anti-inflammatory/tumor-promoting phenotype. Besides their role in thrombosis and hemostasis, platelets can also stimulate fibrosis and tumor development by secreting profibrogenic factors and regulating the innate immune response, e.g., by interacting with monocytes and macrophages. Here, we review recent literature on the role of macrophages and platelets and their interplay in liver fibrosis and hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular , Várices Esofágicas y Gástricas , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Várices Esofágicas y Gástricas/metabolismo , Várices Esofágicas y Gástricas/patología , Neoplasias Hepáticas/patología , Hemorragia Gastrointestinal , Cirrosis Hepática , Macrófagos , Fibrosis
16.
Hamostaseologie ; 43(5): 348-359, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37857297

RESUMEN

Intravital microscopy is a powerful tool to study thrombosis in real time. The kinetics of thrombus formation and progression in vivo is studied after inflicting damage to the endothelium through mechanical, chemical, or laser injury. Mouse models of atherosclerosis are also used to induce thrombus formation. Vessels of different sizes and from different vascular beds such as carotid artery or vena cava, mesenteric or cremaster arterioles, can be targeted. Using fluorescent dyes, antibodies, or reporter mouse strains allows to visualize key cells and factors mediating the thrombotic processes. Here, we review the latest literature on using intravital microscopy to study thrombosis as well as thromboinflammation following transient middle cerebral artery occlusion, infection-induced immunothrombosis, and liver ischemia reperfusion.


Asunto(s)
Trombosis , Animales , Ratones , Inflamación , Modelos Animales de Enfermedad , Microscopía Intravital , Endotelio
17.
EMBO Mol Med ; 15(9): e16858, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37490001

RESUMEN

Hyperreactive platelets are commonly observed in diabetic patients indicating a potential link between glucose homeostasis and platelet reactivity. This raises the possibility that platelets may play a role in the regulation of metabolism. Pancreatic ß cells are the central regulators of systemic glucose homeostasis. Here, we show that factor(s) derived from ß cells stimulate platelet activity and platelets selectively localize to the vascular endothelium of pancreatic islets. Both depletion of platelets and ablation of major platelet adhesion or activation pathways consistently resulted in impaired glucose tolerance and decreased circulating insulin levels. Furthermore, we found platelet-derived lipid classes to promote insulin secretion and identified 20-Hydroxyeicosatetraenoic acid (20-HETE) as the main factor promoting ß cells function. Finally, we demonstrate that the levels of platelet-derived 20-HETE decline with age and that this parallels with reduced impact of platelets on ß cell function. Our findings identify an unexpected function of platelets in the regulation of insulin secretion and glucose metabolism, which promotes metabolic fitness in young individuals.


Asunto(s)
Células Secretoras de Insulina , Humanos , Secreción de Insulina , Insulina/metabolismo , Plaquetas , Glucosa/metabolismo
18.
Nat Metab ; 5(7): 1174-1187, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37414930

RESUMEN

The gut microbiota influences intestinal barrier integrity through mechanisms that are incompletely understood. Here we show that the commensal microbiota weakens the intestinal barrier by suppressing epithelial neuropilin-1 (NRP1) and Hedgehog (Hh) signaling. Microbial colonization of germ-free mice dampens signaling of the intestinal Hh pathway through epithelial Toll-like receptor (TLR)-2, resulting in decreased epithelial NRP1 protein levels. Following activation via TLR2/TLR6, epithelial NRP1, a positive-feedback regulator of Hh signaling, is lysosomally degraded. Conversely, elevated epithelial NRP1 levels in germ-free mice are associated with a strengthened gut barrier. Functionally, intestinal epithelial cell-specific Nrp1 deficiency (Nrp1ΔIEC) results in decreased Hh pathway activity and a weakened gut barrier. In addition, Nrp1ΔIEC mice have a reduced density of capillary networks in their small intestinal villus structures. Collectively, our results reveal a role for the commensal microbiota and epithelial NRP1 signaling in the regulation of intestinal barrier function through postnatal control of Hh signaling.


Asunto(s)
Proteínas Hedgehog , Neuropilina-1 , Ratones , Animales , Neuropilina-1/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Células Epiteliales/metabolismo , Bacterias/metabolismo
19.
Front Cell Dev Biol ; 9: 623906, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33644061

RESUMEN

Leukocytes are among the most mobile and versatile cells that have many essential functions in homeostasis and survival. Especially cells from the innate immune system, i.e., neutrophils and macrophages, play an important role as rapid first responders against invading microorganisms. With the advent of novel imaging techniques, new ways of visualizing innate immune cells have become available in recent years, thereby enabling more and more detailed discoveries about their nature, function and interaction partners. Besides intravital spinning-disc and 2-photon microscopy, clearing and 3D-imaging techniques provide new insights into the mechanism of innate immune cell behavior in their natural environment. This mini review focuses on the contributions of novel-imaging techniques to provide insight into the functions of neutrophils and macrophages under homeostasis and in infections. Imaging setups for different organs like the liver, kidney, heart, lung, and the peritoneal cavity are discussed as well as the current limitations of these imaging techniques.

20.
Thromb Haemost ; 121(8): 1021-1030, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33307564

RESUMEN

Neutrophil extracellular traps (NETs) and polyphosphates (polyP) have been recognized as procoagulant polyanions. This review summarizes the activities and regulation of the two procoagulant mediators and compares their functions. NETs are composed of DNA which like polyP is built of phosphate units linked by high-energy phosphoanhydride bonds. Both NETs and polyP form insoluble particulate surfaces composed of a DNA/histone meshwork or Ca2+-rich nanoparticles, respectively. These polyanionic molecules modulate coagulation involving an array of mechanisms and trigger thrombosis via activation of the factor XII-driven procoagulant and proinflammatory contact pathway. Here, we outline the current knowledge on NETs and polyP with respect to their procoagulant and prothrombotic nature, strategies for interference of their activities in circulation, as well as the crosstalk between these two molecules. A better understanding of the underlying, cellular mechanisms will shed light on the therapeutic potential of targeting NETs and polyP in coagulation and thrombosis.


Asunto(s)
Coagulación Sanguínea , Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Polifosfatos/sangre , Trombosis/sangre , Animales , Coagulación Sanguínea/efectos de los fármacos , Trampas Extracelulares/efectos de los fármacos , Fibrinolíticos/uso terapéutico , Humanos , Neutrófilos/efectos de los fármacos , Polifosfatos/antagonistas & inhibidores , Transducción de Señal , Trombosis/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA