Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 28(13): e18457, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38963011

RESUMEN

Allogeneic haematopoietic stem cell transplantation (allo-HSCT) can potentially cure malignant blood disorders and benign conditions such as haemoglobinopathies and immunologic diseases. However, allo-HSCT is associated with significant complications. The most common and debilitating among them is graft-versus-host disease (GVHD). In GVHD, donor-derived T cells mount an alloimmune response against the recipient. The alloimmune response involves several steps, including recognition of recipient antigens, activation and proliferation of T cells in secondary lymphoid organs, and homing into GVHD-targeted organs. Adhesion molecules on T cells and endothelial cells mediate homing of T cells into lymphoid and non-lymphoid tissues. In this study, we showed that Von Willebrand factor (VWF), an adhesion molecule secreted by activated endothelial cells, plays an important role in mouse models of GVHD. We investigated the effect of the VWF-cleaving protease ADAMTS13 on GVHD. We found that ADAMTS13 reduced the severity of GVHD after bone marrow transplantation from C57BL6 donor to BALB/C recipient mice. A recombinant VWF-A2 domain peptide also reduced GVHD in mice. We showed that ADAMTS13 and recombinant VWF-A2 reduced the binding of T cells to endothelial cells and VWF in vitro, and reduced the number of T cells in lymph nodes, Peyer's patches and GVHD-targeted organs in vivo. We identified LFA-1 (αLß2) as the binding site of VWF on T cells. Our results showed that blocking T-cell homing by ADAMTS13 or VWF-A2 peptide reduced the severity of the GVHD after allo-HSCT, a potentially novel method for treating and preventing GVHD.


Asunto(s)
Proteína ADAMTS13 , Enfermedad Injerto contra Huésped , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T , Factor de von Willebrand , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/inmunología , Animales , Proteína ADAMTS13/metabolismo , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de von Willebrand/metabolismo , Humanos , Trasplante Homólogo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Modelos Animales de Enfermedad , Trasplante de Médula Ósea , Células Endoteliales/metabolismo
2.
Cancer Metastasis Rev ; 41(1): 147-172, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35022962

RESUMEN

We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.


Asunto(s)
COVID-19 , Neoplasias , Animales , Biología , Plaquetas/metabolismo , Hemostasis , Humanos , Mamíferos , Neoplasias/metabolismo , SARS-CoV-2
3.
Curr Opin Hematol ; 27(5): 341-352, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32701617

RESUMEN

PURPOSE OF REVIEW: To discuss the crosstalk between the complement system and hemostatic factors (coagulation cascade, platelet, endothelium, and Von Willebrand Factor), and the consequences of this interaction under physiologic and pathologic conditions. RECENT FINDINGS: The complement and coagulation systems are comprised of serine proteases and are genetically related. In addition to the common ancestral genes, the complement system and hemostasis interact directly, through protein-protein interactions, and indirectly, on the surface of platelets and endothelial cells. The close interaction between the complement system and hemostatic factors is manifested both in physiologic and pathologic conditions, such as in the inflammatory response to thrombosis, thrombosis at the inflamed area, and thrombotic complications of complement disorders. SUMMARY: The interaction between the complement system and hemostasis is vital for homeostasis and the protective response of the host to tissue injury, but also results in the pathogenesis of several thrombotic and inflammatory disorders.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Hemostasis/fisiología , Humanos
4.
Biol Blood Marrow Transplant ; 25(10): 1920-1924, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271886

RESUMEN

Acute graft-versus-host disease (GVHD) affects different organs, including the skin, liver, and gastrointestinal tract. Although kidneys are not among the organs commonly known to be the target of acute GVHD, kidney damage is frequently reported after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We have studied the effect of bone marrow transplantation (BMT) on the kidneys in different murine models of GVHD. We found that glomerular damage in the kidneys is a common pathological finding in mice after BMT. The histopathological features of glomeruli damage included mesengiolysis, mesangial proliferation and edema, subendothelial and endothelial thickening, splitting of capillary walls in glomeruli, narrowing and collapsing of capillary lumens, fibrinoid necrosis of afferent arterioles, intimal hyperplasia, and microthrombi. These pathological features are similar to those detected in kidneys of patients with thrombotic microangiopathy (TMA) after allo-HSCT. We previously showed that activation of the complement system plays a role in GVHD-induced tissue injury in mice. Here we report the presence of complement activation products in the kidney specimens of mice after BMT. We also report that complement deficiency reduced the extent and severity of post-BMT glomerular damage in mice. We conclude that BMT in mice is associated with glomerular injury and tubulointerstitial nephritis, and that kidney damage is at least partially mediated by activation of the complement system.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Riñón/lesiones , Acondicionamiento Pretrasplante/efectos adversos , Animales , Modelos Animales de Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Ratones , Acondicionamiento Pretrasplante/métodos
5.
Blood ; 130(10): 1235-1242, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28679740

RESUMEN

We investigated the effect of platelets on ovarian cancer and the role of adenosine diphosphate (ADP) receptors (P2Y12 and P2Y1) on platelets in the growth of primary ovarian cancer tumors. We showed that in murine models of ovarian cancer, a P2Y12 inhibitor (ticagrelor) reduced tumor growth by 60% compared with aspirin and by 75% compared with placebo. In P2Y12-/- mice, the growth of syngeneic ovarian cancer tumors was reduced by >85% compared with wild-type (WT) mice. In contrast, there was no difference in tumor growth between P2Y1-/- and WT mice. Reconstitution of hematopoiesis in irradiated P2Y12-/- mice by hematopoietic progenitor cells from WT mice (WT→P2Y12-/-) restored tumor growth in P2Y12-/- mice. Finally, knockdown of ecto-apyrase (CD39) on ovarian cancer cells increased tumor growth in tumor-bearing mice. Although in the absence of platelets, ADP, the P2Y12 inhibitor, recombinant apyrase, or knockdown of CD39 did not affect cancer cell proliferation, in the presence of platelets, the P2Y12 inhibitor and recombinant apyrase reduced and knockdown of CD39 increased platelet-enhanced cancer cell proliferation. These results suggest that P2Y12 on platelets and ADP concentration at the interface between cancer cells and platelets affect the growth of primary ovarian cancer tumors in mice. If additional studies in mice and in pilot human trials confirm our results, inhibition of P2Y12 might be a new therapeutic option that can be used in adjuvant to the traditional surgery and chemotherapy in patients with ovarian cancer.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Receptores Purinérgicos P2Y12/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apirasa/metabolismo , Plaquetas/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Hematopoyesis/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Neoplasias Ováricas/metabolismo , Ticagrelor
6.
Blood ; 137(25): 3462-3463, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34165547
7.
J Immunol ; 196(3): 1412-8, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718342

RESUMEN

We have previously shown that complement component 3 (C3) is secreted by malignant epithelial cells. To understand the mechanism of upregulation of C3 expression in tumor cells, we studied the C3 promoter and identified that twist basic helix-loop-helix transcription factor 1 (TWIST1) binds to the C3 promoter and enhances its expression. Because TWIST1 mediates epithelial-mesenchymal transition (EMT), we studied the effect of C3 on EMT and found that C3 decreased E-cadherin expression on cancer cells and promoted EMT. We showed that C3-induced reduction in E-cadherin expression in ovarian cancer cells was mediated by C3a and is Krüppel-like factor 5 dependent. We investigated the association between TWIST1 and C3 in malignant tumors and in murine embryos. TWIST1 and C3 colocalized at the invasive tumor edges, and in the neural crest and limb buds of mouse embryos. Our results identified TWIST1 as a transcription factor that regulates C3 expression during pathologic and physiologic EMT.


Asunto(s)
Complemento C3/biosíntesis , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias/patología , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Complemento C3/genética , Femenino , Humanos , Inmunohistoquímica , Ratones , Mutagénesis Sitio-Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Proteína 1 Relacionada con Twist/genética
8.
Proc Natl Acad Sci U S A ; 112(46): 14266-71, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578771

RESUMEN

Thrombocytosis and platelet hyperreactivity are known to be associated with malignancy; however, there have been no ultrastructure studies of platelets from patients with ovarian cancer. Here, we used electron cryotomography (cryo-ET) to examine frozen-hydrated platelets from patients with invasive ovarian cancer (n = 12) and control subjects either with benign adnexal mass (n = 5) or free from disease (n = 6). Qualitative inspections of the tomograms indicate significant morphological differences between the cancer and control platelets, including disruption of the microtubule marginal band. Quantitative analysis of subcellular features in 120 platelet electron tomograms from these two groups showed statistically significant differences in mitochondria, as well as microtubules. These structural variations in the platelets from the patients with cancer may be correlated with the altered platelet functions associated with malignancy. Cryo-ET of platelets shows potential as a noninvasive biomarker technology for ovarian cancer and other platelet-related diseases.


Asunto(s)
Biomarcadores de Tumor/sangre , Plaquetas/ultraestructura , Microscopía por Crioelectrón , Microtúbulos/ultraestructura , Mitocondrias/ultraestructura , Neoplasias Ováricas , Femenino , Humanos , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología
9.
Blood ; 125(6): 1034-7, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25395424

RESUMEN

Several complement proteins interact with hemostatic factors. We discovered that von Willebrand factor (VWF) acts as a cofactor for factor I-mediated cleavage of complement C3b, thereby shutting down complement activation. The complement regulatory function of VWF multimers depends on their size. Smaller VWF multimers enhance cleavage of C3b but large and ultra-large VWF (ULVWF) multimers have no effect on C3b cleavage and permit default complement activation. We conclude that normal plasma VWF multimers prevent complement activation and steer the complement pathway toward generation of inactivated C3b (iC3b). ULVWF multimers, as are present in patients with thrombotic microangiopathy, lack an inhibitory effect on complement and permit complement activation.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Fibrinógeno/inmunología , Factor de von Willebrand/inmunología , Proteínas del Sistema Complemento/metabolismo , Fibrinógeno/metabolismo , Humanos , Multimerización de Proteína , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
10.
Platelets ; 28(1): 99-102, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27534811

RESUMEN

Cyclooxygenase (COX) is the rate-limiting enzyme in conversion of arachidonic acid to prostanoids, and has two isoforms, COX1 and COX2, which share ~65% amino acid homology. COX1 is universally expressed in many cell types including platelets; however, expression of COX2 is known to be more limited. We examined expression of COX2 mRNA and protein in platelets and platelet-derived microparticles (MPs); using quantitative RT-PCR, immunostaining, and Western blotting. We have detected a significant amount of COX2 in platelets, both at mRNA and protein levels. We found that COX1/COX2 mRNA and protein ratios in platelets were 370:1 and 17:1, respectively. Expression level of COX2 in platelets was less than COX1, but comparable to the expression of COX2 in malignant epithelial cells. Considering the important role of COX2 in tumorigenesis and thrombosis, and the large number of circulating platelets, we propose that platelet COX2 may play an important role in physiologic and pathologic conditions.


Asunto(s)
Plaquetas/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Expresión Génica , Micropartículas Derivadas de Células/metabolismo , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Humanos , Inmunohistoquímica , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Blood ; 124(2): 184-7, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24868077

RESUMEN

Paraneoplastic thrombocytosis is associated with many solid tumors and often correlates with reduced survival. Recent studies suggest that a pathogenic feed back loop may be operative between platelets and tumor cells, with reciprocal interactions between tumor growth/metastasis and thrombocytosis/platelet activation. Specific molecular pathways have been identified in which tumors can stimulate platelet production and activation; activated platelets can, in turn, promote tumor growth and metastasis. Taken together, these findings provide exciting new potential targets for therapeutic intervention.


Asunto(s)
Síndromes Paraneoplásicos/patología , Animales , Antineoplásicos/uso terapéutico , Plaquetas/patología , Plaquetas/fisiología , Proliferación Celular , Humanos , Terapia Molecular Dirigida , Síndromes Paraneoplásicos/tratamiento farmacológico , Síndromes Paraneoplásicos/etiología , Trombocitosis/tratamiento farmacológico , Trombocitosis/etiología , Trombocitosis/patología , Microambiente Tumoral/fisiología
13.
Platelets ; 27(7): 716-718, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27185008

RESUMEN

Paraneoplastic thrombocytosis has been reported in different types of solid tumors, including ovarian epithelial cancer, and found to be associated with a worse outcome. Although the effect of cancer on increasing platelet counts is well documented, the effect of cancer on platelet functions is not well known. We compared in vitro aggregation response of platelets isolated from 34 patients with ovarian cancer to those of platelets from 19 patients with benign ovarian tumors. Aggregation studies were conducted in a light transmission aggregometer, using both a high and a low dose of ADP and collagen. We evaluated platelet preactivation by measuring the plasma concentration of ß-thromboglobulin (ß-TG) and platelet factor-4 (PF-4) as markers of platelet α granule secretion, using ELISA. We found that ovarian cancer is not associated with an enhanced aggregation response of platelets to ADP or collagen, and plasma concentration of ß-TG and PF-4 is not higher in patients with ovarian cancer compared to those in patients with benign ovarian tumors.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Ováricas/sangre , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Colágeno/metabolismo , Colágeno/farmacología , Femenino , Humanos , Neoplasias Ováricas/complicaciones , Activación Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Trombocitosis/etiología
14.
Biol Blood Marrow Transplant ; 21(8): 1392-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25985919

RESUMEN

Detection of donor-specific anti-HLA antibodies (DSA) has been associated with graft rejection in all forms of transplantation. The mechanism by which DSA increase the risk of graft failure remains unclear. We hypothesized that complement-binding DSA are associated with engraftment failure in hematopoietic stem cell transplantation (HSCT) and analyzed 122 haploidentical transplant recipients tested prospectively for DSA. Retrospective analysis to detect C1q binding DSA (C1q+DSA) was performed on 22 allosensitized recipients. Twenty-two of 122 patients (18%) had DSA, 19 of which were women (86%). Seven patients with DSA (32%) rejected the graft. Median DSA level at transplant for patients who failed to engraft was 10,055 mean fluorescence intensity (MFI) versus 2065 MFI for those who engrafted (P = .007). Nine patients with DSA were C1q positive in the initial samples with median DSA levels of 15,279 MFI (range, 1554 to 28,615), compared with 7 C1q-negative patients with median DSA levels of 2471 MFI (range, 665 to 12,254) (P = .016). Of 9 patients who were C1q positive in the initial samples, 5 patients remained C1q positive at time of transplant (all with high DSA levels [median, 15,279; range, 6487 to 22,944]) and experienced engraftment failure, whereas 4 patients became C1q negative pretransplant and all engrafted the donor cells (P = .008). In conclusion, patients with high DSA levels (>5000 MFI) and complement-binding DSA antibodies (C1q positive) appear to be at much higher risk of primary graft failure. The presence of C1q+DSA should be assessed in allosensitized patients before HSCT. Reduction of C1q+DSA levels might prevent engraftment failure in HSCT.


Asunto(s)
Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Prueba de Histocompatibilidad/métodos , Isoanticuerpos/sangre , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
15.
N Engl J Med ; 366(7): 610-8, 2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22335738

RESUMEN

BACKGROUND: The mechanisms of paraneoplastic thrombocytosis in ovarian cancer and the role that platelets play in abetting cancer growth are unclear. METHODS: We analyzed clinical data on 619 patients with epithelial ovarian cancer to test associations between platelet counts and disease outcome. Human samples and mouse models of epithelial ovarian cancer were used to explore the underlying mechanisms of paraneoplastic thrombocytosis. The effects of platelets on tumor growth and angiogenesis were ascertained. RESULTS: Thrombocytosis was significantly associated with advanced disease and shortened survival. Plasma levels of thrombopoietin and interleukin-6 were significantly elevated in patients who had thrombocytosis as compared with those who did not. In mouse models, increased hepatic thrombopoietin synthesis in response to tumor-derived interleukin-6 was an underlying mechanism of paraneoplastic thrombocytosis. Tumor-derived interleukin-6 and hepatic thrombopoietin were also linked to thrombocytosis in patients. Silencing thrombopoietin and interleukin-6 abrogated thrombocytosis in tumor-bearing mice. Anti-interleukin-6 antibody treatment significantly reduced platelet counts in tumor-bearing mice and in patients with epithelial ovarian cancer. In addition, neutralizing interleukin-6 significantly enhanced the therapeutic efficacy of paclitaxel in mouse models of epithelial ovarian cancer. The use of an antiplatelet antibody to halve platelet counts in tumor-bearing mice significantly reduced tumor growth and angiogenesis. CONCLUSIONS: These findings support the existence of a paracrine circuit wherein increased production of thrombopoietic cytokines in tumor and host tissue leads to paraneoplastic thrombocytosis, which fuels tumor growth. We speculate that countering paraneoplastic thrombocytosis either directly or indirectly by targeting these cytokines may have therapeutic potential. (Funded by the National Cancer Institute and others.).


Asunto(s)
Interleucina-6/antagonistas & inhibidores , Neoplasias Glandulares y Epiteliales/complicaciones , Neoplasias Ováricas/complicaciones , Síndromes Paraneoplásicos , Trombocitosis/etiología , Animales , Anticuerpos Monoclonales/uso terapéutico , Plaquetas/inmunología , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Interleucina-6/sangre , Interleucina-6/inmunología , Estimación de Kaplan-Meier , Ratones , Ratones Noqueados , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/sangre , Neoplasias Ováricas/tratamiento farmacológico , Recuento de Plaquetas , Modelos de Riesgos Proporcionales , Receptores de Interleucina-6/deficiencia , Transducción de Señal , Trombopoyetina/antagonistas & inhibidores , Trombopoyetina/sangre
16.
Blood ; 122(8): 1487-93, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23847193

RESUMEN

Complement dysregulation leads to atypical hemolytic uremic syndrome (aHUS), while ADAMTS13 deficiency causes thrombotic thrombocytopenic purpura. We investigated whether genetic variations in the ADAMTS13 gene partially explain the reduced activity known to occur in some patients with aHUS. We measured complement activity and ADAMTS13 function, and completed mutation screening of multiple complement genes and ADAMTS13 in a large cohort of aHUS patients. In over 50% of patients we identified complement gene mutations. Surprisingly, 80% of patients also carried at least 1 nonsynonymous change in ADAMTS13, and in 38% of patients, multiple ADAMTS13 variations were found. Six of the 9 amino acid substitutions in ADAMTS13 were common single nucleotide polymorphisms; however, 3 variants-A747V, V832M, and R1096H- were rare, with minor allele frequencies of 0.0094%, 0.5%, and 0.32%, respectively. Reduced complement and ADAMTS13 activity (<60% of normal activity) were found in over 60% and 50% of patients, respectively. We concluded that partial ADAMTS13 deficiency is a common finding in aHUS patients and that genetic screening and functional tests of ADAMTS13 should be considered in these patients.


Asunto(s)
Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/genética , Proteína ADAMTS13 , Adolescente , Adulto , Alelos , Síndrome Hemolítico Urémico Atípico , Autoanticuerpos/inmunología , Niño , Preescolar , Estudios de Cohortes , Factor H de Complemento/inmunología , ADN/genética , Femenino , Humanos , Lactante , Masculino , Mutación , Polimorfismo Genético , Proteínas Recombinantes de Fusión/metabolismo , Adulto Joven
17.
Blood ; 129(16): 2214-2215, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28428238
18.
Blood ; 120(24): 4869-72, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22966171

RESUMEN

Platelets promote metastasis and angiogenesis, but their effect on tumor cell growth is uncertain. Here we report a direct proliferative effect of platelets on cancer cells both in vitro and in vivo. Incubation of platelets with ovarian cancer cells from murine (ID8 and 2C6) or human (SKOV3 and OVCAR5) origin increased cell proliferation. The proliferative effect of platelets was not dependent on direct contact with cancer cells, and preincubation of platelets with blocking antibodies against platelet adhesion molecules did not alter their effect on cancer cells. The proliferative effect of platelets was reduced by fixing platelets with paraformaldehyde, preincubating platelets with a TGF-ß1-blocking antibody, or reducing expression of TGF-ßR1 receptor on cancer cells with siRNA. Infusing platelets into mice with orthotopic ovarian tumors significantly increased the proliferation indices in these tumors. Ovarian cancer patients with thrombocytosis had higher tumor proliferation indices compared with patients with normal platelet counts.


Asunto(s)
Plaquetas/citología , Proliferación Celular , Neoplasias Ováricas/patología , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/terapia , Recuento de Plaquetas , Transfusión de Plaquetas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Trombocitosis/complicaciones
19.
Arterioscler Thromb Vasc Biol ; 33(11): 2524-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24008159

RESUMEN

OBJECTIVE: Ultralarge von Willebrand factor (vWF) strings are secreted by, and anchored to, stimulated human endothelial cells. A disintegrin and metalloprotease with thrombospondin domains-type 13 cleaves the ultralarge vWF strings into large soluble vWF multimers. Normal plasma contains a nonproteolytic reducing activity that subsequently rapidly diminishes the size of the large soluble vWF multimers. APPROACH AND RESULTS: The vWF reductase activity was isolated from normal cryoprecipitate-poor plasma by chromatography and identified as the complement regulatory protein, factor H (FH), by mass spectroscopy, SDS-PAGE, and monospecific anti-FH antibody. Removal of FH from partially purified vWF reductase by immunoabsorption eliminated the reducing activity, and the activity was recovered in the eluates. Recombinant human FH reduced large soluble vWF multimers in a free thiol-dependent reaction that was not inhibited by a variety of protease inhibitors. CONCLUSIONS: FH contributes to the reduction of large soluble vWF multimers.


Asunto(s)
Endotelio Vascular/metabolismo , Oxidorreductasas/metabolismo , Factor de von Willebrand/metabolismo , Factor H de Complemento/metabolismo , Activación Enzimática/fisiología , Humanos , Peso Molecular , Solubilidad , Compuestos de Sulfhidrilo/metabolismo , Factor de von Willebrand/química
20.
Cardiovasc Intervent Radiol ; 47(5): 556-566, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38548981

RESUMEN

PURPOSE: Venous thromboembolism (VTE) is a major contributor to the mortality of cancer patients. Mechanical thrombectomy (MT) is an endovascular technique that physically removes a thrombus without thrombolytics. The purpose of this study was to evaluate safety, efficacy, and clinical outcomes following MT for lower extremity DVT in cancer patients. METHODS: This single-center, retrospective study evaluated outcomes following MT of lower extremity DVT in cancer patients from November 2019 to May 2023. The primary outcome measure was clinical success, defined as a decrease in Villalta score by at least 2 points following the intervention. Secondary outcomes included repeat intervention-free survival and overall survival. Technical success was defined as restoring venous flow with mild (< 10%) or no residual filling defect. RESULTS: In total, 90 patients and 113 procedures were included. Technical and clinical success was achieved in 81% and 87% of procedures performed. Repeat intervention-free survival at 1 month, 3 months, and 6 months post-procedure was 92%, 82%, and 77%, respectively. The complication rate was 2.7%. Pathologic analysis of the extracted thrombus revealed tumor thrombus in 18.4% (18/98) samples. Overall survival for the study cohort was 87% at 1 month, 74% at 3 months, and 62% at 6 months. Patients who were found to have tumor thrombi were noted to have a decreased overall survival compared to patients with non-tumor thrombi (P = 0.012). CONCLUSION: MT is safe and efficacious in reducing cancer patients' VTE-related symptoms. The high rate of tumor thrombus in thrombectomy specimens suggests this phenomenon is more common than suspected.


Asunto(s)
Neoplasias , Trombectomía , Trombosis de la Vena , Humanos , Estudios Retrospectivos , Femenino , Masculino , Persona de Mediana Edad , Trombosis de la Vena/diagnóstico por imagen , Trombosis de la Vena/terapia , Neoplasias/complicaciones , Anciano , Trombectomía/métodos , Adulto , Resultado del Tratamiento , Anciano de 80 o más Años
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA