Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(1): 75-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25347465

RESUMEN

In lymph nodes, fibroblastic reticular cells (FRCs) form a collagen-based reticular network that supports migratory dendritic cells (DCs) and T cells and transports lymph. A hallmark of FRCs is their propensity to contract collagen, yet this function is poorly understood. Here we demonstrate that podoplanin (PDPN) regulates actomyosin contractility in FRCs. Under resting conditions, when FRCs are unlikely to encounter mature DCs expressing the PDPN receptor CLEC-2, PDPN endowed FRCs with contractile function and exerted tension within the reticulum. Upon inflammation, CLEC-2 on mature DCs potently attenuated PDPN-mediated contractility, which resulted in FRC relaxation and reduced tissue stiffness. Disrupting PDPN function altered the homeostasis and spacing of FRCs and T cells, which resulted in an expanded reticular network and enhanced immunity.


Asunto(s)
Colágeno/metabolismo , Fibroblastos/citología , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/citología , Glicoproteínas de Membrana/metabolismo , Amidas/farmacología , Animales , Supervivencia Celular/inmunología , Colágeno/inmunología , Citoesqueleto/inmunología , Citoesqueleto/ultraestructura , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/inmunología , Fibroblastos/ultraestructura , Lectinas Tipo C/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/ultraestructura , Masculino , Glicoproteínas de Membrana/inmunología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosforilación , Piridinas/farmacología , Organismos Libres de Patógenos Específicos
2.
Blood ; 143(13): 1293-1309, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38142410

RESUMEN

ABSTRACT: Although it is caused by a single-nucleotide mutation in the ß-globin gene, sickle cell anemia (SCA) is a systemic disease with complex, incompletely elucidated pathologies. The mononuclear phagocyte system plays critical roles in SCA pathophysiology. However, how heterogeneous populations of hepatic macrophages contribute to SCA remains unclear. Using a combination of single-cell RNA sequencing and spatial transcriptomics via multiplexed error-robust fluorescence in situ hybridization, we identified distinct macrophage populations with diversified origins and biological functions in SCA mouse liver. We previously found that administering the von Willebrand factor (VWF)-cleaving protease ADAMTS13 alleviated vaso-occlusive episode in mice with SCA. Here, we discovered that the ADAMTS13-cleaved VWF was cleared from the circulation by a Clec4f+Marcohigh macrophage subset in a desialylation-dependent manner in the liver. In addition, sickle erythrocytes were phagocytized predominantly by Clec4f+Marcohigh macrophages. Depletion of macrophages not only abolished the protective effect of ADAMTS13 but exacerbated vaso-occlusive episode in mice with SCA. Furthermore, promoting macrophage-mediated VWF clearance reduced vaso-occlusion in SCA mice. Our study demonstrates that hepatic macrophages are important in the pathogenesis of SCA, and efficient clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in SCA mice.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Ratones , Animales , Factor de von Willebrand/genética , Hibridación Fluorescente in Situ , Anemia de Células Falciformes/patología , Macrófagos/patología , Proteína ADAMTS13/genética
3.
Blood ; 142(12): 1071-1081, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37294924

RESUMEN

Rebalance of coagulation and anticoagulation to achieve a hemostatic effect has recently gained attention as an alternative therapeutic strategy for hemophilia. We engineered a humanized chimeric antibody, SR604, based on a previously published murine antibody, HAPC1573, which selectively blocks the anticoagulant activity of human activated protein C (APC). SR604 effectively blocked the anticoagulation activities of APC in human plasma deficient in various coagulation factors in vitro with affinities ∼60 times greater than that of HAPC1573. SR604 exhibited prophylactic and therapeutic efficacy in the tail-bleeding and knee-injury models of hemophilia A and B mice expressing human APC (humanized hemophilic mice). SR604 did not interfere with the cytoprotection and endothelial barrier function of APC, nor were there obvious toxicity effects in humanized hemophilic mice. Pharmacokinetic study showed a high bioavailability (106%) of subcutaneously injected SR604 in cynomolgus monkeys. These results demonstrate that SR604 is expected to be a safe and effective therapeutic and/or prophylactic agent with a prolonged half-life for patients with congenital factor deficiencies including hemophilia A and B.


Asunto(s)
Hemofilia A , Proteína C , Humanos , Ratones , Animales , Proteína C/uso terapéutico , Hemofilia A/tratamiento farmacológico , Modelos Animales de Enfermedad , Coagulación Sanguínea , Anticoagulantes/uso terapéutico
4.
Circ Res ; 133(6): 463-480, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37555328

RESUMEN

BACKGROUND: Cardiac valve disease is observed in 2.5% of the general population and 10% of the elderly people. Effective pharmacological treatments are currently not available, and patients with severe cardiac valve disease require surgery. PROX1 (prospero-related homeobox transcription factor 1) and FOXC2 (Forkhead box C2 transcription factor) are transcription factors that are required for the development of lymphatic and venous valves. We found that PROX1 and FOXC2 are expressed in a subset of valvular endothelial cells (VECs) that are located on the downstream (fibrosa) side of cardiac valves. Whether PROX1 and FOXC2 regulate cardiac valve development and disease is not known. METHODS: We used histology, electron microscopy, and echocardiography to investigate the structure and functioning of heart valves from Prox1ΔVEC mice in which Prox1 was conditionally deleted from VECs. Isolated valve endothelial cells and valve interstitial cells were used to identify the molecular mechanisms in vitro, which were tested in vivo by RNAScope, additional mouse models, and pharmacological approaches. The significance of our findings was tested by evaluation of human samples of mitral valve prolapse and aortic valve insufficiency. RESULTS: Histological analysis revealed that the aortic and mitral valves of Prox1ΔVEC mice become progressively thick and myxomatous. Echocardiography revealed that the aortic valves of Prox1ΔVEC mice are stenotic. FOXC2 was downregulated and PDGF-B (platelet-derived growth factor-B) was upregulated in the VECs of Prox1ΔVEC mice. Conditional knockdown of FOXC2 and conditional overexpression of PDGF-B in VECs recapitulated the phenotype of Prox1ΔVEC mice. PDGF-B was also increased in mice lacking FOXC2 and in human mitral valve prolapse and insufficient aortic valve samples. Pharmacological inhibition of PDGF-B signaling with imatinib partially ameliorated the valve defects of Prox1ΔVEC mice. CONCLUSIONS: PROX1 antagonizes PDGF-B signaling partially via FOXC2 to maintain the extracellular matrix composition and prevent myxomatous degeneration of cardiac valves.


Asunto(s)
Enfermedades de las Válvulas Cardíacas , Prolapso de la Válvula Mitral , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/prevención & control , Enfermedades de las Válvulas Cardíacas/metabolismo , Válvula Mitral/metabolismo , Prolapso de la Válvula Mitral/metabolismo , Factores de Transcripción/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-38899470

RESUMEN

BACKGROUND: Integrin-regulated monocyte recruitment and cellular responses of monocyte-derived macrophages are critical for the pathogenesis of atherosclerosis. In the canonical model, talin1 controls ligand binding to integrins, a prerequisite for integrins to mediate leukocyte recruitment and induce immune responses. However, the role of talin1 in the development of atherosclerosis has not been studied. Our study investigated how talin1 in myeloid cells regulates the progression of atherosclerosis. METHODS: On an Apoe-/- background, myeloid talin1-deficient mice and the control mice were fed with a high-fat diet for 8 or 12 weeks to induce atherosclerosis. The atherosclerosis development in the aorta and monocyte recruitment into atherosclerotic lesions were analyzed. RESULTS: Myeloid talin1 deletion facilitated the formation of atherosclerotic lesions and macrophage deposition in lesions. Talin1 deletion abolished integrin ß2-mediated adhesion of monocytes but did not impair integrin α4ß1-dependent cell adhesion in a flow adhesion assay. Strikingly, talin1 deletion did not prevent Mn2+- or chemokine-induced activation of integrin α4ß1 to the high-affinity state for ligands. In an in vivo competitive homing assay, monocyte infiltration into inflamed tissues was prohibited by antibodies to integrin α4ß1 but was not affected by talin1 deletion or antibodies to integrin ß2. Furthermore, quantitative polymerase chain reaction and ELISA analysis showed that macrophages produced cytokines to promote inflammation and the proliferation of smooth muscle cells. Ligand binding to integrin ß3 inhibited cytokine generation in macrophages, although talin1 deletion abolished the negative effects of integrin ß3. CONCLUSIONS: Integrin α4ß1 controls monocyte recruitment during atherosclerosis. Talin1 is dispensable for integrin α4ß1 activation to the high-affinity state and integrin α4ß1-mediated monocyte recruitment. Yet, talin1 is required for integrin ß3 to inhibit the production of inflammatory cytokines in macrophages. Thus, intact monocyte recruitment and elevated inflammatory responses cause enhanced atherosclerosis in talin1-deficient mice. Our study provides novel insights into the roles of myeloid talin1 and integrins in the progression of atherosclerosis.

6.
Proc Natl Acad Sci U S A ; 119(34): e2207592119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969769

RESUMEN

Vaso-occlusive episode (VOE) is a common and critical complication of sickle cell disease (SCD). Its pathogenesis is incompletely understood. von Willebrand factor (VWF), a multimeric plasma hemostatic protein synthesized and secreted by endothelial cells and platelets, is increased during a VOE. However, whether and how VWF contributes to the pathogenesis of VOE is not fully understood. In this study, we found increased VWF levels during tumor necrosis factor (TNF)-induced VOE in a humanized mouse model of SCD. Deletion of endothelial VWF decreased hemolysis, vascular occlusion, and organ damage caused by TNF-induced VOE in SCD mice. Moreover, administering ADAMTS13, the VWF-cleaving plasma protease, reduced plasma VWF levels, decreased inflammation and vaso-occlusion, and alleviated organ damage during VOE. These data suggest that promoting VWF cleavage via ADAMTS13 may be an effective treatment for reducing hemolysis, inflammation, and vaso-occlusion during VOE.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Factor de von Willebrand , Proteína ADAMTS13/metabolismo , Proteína ADAMTS13/farmacología , Proteína ADAMTS13/uso terapéutico , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Eliminación de Gen , Hemólisis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/etiología , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
7.
Blood ; 139(16): 2523-2533, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35157766

RESUMEN

Microvascular thrombosis in patients with thrombotic thrombocytopenic purpura (TTP) is initiated by GPIbα-mediated platelet binding to von Willebrand factor (VWF). Binding of VWF to GPIbα causes activation of the platelet surface integrin αIIbß3. However, the mechanism of GPIbα-initiated activation of αIIbß3 and its clinical importance for microvascular thrombosis remain elusive. Deletion of platelet C-type lectin-like receptor 2 (CLEC-2) did not prevent VWF binding to platelets but specifically inhibited platelet aggregation induced by VWF binding in mice. Deletion of platelet CLEC-2 also inhibited αIIbß3 activation induced by the binding of VWF to GPIbα. Using a mouse model of TTP, which was created by infusion of anti-mouse ADAMTS13 monoclonal antibodies followed by infusion of VWF, we found that deletion of platelet CLEC-2 decreased pulmonary arterial thrombosis and the severity of thrombocytopenia. Importantly, prophylactic oral administration of aspirin, an inhibitor of platelet activation, and therapeutic treatment of the TTP mice with eptifibatide, an integrin αIIbß3 antagonist, reduced pulmonary arterial thrombosis in the TTP mouse model. Our observations demonstrate that GPIbα-mediated activation of integrin αIIbß3 plays an important role in the formation of thrombosis in TTP. These observations suggest that prevention of platelet activation with aspirin may reduce the risk for thrombosis in patients with TTP.


Asunto(s)
Hipertensión Pulmonar , Púrpura Trombocitopénica Trombótica , Trombosis , Aspirina , Plaquetas/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Púrpura Trombocitopénica Trombótica/metabolismo , Trombosis/etiología , Factor de von Willebrand/metabolismo
8.
Genes Dev ; 30(12): 1454-69, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27313318

RESUMEN

Lymphatic vasculature regulates fluid homeostasis by returning interstitial fluid to blood circulation. Lymphatic endothelial cells (LECs) are the building blocks of the entire lymphatic vasculature. LECs originate as a homogeneous population of cells predominantly from the embryonic veins and undergo stepwise morphogenesis to become the lymphatic capillaries, collecting vessels or valves. The molecular mechanisms underlying the morphogenesis of the lymphatic vasculature remain to be fully understood. Here we show that canonical Wnt/ß-catenin signaling is necessary for lymphatic vascular morphogenesis. Lymphatic vascular-specific ablation of ß-catenin in mice prevents the formation of lymphatic and lymphovenous valves. Additionally, lymphatic vessel patterning is defective in these mice, with abnormal recruitment of mural cells. We found that oscillatory shear stress (OSS), which promotes lymphatic vessel maturation, triggers Wnt/ß-catenin signaling in LECs. In turn, Wnt/ß-catenin signaling controls the expression of several molecules, including the lymphedema-associated transcription factor FOXC2. Importantly, FOXC2 completely rescues the lymphatic vessel patterning defects in mice lacking ß-catenin. Thus, our work reveals that mechanical stimulation is a critical regulator of lymphatic vascular development via activation of Wnt/ß-catenin signaling and, in turn, FOXC2.


Asunto(s)
Linfangiogénesis/fisiología , Mecanotransducción Celular/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Células Cultivadas , Células Endoteliales/citología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Humanos , Vasos Linfáticos/embriología , Ratones , beta Catenina/genética
9.
Blood ; 137(20): 2756-2769, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33619517

RESUMEN

During early embryonic development in mammals, including humans and mice, megakaryocytes (Mks) first originate from primitive hematopoiesis in the yolk sac. These embryonic Mks (eMks) circulate in the vasculature with unclear function. Herein, we report that podoplanin (PDPN), the ligand of C-type lectin-like receptor (CLEC-2) on Mks/platelets, is temporarily expressed in neural tissue during midgestation in mice. Loss of PDPN or CLEC-2 resulted in aneurysms and spontaneous hemorrhage, specifically in the lower diencephalon during midgestation. Surprisingly, more eMks/platelets had enhanced granule release and localized to the lower diencephalon in mutant mouse embryos than in wild-type littermates before hemorrhage. We found that PDPN counteracted the collagen-1-induced secretion of angiopoietin-1 from fetal Mks, which coincided with enhanced TIE-2 activation in aneurysm-like sprouts of PDPN-deficient embryos. Blocking platelet activation prevented the PDPN-deficient embryo from developing vascular defects. Our data reveal a new role for PDPN in regulating eMk function during midgestation.


Asunto(s)
Encéfalo/irrigación sanguínea , Aneurisma Intracraneal/etiología , Megacariocitos/patología , Glicoproteínas de Membrana/deficiencia , Aneurisma Roto/embriología , Aneurisma Roto/etiología , Angiopoyetina 1/metabolismo , Animales , Encéfalo/embriología , Células Cultivadas , Hemorragia Cerebral/embriología , Hemorragia Cerebral/etiología , Colágeno/farmacología , Diencéfalo/irrigación sanguínea , Diencéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Aneurisma Intracraneal/embriología , Aneurisma Intracraneal/genética , Aneurisma Intracraneal/patología , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas Tipo C/fisiología , Megacariocitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Activación Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Receptor TIE-2/metabolismo
10.
Phys Rev Lett ; 130(22): 223601, 2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37327414

RESUMEN

Integrated quantum photonics has recently emerged as a powerful platform for generating, manipulating, and detecting entangled photons. Multipartite entangled states lie at the heart of the quantum physics and are the key enabling resources for scalable quantum information processing. Dicke state is an important class of genuinely entangled state, which has been systematically studied in the light-matter interactions, quantum state engineering, and quantum metrology. Here, by using a silicon photonic chip, we report the generation and collectively coherent control of the entire family of four-photon Dicke states, i.e., with arbitrary excitations. We generate four entangled photons from two microresonators and coherently control them in a linear-optic quantum circuit, in which the nonlinear and linear processing are achieved in a chip-scale device. The generated photons are in telecom band, which lays the groundwork for large-scale photonic quantum technologies for multiparty networking and metrology.


Asunto(s)
Cognición , Ojo , Corazón , Fotones , Física
11.
Blood ; 144(3): 247-248, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39023873
12.
Haematologica ; 106(3): 759-769, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32303557

RESUMEN

Sialic acid is a common terminal residue of glycans on proteins and acidic sphingolipids such as gangliosides and has important biological functions. The sialylation process is controlled by more than 20 different sialyltransferases, many of which exhibit overlapping functions. Thus, it is difficult to determine the overall biological function of sialylation by targeted deletion of individual sialyltransferases. To address this issue, we established a mouse line with the Slc35a1 gene flanked by loxP sites. Slc35a1 encodes the cytidine-5'-monophosphate (CMP)-sialic acid transporter that transports CMP-sialic acid from the cytoplasm into the Golgi apparatus for sialylation. Here we report our study regarding the role of sialylation on megakaryocytes and platelets using a mouse line with significantly reduced sialylation in megakaryocytes and platelets (Plt Slc35a1­ /­). The major phenotype of Plt Slc35a1­/­ mice was thrombocytopenia. The number of bone marrow megakaryocytes in Plt Slc35a1­/­ mice was reduced, and megakaryocyte maturation was also impaired. In addition, an increased number of desialylated platelets was cleared by Küpffer cells in the liver of Plt Slc35a1­/­ mice. This study provides new insights into the role of sialylation in platelet homeostasis and the mechanisms of thrombocytopenia in diseases associated with platelet desialylation, such as immune thrombocytopenia and a rare congenital disorder of glycosylation (CDG), SLC35A1-CDG, which is caused by SLC35A1 mutations.


Asunto(s)
Proteínas de Transporte de Nucleótidos , Trombocitopenia , Plaquetas , Humanos , Hígado , Ácido N-Acetilneuramínico , Proteínas de Transporte de Nucleótidos/genética , Trombocitopenia/genética , Trombopoyesis
13.
J Cell Mol Med ; 24(7): 4356-4361, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32073234

RESUMEN

Hereditary thrombotic thrombocytopenic purpura (TTP) is an autosomal recessive thrombosis disorder, caused by loss-of-function mutations in ADAMTS13. Mutations in the CUB domains of ADAMTS13 are rare, and the exact mechanisms through which these mutations result in the development of TTP have not yet been fully elucidated. In this study, we identified two novel mutations in the CUB domains in a TTP family with an acceptor splice-site mutation (c.3569-1, G>A, intron 25) and a point missense mutation (c.3923, G>A, exon 28), resulting in a glycine to aspartic acid substitution (p.G1308D). In vitro splicing analysis revealed that the intronic mutation resulted in abnormal pre-mRNA splicing, and an in vitro expression assay revealed that the missense mutation significantly impaired ADAMTS13 secretion. Although both the patient and her brother displayed significantly reduced ADAMTS13 activity and increased levels of ultra-large VWF (ULVWF) multimers in plasma, only the female developed acute episodes of TTP. Our findings indicate the importance of the CUB domains for the protein stability and extracellular secretion of ADAMTS13.


Asunto(s)
Proteína ADAMTS13/genética , Multimerización de Proteína/genética , Púrpura Trombocitopénica Trombótica/genética , Precursores del ARN/sangre , Proteína ADAMTS13/sangre , Adulto , Femenino , Humanos , Mutación con Pérdida de Función/genética , Mutación Missense/genética , Estabilidad Proteica , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/patología , Precursores del ARN/genética , Empalme del ARN/genética , Factor de von Willebrand/genética
14.
Proc Natl Acad Sci U S A ; 114(31): 8360-8365, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716912

RESUMEN

Most platelet membrane proteins are modified by mucin-type core 1-derived glycans (O-glycans). However, the biological importance of O-glycans in platelet clearance is unclear. Here, we generated mice with a hematopoietic cell-specific loss of O-glycans (HC C1galt1-/- ). These mice lack O-glycans on platelets and exhibit reduced peripheral platelet numbers. Platelets from HC C1galt1-/- mice show reduced levels of α-2,3-linked sialic acids and increased accumulation in the liver relative to wild-type platelets. The preferential accumulation of HC C1galt1-/- platelets in the liver was reduced in mice lacking the hepatic asialoglycoprotein receptor [Ashwell-Morell receptor (AMR)]. However, we found that Kupffer cells are the primary cells phagocytosing HC C1galt1-/- platelets in the liver. Our results demonstrate that hepatic AMR promotes preferential adherence to and phagocytosis of desialylated and/or HC C1galt1-/- platelets by the Kupffer cell through its C-type lectin receptor CLEC4F. These findings provide insights into an essential role for core 1 O-glycosylation of platelets in their clearance in the liver.


Asunto(s)
Plaquetas/metabolismo , Galactosiltransferasas/genética , Macrófagos del Hígado/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/metabolismo , Animales , Receptor de Asialoglicoproteína/metabolismo , Hepatocitos/metabolismo , Homeostasis/fisiología , Lectinas Tipo C/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitopenia/patología
15.
Gastroenterology ; 155(5): 1608-1624, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30086262

RESUMEN

BACKGROUND & AIMS: Pancreatic ductal adenocarcinomas (PDACs) produce higher levels of truncated O-glycan structures (such as Tn and sTn) than normal pancreata. Dysregulated activity of core 1 synthase glycoprotein-N-acetylgalactosamine 3-ß-galactosyltransferase 1 (C1GALT1) leads to increased expression of these truncated O-glycans. We investigated whether and how truncated O-glycans contributes to the development and progression of PDAC using mice with disruption of C1galt1. METHODS: We crossed C1galt1 floxed mice (C1galt1loxP/loxP) with KrasG12D/+; Trp53R172H/+; Pdx1-Cre (KPC) mice to create KPCC mice. Growth and progression of pancreatic tumors were compared between KPC and KPCC mice; pancreatic tissues were collected and analyzed by immunohistochemistry; immunofluorescence; and Sirius red, alcian blue, and lectin staining. We used the CRISPR/Cas9 system to disrupt C1GALT1 in human PDAC cells (T3M4 and CD18/HPAF) and levels of O-glycans were analyzed by lectin blotting, mass spectrometry, and lectin pulldown assay. Orthotopic studies and RNA sequencing analyses were performed with control and C1GALT1 knockout PDAC cells. C1GALT1 expression was analyzed in well-differentiated (n = 36) and poorly differentiated (n = 23) PDAC samples by immunohistochemistry. RESULTS: KPCC mice had significantly shorter survival times (median 102 days) than KPC mice (median 200 days) and developed early pancreatic intraepithelial neoplasias at 3 weeks, PDAC at 5 weeks, and metastasis at 10 weeks compared with KPC mice. Pancreatic tumors that developed in KPCC mice were more aggressive (more invasive and metastases) than those in KPC mice, had a decreased amount of stroma, and had increased production of Tn. Poorly differentiated PDAC specimens had significantly lower levels of C1GALT1 than well-differentiated PDACs. Human PDAC cells with knockout of C1GALT1 had aberrant glycosylation of MUC16 compared with control cells and increased expression of genes that regulate tumorigenesis and metastasis. CONCLUSIONS: In studies of KPC mice with disruption of C1galt1, we found that loss of C1galt1 promotes development of aggressive PDACs and increased metastasis. Knockout of C1galt1 leads to increased tumorigenicity and truncation of O-glycosylation on MUC16, which could contribute to increased aggressiveness.


Asunto(s)
Adenocarcinoma/etiología , Galactosiltransferasas/fisiología , Neoplasias Pancreáticas/etiología , Adenocarcinoma/secundario , Animales , Sistemas CRISPR-Cas , Carcinoma Ductal Pancreático , Proliferación Celular , Galactosiltransferasas/genética , Glicosilación , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/patología
16.
Blood ; 140(22): 2314-2315, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36454596
17.
BMC Cancer ; 19(1): 599, 2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-31208371

RESUMEN

BACKGROUND: Podoplanin (PDPN), a transmembrane O-glycoprotein, is up-regulated in many tumors and is involved in tumor metastasis and malignant progression. In previous studies, we generated a functional blocking monoclonal antibody (mAb, SZ168) against the extracellular domain of human PDPN. This study is aimed to investigate whether blocking PDPN by SZ168 inhibits tumor growth and metastasis. METHODS: Malignant melanoma xenograft model by inoculating subcutaneously human malignant melanoma cell line C8161 into the back of BALB/c nude mice was used. Endogenous PDPN expression in C8161 cells and nasopharyngeal cancer cell line CNE-2 was detected using western blot and flow cytometry. RESULTS: SZ168 significantly inhibited C8161 or CNE-2 cell-induced platelet aggregation in a dose-dependent manner with a maximal inhibition of 73.9 ± 3.0% in C8161 cells or 77.1 ± 2.7% in CNE-2 cells. Moreover, SZ168 inhibited the growth and pulmonary metastasis of C8161cells in vivo. The number of lung metastatic foci in the SZ168-treated group was significantly decreased compared with that in the control mouse IgG group (1.61 ± 0.44 vs.3.83 ± 0.60, P < 0.01). Subcutaneous tumor volume, weight, and incidence were also significantly reduced in the SZ168-treated group compared to the control group (P < 0.05). Additionally, SZ168 recognized PDPN in immunohistochemical analyses of tumor tissue sections. CONCLUSIONS: SZ168 blocks growth and pulmonary metastasis of human malignant melanoma by inhibiting the interaction between tumor PDPN and platelet CLEC-2 and therefore is a promising antibody for therapeutic development against malignant melanoma.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetulus , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nature ; 502(7469): 105-9, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23995678

RESUMEN

Circulating lymphocytes continuously enter lymph nodes for immune surveillance through specialized blood vessels named high endothelial venules, a process that increases markedly during immune responses. How high endothelial venules (HEVs) permit lymphocyte transmigration while maintaining vascular integrity is unknown. Here we report a role for the transmembrane O-glycoprotein podoplanin (PDPN, also known as gp38 and T1α) in maintaining HEV barrier function. Mice with postnatal deletion of Pdpn lost HEV integrity and exhibited spontaneous bleeding in mucosal lymph nodes, and bleeding in the draining peripheral lymph nodes after immunization. Blocking lymphocyte homing rescued bleeding, indicating that PDPN is required to protect the barrier function of HEVs during lymphocyte trafficking. Further analyses demonstrated that PDPN expressed on fibroblastic reticular cells, which surround HEVs, functions as an activating ligand for platelet C-type lectin-like receptor 2 (CLEC-2, also known as CLEC1B). Mice lacking fibroblastic reticular cell PDPN or platelet CLEC-2 exhibited significantly reduced levels of VE-cadherin (also known as CDH5), which is essential for overall vascular integrity, on HEVs. Infusion of wild-type platelets restored HEV integrity in Clec-2-deficient mice. Activation of CLEC-2 induced release of sphingosine-1-phosphate from platelets, which promoted expression of VE-cadherin on HEVs ex vivo. Furthermore, draining peripheral lymph nodes of immunized mice lacking sphingosine-1-phosphate had impaired HEV integrity similar to Pdpn- and Clec-2-deficient mice. These data demonstrate that local sphingosine-1-phosphate release after PDPN-CLEC-2-mediated platelet activation is critical for HEV integrity during immune responses.


Asunto(s)
Endotelio Linfático/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio Linfático/inmunología , Femenino , Regulación de la Expresión Génica , Uniones Intercelulares/genética , Uniones Intercelulares/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Lisofosfolípidos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Esfingosina/análogos & derivados , Esfingosina/metabolismo
19.
J Biol Chem ; 292(40): 16491-16497, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28842487

RESUMEN

The kidney's filtration activity is essential for removing toxins and waste products from the body. The vascular endothelial cells of the glomerulus are fenestrated, flattened, and surrounded by podocytes, specialized cells that support glomerular endothelial cells. Mucin-type core 1-derived O-glycans (O-glycans) are highly expressed on both glomerular capillary endothelial cells and their supporting podocytes, but their biological role is unclear. Biosynthesis of core 1-derived O-glycans is catalyzed by the glycosyltransferase core 1 ß1,3-galactosyltransferase (C1galt1). Here we report that neonatal or adult mice with inducible deletion of C1galt1 (iC1galt1-/-) exhibit spontaneous proteinuria and rapidly progressing glomerulosclerosis. Ultrastructural analysis of the glomerular filtration barrier components revealed that loss of O-glycans results in altered podocyte foot processes. Further analysis indicated that O-glycan is essential for the normal signaling function of podocalyxin, a podocyte foot process-associated glycoprotein. Our results reveal a new function of O-glycosylation in the integrity of the glomerular filtration barrier.


Asunto(s)
Galactosiltransferasas/metabolismo , Mucinas , Podocitos/metabolismo , Polisacáridos/metabolismo , Sialoglicoproteínas/metabolismo , Transducción de Señal/fisiología , Animales , Galactosiltransferasas/genética , Ratones , Ratones Noqueados , Polisacáridos/genética , Sialoglicoproteínas/genética
20.
Cancer Sci ; 109(2): 403-411, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29266546

RESUMEN

Podoplanin (PDPN) is expressed on many tumors and is involved in tumor metastasis. The objective of the present study was to develop an ELISA for determining soluble PDPN (sPDPN) levels as a potential novel tumor marker in plasma of patients with cancers for detection of tumor occurrence and metastasis. Mouse monoclonal antibodies (mAb) against human PDPN were developed and characterized. Two anti-PDPN mAb, SZ-163 and SZ-168, were used in a sandwich ELISA to detect plasma sPDPN in patients with cancers and in normal individuals. The levels of sPDPN were detected in patients with adenocarcinoma (87 cases, 31.09 ± 5.48 ng/ml), squamous cell carcinoma (86 cases, 6.91 ± 0.59 ng/ml), lung cancer (45 cases, 26.10 ± 7.62 ng/ml), gastric cancer (38 cases, 23.71 ± 6.90 ng/ml) and rectal cancer (27 cases, 32.98 ± 9.88 ng/ml), which were significantly higher than those in normal individuals (99 cases, 1.31 ± 0.13 ng/ml) (P < .0001). Moreover, the sPDPN levels in patients with metastatic cancers were higher (192 cases, 30.35 ± 3.63 ng/ml) than those in non-metastatic cancer patients (92 cases, 6.28 ± 0.77 ng/ml) (P < .0001). The post-treatment sPDPN levels of cancer patients (n = 156) (4.47 ± 0.35 ng/ml) were significantly lower compared with those seen pre-treatment (n = 128) (43.74 ± 4.97 ng/ml) (P < .0001). These results showed that an ELISA method was successfully established for quantitation of plasma sPDPN and plasma sPDPN levels correlate significantly with tumor occurrence and metastasis.


Asunto(s)
Biomarcadores de Tumor/sangre , Glicoproteínas de Membrana/sangre , Neoplasias/diagnóstico , Animales , Anticuerpos Monoclonales/metabolismo , Células CHO , Línea Celular Tumoral , Cricetulus , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias/sangre , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA