Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958798

RESUMEN

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Asunto(s)
Colon/fisiología , Motilidad Gastrointestinal , Macrófagos/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Colon/fisiopatología , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Dinoprostona/análisis , Dinoprostona/metabolismo , Femenino , Mucosa Gástrica/citología , Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Contracción Muscular , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
2.
J Biol Chem ; 298(5): 101816, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35278430

RESUMEN

Jumonji domain-containing protein-3 (JMJD3), a histone H3 lysine 27 (H3K27) demethylase, promotes endothelial regeneration, but its function in neointimal hyperplasia (NIH) of arteriovenous fistulas (AVFs) has not been explored. In this study, we examined the contribution of endothelial JMJD3 to NIH of AVFs and the mechanisms underlying JMJD3 expression during kidney failure. We found that endothelial JMJD3 expression was negatively associated with NIH of AVFs in patients with kidney failure. JMJD3 expression in endothelial cells (ECs) was also downregulated in the vasculature of chronic kidney disease (CKD) mice. In addition, specific knockout of endothelial JMJD3 delayed EC regeneration, enhanced endothelial mesenchymal transition, impaired endothelial barrier function as determined by increased Evans blue staining and inflammatory cell infiltration, and accelerated neointima formation in AVFs created by venous end to arterial side anastomosis in CKD mice. Mechanistically, JMJD3 expression was downregulated via binding of transforming growth factor beta 1-mediated Hes family transcription factor Hes1 to its gene promoter. Knockdown of JMJD3 enhanced H3K27 methylation, thereby inhibiting transcriptional activity at promoters of EC markers and reducing migration and proliferation of ECs. Furthermore, knockdown of endothelial JMJD3 decreased endothelial nitric oxide synthase expression and nitric oxide production, leading to the proliferation of vascular smooth muscle cells. In conclusion, we demonstrate that decreased expression of endothelial JMJD3 impairs EC regeneration and function and accelerates neointima formation in AVFs. We propose increasing the expression of endothelial JMJD3 could represent a new strategy for preventing endothelial dysfunction, attenuating NIH, and improving AVF patency in patients with kidney disease.


Asunto(s)
Fístula Arteriovenosa , Histona Demetilasas con Dominio de Jumonji/genética , Insuficiencia Renal Crónica , Animales , Fístula Arteriovenosa/genética , Fístula Arteriovenosa/patología , Regulación hacia Abajo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Hiperplasia/genética , Hiperplasia/patología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Neointima/genética
3.
Int J Mol Sci ; 24(14)2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37511512

RESUMEN

Mesenchymal stem cell (MSC)-seeded polymeric perivascular wraps have been shown to enhance arteriovenous fistula (AVF) maturation. However, the wraps' radiolucency makes their placement and integrity difficult to monitor. Through electrospinning, we infused gold nanoparticles (AuNPs) into polycaprolactone (PCL) wraps to improve their radiopacity and tested whether infusion affects the previously reported beneficial effects of the wraps on the AVF's outflow vein. Sprague Dawley rat MSCs were seeded on the surface of the wraps. We then compared the effects of five AVF treatments-no perivascular wrap (i.e., control), PCL wrap, PCL + MSC wrap, PCL-Au wrap, and PCL-Au + MSC wrap-on AVF maturation in a Sprague Dawley rat model of chronic kidney disease (n = 3 per group). Via micro-CT, AuNP-infused wraps demonstrated a significantly higher radiopacity compared to that of the wraps without AuNPs. Wraps with and without AuNPs equally reduced vascular stenoses, as seen via ultrasonography and histomorphometry. In the immunofluorescence analysis, representative MSC-seeded wraps demonstrated reduced neointimal staining for markers of infiltration with smooth muscle cells (α-SMA), inflammatory cells (CD45), and fibroblasts (vimentin) compared to that of the control and wraps without MSCs. In conclusion, AuNP infusion allows in vivo monitoring via micro-CT of MSC-seeded polymeric wraps over time, without compromising the benefits of the wrap for AVF maturation.


Asunto(s)
Fístula Arteriovenosa , Células Madre Mesenquimatosas , Nanopartículas del Metal , Ratas , Animales , Oro , Ratas Sprague-Dawley , Implantes Absorbibles , Fístula Arteriovenosa/terapia
4.
J Transl Med ; 20(1): 543, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36419038

RESUMEN

BACKGROUND: Arteriovenous fistula (AVF) maturation is a process involving remodeling of venous arm of the AVFs. It is a challenge to balance adaptive AVF remodeling and neointima formation. In this study we temporally controlled Notch activation to promote AVF maturation while avoiding neointima formation. METHODS: Temporal Notch activation was controlled by regulating the expression of Notch transcription factor, RBP-Jκ, or dnMAML1 (dominant negative MAML2) in vascular smooth muscle cells (VSMCs). AVF mouse model was created and VSMC phenotype dynamic changes during AVF remodeling were determined. RESULTS: Activated Notch was found in the nuclei of neointimal VSMCs in AVFs from uremic mice. We found that the VSMCs near the anastomosis became dedifferentiated and activated after AVF creation. These dedifferentiated VSMCs regained smooth muscle contractile markers later during AVF remodeling. However, global or VSMC-specific KO of RBP-Jκ at early stage (before or 1 week after AVF surgery) blocked VSMC differentiation and neointima formation in AVFs. These un-matured AVFs showed less intact endothelium and increased infiltration of inflammatory cells. Consequently, the VSMC fate in the neointima was completely shut down, leading to an un-arterialized AVF. In contrast, KO of RBP-Jκ at late stage (3 weeks after AVF surgery), it could not block neointima formation and vascular stenosis. Inhibition of Notch activation at week 1 or 2, could maintain VSMC contractile markers expression and facilitate AVF maturation. CONCLUSIONS: This work uncovers the molecular and cellular events in each segment of AVF remodeling and found that neither sustained increasing nor blocking of Notch signaling improves AVF maturation. It highlights a novel strategy to improve AVF patency: temporally controlled Notch activation can achieve a balance between adaptive AVF remodeling and neointima formation to improve AVF maturation. TRANSLATIONAL PERSPECTIVE: Adaptive vascular remodeling is required for AVF maturation. The balance of wall thickening of the vein and neointima formation in AVF determines the fate of AVF function. Sustained activation of Notch signaling in VSMCs promotes neointima formation, while deficiency of Notch signaling at early stage during AVF remodeling prevents VSMC accumulation and differentiation from forming a functional AVFs. These responses also delay EC regeneration and impair EC barrier function with increased inflammation leading to failed vascular remodeling of AVFs. Thus, a strategy to temporal regulate Notch activation will improve AVF maturation.


Asunto(s)
Fístula Arteriovenosa , Derivación Arteriovenosa Quirúrgica , Animales , Ratones , Neointima , Remodelación Vascular , Miocitos del Músculo Liso
5.
Arterioscler Thromb Vasc Biol ; 39(1): 48-62, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30580569

RESUMEN

Objective- Hypoxic pulmonary hypertension (HPH) is characterized by proliferative vascular remodeling. Abnormal pulmonary artery smooth muscle cells proliferation and endothelial dysfunction are the primary cellular bases of vascular remodeling. AQP1 (aquaporin-1) is regulated by oxygen level and has been observed to play a role in the proliferation and migration of pulmonary artery smooth muscle cells. The role of AQP1 in HPH pathogenesis has not been directly determined to date. To determine the possible roles of AQP1 in the pathogenesis of HPH and explore its possible mechanisms. Approach and Results- Aqp1 knockout mice were used, and HPH model was established in this study. Primary pulmonary artery smooth muscle cells, primary mouse lung endothelial cells, and lung tissue sections from HPH model were used. Immunohistochemistry, immunofluorescence and Western blot, cell cycle, apoptosis, and migration analysis were performed in this study. AQP1 expression was upregulated by chronic hypoxia exposure, both in pulmonary artery endothelia and medial smooth muscle layer of mice. Aqp1 deficiency attenuated the elevation of right ventricular systolic pressures and mitigated pulmonary vascular structure remodeling. AQP1 deletion reduced abnormal cell proliferation in pulmonary artery and accompanied with accumulation of HIF (hypoxia-inducible factor). In vitro, Aqp1 deletion reduced hypoxia-induced proliferation, apoptosis resistance, and migration ability of primary cultured pulmonary artery smooth muscle cells and repressed HIF-1α protein stability. Furthermore, Aqp1 deficiency protected lung endothelial cells from apoptosis in response to hypoxic injury. Conclusions- Our data showed that Aqp1 deficiency could attenuate hypoxia-induced vascular remodeling in the development of HPH. AQP1 may be a potential target for pulmonary hypertension treatment.


Asunto(s)
Acuaporina 1/fisiología , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Animales , Acuaporina 1/genética , Células Cultivadas , Ciclina D1/fisiología , Hipertensión Pulmonar/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Remodelación Vascular
6.
Kidney Int ; 95(6): 1347-1358, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30799025

RESUMEN

Neointima formation is a major contributor to arteriovenous fistula (AVF) failure. We have previously shown that activation of the Notch signaling pathway contributes to neointima formation by promoting the migration of vascular smooth muscle cells (VSMCs) into the venous anastomosis. In the current study we investigated the mechanisms underlying the dedifferentiation and migration of VSMCs, and in particular the role of bone marrow-derived fibroblast specific protein 1 (FSP-1)+ cells, another cell type found in models of vascular injury. Using VSMC-specific reporter mice, we found that most of the VSMCs participating in AVF neointima formation originated from dedifferentiated VSMCs. We also observed infiltration of bone marrow-derived FSP-1+ cells into the arterial anastomosis where they could interact with VSMCs. In vitro, conditioned media from FSP-1+ cells stimulated VSMC proliferation and phenotype switching. Activated Notch signaling transformed FSP-1+ cells into type I macrophages and stimulated secretion of cytokines and growth factors. Pretreatment with a Notch inhibitor or knockout of the canonical downstream factor RBP-Jκ in bone marrow-derived FSP1+ cells decreased FSP1+ cell infiltration into murine AVFs, attenuating VSMC dedifferentiation and neointima formation. Our results suggest that targeting Notch signaling could provide a new therapeutic strategy to improve AVF patency.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Miocitos del Músculo Liso/patología , Neointima/patología , Receptores Notch/metabolismo , Diálisis Renal/efectos adversos , Animales , Desdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/etiología , Neointima/prevención & control , Cultivo Primario de Células , Receptores Notch/antagonistas & inhibidores , Diálisis Renal/métodos , Insuficiencia Renal Crónica/terapia , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal/efectos de los fármacos , Grado de Desobstrucción Vascular/efectos de los fármacos
7.
J Am Soc Nephrol ; 29(2): 505-517, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29127112

RESUMEN

Neointima formation is the leading cause of arteriovenous fistula (AVF) failure. We have shown that CKD accelerates this process by transforming the vascular smooth muscle cells (SMCs) lining the AVF from a contractile to the synthetic phenotype. However, the underlying mechanisms affecting this transformation are not clear. Previous studies have shown that the α-class glutathione transferase isozymes have an important role in regulating 4-hydroxynonenal (4-HNE)-mediated proliferative signaling of cells. Here, using both the loss- and gain-of-function approaches, we investigated the role of glutathione S-transferase α4 (GSTA4) in modulating cellular 4-HNE levels for the transformation and proliferation of SMCs. Compared with non-CKD controls, mice with CKD had downregulated expression of GSTA4 at the mRNA and protein levels, with concomitant increase in 4-HNE in arteries and veins. This effect was associated with upregulated phosphorylation of MAPK signaling pathway proteins in proliferating SMCs. Overexpressing GSTA4 blocked 4-HNE-induced SMC proliferation. Additionally, inhibitors of MAPK signaling inhibited the 4-HNE-induced responses. Compared with wild-type mice, mice lacking GSTA4 exhibited increased CKD-induced neointima formation in AVF. Transient expression of an activated form of GSTA4, achieved using a combined Tet-On/Cre induction system in mice, lowered levels of 4-HNE and reduced the proliferation of SMCs. Together, these results demonstrate the critical role of GSTA4 in blocking CKD-induced neointima formation and AVF failure.


Asunto(s)
Aldehídos/metabolismo , Glutatión Transferasa/genética , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Neointima/patología , Insuficiencia Renal Crónica/fisiopatología , Túnica Íntima/patología , Animales , Arterias/metabolismo , Derivación Arteriovenosa Quirúrgica , Proliferación Celular , Regulación hacia Abajo , Expresión Génica , Glutatión Transferasa/metabolismo , Hiperplasia/genética , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Fenotipo , Fosforilación/genética , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/terapia , Venas/metabolismo
8.
J Allergy Clin Immunol ; 141(2): 608-619.e7, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28807414

RESUMEN

BACKGROUND: Chronic itch is a highly debilitating symptom that underlies many medical disorders with no universally effective treatments. Although unique neuronal signaling cascades in the sensory ganglia and spinal cord have been shown to critically promote the pathogenesis of chronic itch, the role of skin-associated cells remains poorly understood. OBJECTIVE: We sought to examine the cutaneous mechanisms underlying transient receptor potential vanilloid 4 (TRPV4)-mediated allergic and nonallergic chronic itch. METHODS: Expression of TRPV4 in chronic itch and healthy control skin preparations was examined by using real-time RT-PCR. Trpv4eGFP mice were used to study the expression and function of TRPV4 in the skin by means of immunofluorescence staining, flow cytometry, calcium imaging, and patch-clamp recordings. Genetic and pharmacologic approaches were used to examine the role and underlying mechanisms of TRPV4 in mouse models of dry skin-associated chronic itch and spontaneous scratching associated with squaric acid dibutylester-induced allergic contact dermatitis. RESULTS: TRPV4 is selectively expressed by dermal macrophages and epidermal keratinocytes in mice. Lineage-specific deletion of TRPV4 in macrophages and keratinocytes reduces allergic and nonallergic chronic itch in mice, respectively. Importantly, TRPV4 expression is significantly increased in skin biopsy specimens from patients with chronic idiopathic pruritus in comparison with skin from healthy control subjects. Moreover, TRPV4-dependent chronic itch requires 5-hydroxytryptamine (5-HT) signaling secondary to activation of distinct 5-HT receptors in mice with allergic and those with nonallergic chronic itch conditions. CONCLUSION: Our study reveals previously unrecognized mechanisms by which TRPV4-expressing epithelial and immune cells in the skin critically and dynamically mediate chronic itch and unravels novel targets for therapeutics in the setting of chronic itch.


Asunto(s)
Dermatitis Alérgica por Contacto/inmunología , Dermis/inmunología , Regulación de la Expresión Génica/inmunología , Queratinocitos/inmunología , Macrófagos/inmunología , Prurito/inmunología , Canales Catiónicos TRPV/inmunología , Animales , Enfermedad Crónica , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/patología , Dermis/patología , Femenino , Regulación de la Expresión Génica/genética , Humanos , Queratinocitos/patología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Prurito/genética , Prurito/patología , Canales Catiónicos TRPV/genética
9.
Cell Physiol Biochem ; 49(3): 985, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196283

RESUMEN

BACKGROUND/AIMS: Neointimal hyperplasia is responsible for stenosis, which requires corrective vascular surgery, and is also a major morphological feature of many cardiovascular diseases. This hyperplasia involves the endothelial-to-mesenchymal transition (EndMT). We investigated whether integrin ß3 can modulate the EndMT, as well as its underlying mechanism. METHODS: Integrin ß3 was overexpressed or knocked down in human umbilical vein endothelial cells (HUVECs). The expression of endothelial markers and mesenchymal markers was determined by real-time reverse transcription PCR (RT-PCR), immunofluorescence staining, and western blot analysis. Notch signaling pathway components were detected by real-time RT-PCR and western blot analysis. Cell mobility was evaluated by wound-healing, Transwell, and spreading assays. Fibroblast-specific protein 1 (FSP-1) promoter activity was determined by luciferase assay. RESULTS: Transforming growth factor (TGF)-ß1 treatment or integrin ß3 overexpression significantly promoted the EndMT by downregulating VE-cadherin and CD31 and upregulating smooth muscle actin α and FSP-1 in HUVECs, and by enhancing cell migration. Knockdown of integrin ß3 reversed these effects. Notch signaling was activated after TGF-ß1 treatment of HUVECs. Knockdown of integrin ß3 suppressed TGF-ß1-induced Notch activation and expression of the Notch downstream target FSP-1. CONCLUSION: Integrin ß3 may promote the EndMT in HUVECs through activation of the Notch signaling pathway.


Asunto(s)
Transición Epitelial-Mesenquimal , Integrina beta3/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Actinas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/patología , Cadherinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta3/química , Integrina beta3/genética , Persona de Mediana Edad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína de Unión al Calcio S100A4 , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos
10.
J Immunol ; 196(4): 1874-81, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773152

RESUMEN

Bone marrow-derived monocyte-to-fibroblast transition is a key step in renal fibrosis pathogenesis, which is regulated by the inflammatory microenvironment. However, the mechanism by which the inflammatory microenvironment regulates this transition is not fully understood. In this study, we examined how the CD8(+) T cell/IFN-γ microenvironment regulates the monocyte-to-fibroblast transition in renal fibrosis. Genetic ablation of CD8 promoted a monocyte-to-fibroblast transition and increased renal interstitial fibrosis, whereas reconstitution of CD8 knockout (KO) mice with CD8(+) T cells decreased fibrosis. However, depletion of CD4(+) T cells in CD8 KO mice also reduced fibrosis. To elucidate the role of CD4(+) T cells in mediating CD8-regulated monocyte-to-fibroblast transition, CD4(+) T cells were isolated from obstructed kidneys of CD8 KO or wild-type mice. CD4(+) T cells isolated from CD8 KO obstructed kidney expressed more IL-4 and GATA3 and less IFN-γ and T-bet and showed increased monocyte-to-fibroblast transition in vitro compared with those isolated from wild-type obstructed kidney. To examine the role of IFN-γ-expressing CD8(+) T cells, we reconstituted CD8 KO mice with CD8(+) T cells isolated from IFN-γ KO mice. The IFN-γ KO CD8(+) cells had no effect on IL-4, GATA3, IFN-γ, and T-bet mRNA expression in obstructed kidneys or renal fibrosis. Taken together, our findings identify the axis of CD8(+) T cells and IFN-γ-CD4(+) T cells as an important microenvironment for the monocyte-to-fibroblast transition, which negatively regulates renal fibrosis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/fisiología , Fibroblastos/citología , Riñón/patología , Animales , Western Blotting , Células de la Médula Ósea/citología , Separación Celular , Modelos Animales de Enfermedad , Fibrosis/patología , Citometría de Flujo , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología
11.
J Am Soc Nephrol ; 28(11): 3278-3290, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28768710

RESUMEN

In damaged kidneys, increased extracellular matrix (ECM) and tissue stiffness stimulate kidney fibrosis through incompletely characterized molecular mechanisms. The transcriptional coactivators yes-associated protein (Yap) and transcriptional coactivator with PDZ-binding motif (Taz) function as mechanosensors in cancer cells and have been implicated in the regulation of myofibroblasts in the kidney. We hypothesized that the development of kidney fibrosis depends on Yap-induced activation and proliferation of kidney fibroblasts. In mice, Yap expression increased in renal fibroblasts after unilateral ureteral obstruction (UUO), in association with worsening of interstitial fibrosis. In cultured fibroblasts, inhibition of Yap/Taz signaling blocked TGF-ß1-induced fibroblast-to-myofibroblast transformation and ECM production, whereas constitutive activation of Yap promoted fibroblast transformation and ECM production even in the absence of TGF-ß1. Moreover, in the absence of TGF-ß1, fibroblasts seeded on a stiffened ECM transformed into myofibroblasts in a process dependent on the activation of Yap. In mice with UUO, the Yap inhibitor verteporfin reduced interstitial fibrosis. Furthermore, Gli1+ cell-specific knockout of Yap/Taz in mice suppressed UUO-induced ECM deposition, myofibroblast accumulation, and interstitial fibrosis. In a UUO-release model, induction of Gli1+ cell-specific Yap/Taz knockout partially reversed the development of interstitial fibrosis. Thus, in the kidney, Yap is a tissue mechanosensor that can be activated by ECM and transforms fibroblasts into myofibroblasts; the interaction of Yap/Taz and ECM forms a feed-forward loop resulting in kidney fibrosis. Identifying mechanisms that interrupt this profibrotic cycle could lead to the development of anti-fibrosis therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Riñón/patología , Miofibroblastos , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Matriz Extracelular , Fibroblastos/fisiología , Fibrosis/etiología , Eliminación de Gen , Masculino , Ratones , Miofibroblastos/metabolismo , Fosfoproteínas/genética , Transactivadores , Obstrucción Ureteral/patología , Proteínas Señalizadoras YAP , Proteína con Dedos de Zinc GLI1/biosíntesis
12.
J Am Soc Nephrol ; 27(9): 2797-808, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26880799

RESUMEN

Muscle proteolysis in CKD is stimulated when the ubiquitin-proteasome system is activated. Serum glucocorticoid-regulated kinase 1 (SGK-1) is involved in skeletal muscle homeostasis, but the role of this protein in CKD-induced muscle wasting is unknown. We found that, compared with muscles from healthy controls, muscles from patients and mice with CKD express low levels of SGK-1. In mice, SGK-1-knockout (SGK-1-KO) induced muscle loss that correlated with increased expression of ubiquitin E3 ligases known to facilitate protein degradation by the ubiquitin-proteasome, and CKD substantially aggravated this response. SGK-1-KO also altered the phosphorylation levels of transcription factors FoxO3a and Smad2/3. In C2C12 muscle cells, expression of dominant negative FoxO3a or knockdown of Smad2/3 suppressed the upregulation of E3 ligases induced by loss of SGK-1. Additionally, SGK-1 overexpression increased the level of phosphorylated N-myc downstream-regulated gene 1 protein, which directly interacted with and suppressed the phosphorylation of Smad2/3. Overexpression of SGK-1 in wild-type mice with CKD had similar effects on the phosphorylation of FoxO3a and Smad2/3 and prevented CKD-induced muscle atrophy. Finally, mechanical stretch of C2C12 muscle cells or treadmill running of wild-type mice with CKD stimulated SGK-1 production, and treadmill running inhibited proteolysis in muscle. These protective responses were absent in SGK-1-KO mice. Thus, SGK-1 could be a mechanical sensor that mediates exercise-induced improvement in muscle wasting stimulated by CKD.


Asunto(s)
Proteína Forkhead Box O3/fisiología , Proteínas Inmediatas-Precoces/fisiología , Atrofia Muscular/enzimología , Atrofia Muscular/etiología , Proteínas Serina-Treonina Quinasas/fisiología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/enzimología , Proteína Smad2/fisiología , Proteína smad3/fisiología , Animales , Ratones
13.
Am J Pathol ; 185(5): 1234-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25783760

RESUMEN

Insulin-like growth factor-1 receptor (IGF-1R) can regulate vascular homeostasis and endothelial function. We studied the role of IGF-1R in oxidative stress-induced endothelial dysfunction. Unilateral ureteral obstruction (UUO) was performed in wild-type (WT) mice and mice with endothelial cell (EC)-specific IGF-1R knockout (KO). After UUO in endothelial IGF-1R KO mice, endothelial barrier dysfunction was more severe than in WT mice, as seen by increased inflammatory cell infiltration and vascular endothelial (VE)-cadherin phosphorylation. UUO in endothelial IGF-1R KO mice increased interstitial fibroblast accumulation and enhanced extracellular protein deposition as compared with the WT mice. Endothelial barrier function measured by transendothelial migration in response to hydrogen peroxide (H2O2) was impaired in ECs. Silencing IGF-1R enhanced the influence of H2O2 in disrupting the VE-protein tyrosine phosphatase/VE-cadherin interaction. Overexpression of IGF-1R suppressed H2O2-induced endothelial barrier dysfunction. Furthermore, by using the piggyBac transposon system, we expressed IGF-1R in VE cells in mice. The expression of IGF-1R in ECs also suppressed the inflammatory cell infiltration and renal fibrosis induced by UUO. IGF-1R KO in the VE-cadherin lineage of bone marrow cells had no significant effect on the UUO-induced fibrosis, as compared with control mice. Our results indicate that IGF-1R in the endothelium maintains the endothelial barrier function by stabilization of the VE-protein tyrosine phosphatase/VE-cadherin complex. Decreased expression of IGF-1R impairs endothelial function and increases the fibrosis of kidney disease.


Asunto(s)
Células Endoteliales/metabolismo , Receptor IGF Tipo 1/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , Insuficiencia Renal Crónica/patología , Transfección , Venas Umbilicales , Obstrucción Ureteral
14.
Arterioscler Thromb Vasc Biol ; 35(3): 607-15, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25614287

RESUMEN

OBJECTIVE: Neointima formation is associated with stenosis and subsequent thrombosis in arteriovenous grafts (AVGs). A role of integrin ß3 in the neointima formation of AVGs remains poorly understood. APPROACH AND RESULTS: In integrin ß3(-/-) mice, we found significantly accelerated occlusion of AVGs compared with the wild-type mice. This is caused by the development of neointima and lack of endothelial regeneration. The latter is a direct consequence of impaired functions of circulating angiogenic cells (CACs) and platelets in integrin ß3(-/-) mice. Evidence suggests the involvement of platelet regulating CAC homing to and differentiation at graft sites via transforming growth factor-ß1 and Notch signaling pathway. First, CACs deficient of integrin ß3 impaired adhesion activity toward exposed subendothelium. Second, platelets from integrin ß3(-/-) mice failed to sufficiently stimulate CACs to differentiate into mature endothelial cells. Finally, we found that transforming growth factor-ß1 level was increased in platelets from integrin ß3(-/-) mice and resulted in enhanced Notch1 activation in CACs in AVGs. These results demonstrate that integrin ß3 is critical for endothelial cell homing and differentiation. The increased transforming growth factor-ß1 and Notch1 signaling mediates integrin ß3(-/-)-induced AVG occlusion. This accelerated occlusion of AVGs was reversed in integrin ß3(-/-) mice transplanted with the bone marrow from wild-type mice. CONCLUSIONS: Our results suggest that boosting integrin ß3 function in the endothelial cells and platelets could prevent neointima and thrombosis in AVGs.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Plaquetas/metabolismo , Arteria Carótida Común/cirugía , Proliferación Celular , Células Endoteliales/metabolismo , Oclusión de Injerto Vascular/metabolismo , Integrina beta3/metabolismo , Regeneración , Venas Cavas/cirugía , Animales , Trasplante de Médula Ósea , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Constricción Patológica , Modelos Animales de Enfermedad , Células Endoteliales/patología , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/patología , Integrina beta3/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neointima , Receptor Notch1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Venas Cavas/metabolismo , Venas Cavas/patología
15.
Kidney Int ; 88(3): 490-502, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25786100

RESUMEN

A major factor contributing to failure of arteriovenous fistulas (AVFs) is migration of smooth muscle cells into the forming neointima. To identify the source of smooth muscle cells in neointima, we created end-to-end AVFs by anastomosing the common carotid artery to the jugular vein and studied neural crest-derived smooth muscle cells from the carotid artery, which are Wnt1-positive during development. In Wnt1-cre-GFP mice, smooth muscle cells in the carotid artery but not the jugular vein are labeled with GFP. About half of the cells were GFP-positive in the neointima, indicating their migration from the carotid artery to the jugular vein in AVFs created in these mice. As fibroblast-specific protein-1 (FSP-1) regulates smooth muscle cell migration, we examined FSP-1 in failed AVFs and polytetrafluoroethylene grafts from patients with end-stage kidney disease or from AVFs in mice with chronic kidney disease. In smooth muscle cells of AVFs or polytetrafluoroethylene grafts, FSP-1 and activation of Notch1 are present. In smooth muscle cells, Notch1 increased RBP-Jκ transcription factor activity and RBP-Jκ stimulated FSP-1 expression. Conditional knockout of RBP-Jκ in smooth muscle cells or general knockout of FSP-1 suppressed neointima formation in AVFs in mice. Thus, the artery of AVFs is the major source of smooth muscle cells during neointima formation. Knockout of RBP-Jκ or FSP-1 ameliorates neointima formation and might improve AVF patency during long-term follow-up.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Implantación de Prótesis Vascular/efectos adversos , Movimiento Celular , Oclusión de Injerto Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Receptor Notch1/metabolismo , Insuficiencia Renal Crónica/terapia , Anciano , Animales , Implantación de Prótesis Vascular/instrumentación , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Arteria Carótida Común/cirugía , Células Cultivadas , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/patología , Oclusión de Injerto Vascular/fisiopatología , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Venas Yugulares/cirugía , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Modelos Animales , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Músculo Liso Vascular/cirugía , Miocitos del Músculo Liso/patología , Interferencia de ARN , Receptor Notch1/genética , Diálisis Renal , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Proteínas S100/metabolismo , Transducción de Señal , Transfección , Grado de Desobstrucción Vascular
16.
J Am Soc Nephrol ; 25(4): 773-83, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24480830

RESUMEN

Neointima formation causes the failure of 60% of arteriovenous fistulas (AVFs) within 2 years. Neointima-forming mechanisms are controversial but possibly linked to excess proinflammatory responses and dysregulated Notch signaling. To identify how AVFs fail, we anastomosed the carotid artery to the internal jugular vein in normal and uremic mice and compared these findings with those in failed AVFs from patients with ESRD. Endothelial cells (ECs) of AVFs in uremic mice or patients expressed mesenchymal markers (FSP-1 and/or α-SMA) and exhibited increased expression and nuclear localization of Notch intracellular domain compared with ECs of AVFs in pair-fed control mice. Furthermore, expression of VE-Cadherin decreased, whereas expression of Notch1 and -4, Notch ligands, the downstream transcription factor of Notch, RBP-Jκ, and Notch target genes increased in ECs of AVFs in uremic mice. In cultured ECs, ectopic expression of Notch ligand or treatment with TGF-ß1 triggered the expression of mesenchymal markers and induced endothelial cell barrier dysfunction, both of which were blocked by Notch inhibition or RBP-Jκ knockout. Furthermore, Notch-induced defects in barrier function, invasion of inflammatory cells, and neointima formation were suppressed in mice with heterozygous knockdown of endothelial-specific RBP-Jκ. These results suggest that increased TGF-ß1, a complication of uremia, activates Notch in endothelial cells of AVFs, leading to accelerated neointima formation and AVF failure. Suppression of Notch activation could be a strategy for improving AFV function in uremia.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Células Endoteliales/fisiología , Receptores Notch/fisiología , Insuficiencia Renal Crónica/fisiopatología , Actinas/análisis , Anciano , Animales , Proteínas de Unión al Calcio/análisis , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Masculino , Ratones , Persona de Mediana Edad , Neointima , Insuficiencia Renal Crónica/patología , Proteína de Unión al Calcio S100A4 , Transducción de Señal , Factor de Crecimiento Transformador beta1/fisiología
17.
Basic Res Cardiol ; 109(5): 431, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25107324

RESUMEN

Accumulation of smooth muscle cells (SMC) results in neointima formation in injured vessels. Two graft models consisting of vein and artery grafts were created by anastomosing common carotid arteries to donor vessels. To identify the origin of the neointima cells from anastomosed arteries, we use Wnt1-Cre/reporter mice to label and track SMCs in the common carotid artery. The contribution of SMCs in the neighboring arteries to neointima formation was studied. On evaluating the artery grafts after 1 month, >90 % of the labeled neointima cells were found to have originated from the anastomosing host arteries. Most of the neointima cells were also smooth muscle α-actin positive (SMA-α(+)) and expressed the smooth muscle myosin heavy chain (SMMHC), the SMC terminal differentiation marker. In vein grafts, about 60 % SMA-α-positive cells were from anastomosing arteries. Bone marrow cells did not contribute to neointima SMCs in vein grafts, but did co-stain with markers of inflammatory cells. Wnt1 expression was not detected in the neointima cells in the vein or artery grafts, or the injured femoral arteries. Neointima SMCs showed the synthetic phenotype and were positively labeled with BrdU in vitro and in vivo. Treatment with the IGF-1 receptor inhibitor suppressed SMC proliferation and neointima formation in vein grafts. Our results indicate that SMCs from the neighboring artery are predominantly present in the neointima formed in both vein and artery grafts and that Wnt1-Cre mice can be used to explore the role of SMCs originating from neighboring vessels in vascular remodeling.


Asunto(s)
Arteria Carótida Común/citología , Arteria Carótida Común/trasplante , Puente de Arteria Coronaria/efectos adversos , Miocitos del Músculo Liso/citología , Neointima/patología , Venas Cavas/trasplante , Anastomosis Quirúrgica/efectos adversos , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/patología
18.
Circ Res ; 110(2): 230-40, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22116816

RESUMEN

RATIONALE: Fibroblast-specific protein 1 (FSP-1) plays multiple roles in promoting cell proliferation and motility. Increased FSP-1 expression in smooth muscle cells (SMCs) has been associated with their enhanced proliferation. OBJECTIVE: To study how FSP-1 contributes to neointima formation of vein grafts. METHODS: Arteriovenous grafts were created in wild-type or FSP-1-GFP mice (green fluorescent protein expression regulated by FSP-1 promoter). The effects of FSP-1 on bone marrow (BM) cell migration and on SMC proliferation were studied in vivo and in vitro. RESULTS: On creation of a vein graft, there was rapid deposition of platelets on the denuded surface leading to secretion of the chemokine stromal cell-derived factor-1α (SDF-1α). This was followed by recruitment of BM-derived cells expressing the SDF-1α receptor CXCR4; homing of FSP-1-positive cells was found to be dependent on platelet-derived SDF-1α. FSP-1 was expressed in 8% of the BM cells, and 20% of these express CD45; 85% of FSP-1-positive cells express CD11b. We found that the FSP-1-positive cells migrated into the vein graft in a Rac-1-dependent fashion. FSP-1 expression was also found to stimulate proliferation of SMCs through a MEK5-ERK5 signaling pathway that can be suppressed by a dominant-negative Rac1. Consequently, knocking down FSP-1 expression in BM cells prevented neointimal formation. CONCLUSIONS: BM-derived FSP-1(+) cells enhance neointima formation through an increase in transendothelial invasion with stimulation of SMC proliferation. The Rac1 and ERK5 signaling cascade mediate FSP-1-induced responses in SMCs and BM cells. This novel pathophysiology suggests a new therapeutic target, FSP-1, for preventing the development of neointima in vein grafts.


Asunto(s)
Células de la Médula Ósea/metabolismo , Proliferación Celular , Terapia Genética , Proteínas S100/metabolismo , Túnica Íntima/trasplante , Injerto Vascular , Vena Cava Inferior/trasplante , Animales , Plaquetas/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Quimiocina CXCL12/metabolismo , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hiperplasia , Mediadores de Inflamación/metabolismo , MAP Quinasa Quinasa 5/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neuropéptidos/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Transducción de Señal , Migración Transendotelial y Transepitelial , Túnica Íntima/metabolismo , Túnica Íntima/patología , Vena Cava Inferior/metabolismo , Vena Cava Inferior/patología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
19.
ACS Appl Mater Interfaces ; 16(26): 33159-33168, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38912610

RESUMEN

In the context of arteriovenous fistula (AVF) failure, local delivery enables the release of higher concentrations of drugs that can suppress neointimal hyperplasia (NIH) while reducing systemic adverse effects. However, the radiolucency of polymeric delivery systems hinders long-term in vivo surveillance of safety and efficacy. We hypothesize that using a radiopaque perivascular wrap to deliver anti-NIH drugs could enhance AVF maturation. Through electrospinning, we fabricated multifunctional perivascular polycaprolactone (PCL) wraps loaded with bismuth nanoparticles (BiNPs) for enhanced radiologic visibility and drugs that can attenuate NIH─rosuvastatin (Rosu) and rapamycin (Rapa). The following groups were tested on the AVFs of a total of 24 Sprague-Dawley rats with induced chronic kidney disease: control (i.e., without wrap), PCL-Bi (i.e., wrap with BiNPs), PCL-Bi-Rosu, and PCL-Bi-Rapa. We found that BiNPs significantly improved the wraps' radiopacity without affecting biocompatibility. The drug release profiles of Rosu (hydrophilic drug) and Rapa (hydrophobic drug) differed significantly. Rosu demonstrated a burst release followed by gradual tapering over 8 weeks, while Rapa demonstrated a gradual release similar to that of the hydrophobic BiNPs. In vivo investigations revealed that both drug-loaded wraps can reduce vascular stenosis on ultrasonography and histomorphometry, as well as reduce [18F]Fluorodeoxyglucose uptake on positron emission tomography. Immunohistochemical studies revealed that PCL-Bi-Rosu primarily attenuated endothelial dysfunction and hypoxia in the neointimal layer, while PCL-Bi-Rapa modulated hypoxia, inflammation, and cellular proliferation across the whole outflow vein. In summary, the controlled delivery of drugs with different properties and mechanisms of action against NIH through a multifunctional, radiopaque perivascular wrap can improve imaging and histologic parameters of AVF maturation.


Asunto(s)
Bismuto , Ratas Sprague-Dawley , Rosuvastatina Cálcica , Sirolimus , Animales , Ratas , Sirolimus/química , Sirolimus/farmacología , Rosuvastatina Cálcica/química , Rosuvastatina Cálcica/farmacología , Rosuvastatina Cálcica/farmacocinética , Bismuto/química , Bismuto/farmacología , Poliésteres/química , Masculino , Fístula Arteriovenosa/patología , Nanopartículas del Metal/química , Neointima/patología , Nanopartículas/química , Humanos , Liberación de Fármacos
20.
J Biol Chem ; 287(9): 6177-86, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22210777

RESUMEN

Following injury, skeletal muscle achieves repair by a highly coordinated, dynamic process resulting from interplay among numerous inflammatory, growth factors and myogenic regulators. To identify genes involved in muscle regeneration, we used a microarray analysis; there was a significant increase in the expression of a group of integrin genes. To verify these results, we used RT-PCR and Western blotting and found that 12 integrins were up-regulated from 3 h to 15 days following injury. Following muscle injury, integrin-ß3 was initially expressed, mainly in macrophages. In integrin-ß3 global KO mice, the expression of myogenic genes was decreased and muscle regeneration was impaired, whereas fibrosis was enhanced versus events in wild type (WT) mice. The mechanism for these responses in integrin-ß3 KO mice included an infiltration of macrophages that were polarized into the M2 phenotype. These macrophages produced more TGF-ß1 and increased TGF-ß1/Smad signaling. In vitro, we confirmed that M2 macrophages lacking integrin-ß3 produced more TGF-ß1. Furthermore, transplantation of bone marrow cells from integrin-ß3 KO mice into WT mice led to suppression of the infiltration and accumulation of macrophages into injured muscles. There was also impaired muscle regeneration with an increase in muscle fibrosis. Our results demonstrate that integrin-ß3 plays a fundamental role in muscle regeneration through a regulation of macrophage infiltration and polarization leading to suppressed TGF-ß1 production. This promotes efficient muscle regeneration. Thus, an improvement in integrin-ß3 function could stimulate muscle regeneration.


Asunto(s)
Polaridad Celular/fisiología , Integrina beta3/metabolismo , Macrófagos/fisiología , Músculo Esquelético , Regeneración/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Fibrosis , Expresión Génica/fisiología , Integrina beta3/genética , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/fisiología , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Proteína MioD/genética , Miogenina/genética , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA