Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Carcinogenesis ; 43(4): 360-370, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34965300

RESUMEN

Development of cancer, including renal cancer, is a major problem in immunosuppressed patients. The mTOR inhibitor Rapamycin (RAPA) is used as an immunosuppressive agent in patients with organ transplants and other immunological disorders; and it also has antitumorigenic potential. However, long-term use of RAPA causes reactivation of Akt, and ultimately leads to enhanced tumor growth. Honokiol (HNK) is a natural compound, which possesses both anti-inflammatory and antitumorigenic properties. In this study, we investigated the effect of a novel combination therapy using RAPA + HNK on allograft survival and post-transplantation renal tumor growth. We observed that it effectively modulated the expression of some key regulatory molecules (like Carabin, an endogenous Ras inhibitor; and Rubicon, a negative regulator of autophagy) that play important roles in tumor cell growth and survival. This combination induced toxic autophagy and apoptosis to promote cancer cell death; and was associated with a reduced expression of the tumor-promoting receptor tyrosine kinase AXL. Finally, we utilized a novel murine model to examine the effect of RAPA + HNK on post-transplantation renal tumor growth. The combination treatment prolonged the allograft survival and significantly inhibited post-transplantation tumor growth. It was associated with reduced tumor expression of Rubicon and the cytoprotective/antioxidant heme oxygenase-1 to overcome therapeutic resistance. It also downregulated the coinhibitory programmed death-1 ligand, which plays major role(s) in the immune escape of tumor cells. Together, this combination treatment has a great potential to restrict renal tumor growth in transplant recipients as well as other immunosuppressed patients.


Asunto(s)
Neoplasias Renales , Trasplante de Órganos , Animales , Apoptosis , Autofagia , Compuestos de Bifenilo , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Renales/patología , Lignanos , Ratones , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR
2.
Am J Transplant ; 22(3): 947-954, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34687147

RESUMEN

The statin family of therapeutics is widely used clinically as cholesterol lowering agents, and their effects to target intracellular mevalonate production is a key mechanism of action. In this study, we performed full transcriptomic RNA sequencing and qPCR to evaluate the effects of mevalonate on the immunoregulatory phenotype of endothelial cells (EC). We find that mevalonate-dependent gene regulation includes a reduction in the expression of multiple pro-inflammatory genes including TNFSF4 (OX40-L) and TNFSF18 (GITR-L) and a co-incident induction of immunoregulatory genes including LGALS3 (Galectin-3) and LGALS9 (Galectin-9). In functional assays, pretreatment of EC with simvastatin to inhibit mevalonate metabolism resulted in a dose-dependent reduction in the costimulation of CD45RO+ CD4+ T cell proliferation as well as IL-2, IFNγ and IL-6 production versus vehicle-treated EC. In contrast, pre-treatment of EC with L-mevalonate in combination with simvastatin reversed phenotypic and functional responses. Collectively, these results indicate that relative mevalonate metabolism by EC is critical to sustain EC-dependent mechanisms of immunity. Our findings have broad relevance for the repurposing of statins as therapeutics to augment immunoregulation and/or to inhibit local tissue pro-inflammatory cytokine production following transplantation.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Endoteliales , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/metabolismo , Ácido Mevalónico/farmacología , Fenotipo , Simvastatina/farmacología , Linfocitos T/metabolismo
3.
J Immunol ; 203(8): 2328-2338, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31541025

RESUMEN

The T cell-specific adaptor protein (TSAd), encoded by the SH2D2A gene, is an intracellular molecule that binds Lck to elicit signals that result in cytokine production in CD4+ T effector cells (Teff). Nevertheless, using Sh2d2a knockout (KO; also called TSAd-/-) mice, we find that alloimmune CD4+ Teff responses are fully competent in vivo. Furthermore, and contrary to expectations, we find that allograft rejection is accelerated in KO recipients of MHC class II-mismatched B6.C-H-2bm12 heart transplants versus wild-type (WT) recipients. Also, KO recipients of fully MHC-mismatched cardiac allografts are resistant to the graft-prolonging effects of costimulatory blockade. Using adoptive transfer models, we find that KO T regulatory cells (Tregs) are less efficient in suppressing Teff function and they produce IFN-γ following mitogenic activation. In addition, pyrosequencing demonstrated higher levels of methylation of CpG regions within the Treg-specific demethylated region of KO versus WT Tregs, suggesting that TSAd, in part, promotes Treg stability. By Western blot, Lck is absent in the mitochondria of KO Tregs, and reactive oxygen species production by mitochondria is reduced in KO versus WT Tregs. Full transcriptomic analysis demonstrated that the key mechanism of TSAd function in Tregs relates to its effects on cellular activation rather than intrinsic effects on mitochondria/metabolism. Nevertheless, KO Tregs compensate for a lack of activation by increasing the number of mitochondria per cell. Thus, TSAd serves as a critical cell-intrinsic molecule in CD4+Foxp3+ Tregs to regulate the translocation of Lck to mitochondria, cellular activation responses, and the development of immunoregulation following solid organ transplantation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos T CD4-Positivos/inmunología , Mitocondrias/metabolismo , Trasplante , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/citología , Proliferación Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Am Soc Nephrol ; 30(3): 393-405, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30737270

RESUMEN

BACKGROUND: Although studies have identified >55 genes as causing steroid-resistant nephrotic syndrome (SRNS) and localized its pathogenesis to glomerular podocytes, the disease mechanisms of SRNS remain largely enigmatic. We recently reported that individuals with mutations in COQ6, a coenzyme Q (also called CoQ10, CoQ, or ubiquinone) biosynthesis pathway enzyme, develop SRNS with sensorineural deafness, and demonstrated the beneficial effect of CoQ for maintenace of kidney function. METHODS: To study COQ6 function in podocytes, we generated a podocyte-specific Coq6 knockout mouse (Coq6podKO ) model and a transient siRNA-based COQ6 knockdown in a human podocyte cell line. Mice were monitored for development of proteinuria and assessed for development of glomerular sclerosis. Using a podocyte migration assay, we compared motility in COQ6 knockdown podocytes and control podocytes. We also randomly assigned 5-month-old Coq6podKO mice and controls to receive no treatment or 2,4-dihydroxybenzoic acid (2,4-diHB), an analog of a CoQ precursor molecule that is classified as a food additive by health authorities in Europe and the United States. RESULTS: Abrogation of Coq6 in mouse podocytes caused FSGS and proteinuria (>46-fold increases in albuminuria). In vitro studies revealed an impaired podocyte migration rate in COQ6 knockdown human podocytes. Treating Coq6podKO mice or cells with 2,4-diHB prevented renal dysfunction and reversed podocyte migration rate impairment. Survival of Coq6podKO mice given 2,4diHB was comparable to that of control mice and significantly higher than that of untreated Coq6podKO mice, half of which died by 10 months of age. CONCLUSIONS: These findings reveal a potential novel treatment strategy for those cases of human nephrotic syndrome that are caused by a primary dysfunction in the CoQ10 biosynthesis pathway.

5.
Am J Transplant ; 19(1): 77-88, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29969188

RESUMEN

DEPTOR is an evolutionarily conserved cell-intrinsic binding partner of mTOR that functions as a negative regulator of signaling responses. In this study, we show that DEPTOR is expressed within CD4+ T cells, and we observed that its relative level of expression modulates differentiation as well as glucose utilization within CD4+ T effectors in vitro. Using knock-in mice, we also find that induced expression of DEPTOR within CD4+ T regulatory cells stabilizes Foxp3 expression, shifts metabolism toward oxidative phosphorylation, and increases survival and suppressive function. In vivo, fully MHC mismatched cardiac allograft survival is significantly prolonged in knock-in recipients and sustained recipient expression of DEPTOR in combination with costimulatory blockade induces long-term graft survival. Furthermore, we show that the induced expression of DEPTOR in CD4+ T effectors fails to inhibit acute allograft rejection. Rather, prolonged survival is dominantly mediated via induced expression and function of DEPTOR within recipient CD4+ T regulatory cells. These collective findings identify DEPTOR as a novel protein that functions in CD4+ T cells to augment immunoregulation in vitro and in vivo.


Asunto(s)
Trasplante de Corazón , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trasplante de Piel , Linfocitos T Reguladores/citología , Animales , Factores de Transcripción Forkhead/metabolismo , Glucosa/metabolismo , Rechazo de Injerto , Supervivencia de Injerto/efectos de los fármacos , Masculino , Metilación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/química , Fosforilación , Trasplante Homólogo
6.
J Immunol ; 197(4): 1389-98, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27430720

RESUMEN

Netrin-1 is a neuronal guidance cue that regulates cellular activation, migration, and cytoskeleton rearrangement in multiple cell types. It is a chemotropic protein that is expressed within tissues and elicits both attractive and repulsive migratory responses. Netrin-1 has recently been found to modulate the immune response via the inhibition of neutrophil and macrophage migration. However, the ability of Netrin-1 to interact with lymphocytes and its in-depth effects on leukocyte migration are poorly understood. In this study, we profiled the mRNA and protein expression of known Netrin-1 receptors on human CD4(+) T cells. Neogenin, uncoordinated-5 (UNC5)A, and UNC5B were expressed at low levels in unstimulated cells, but they increased following mitogen-dependent activation. By immunofluorescence, we observed a cytoplasmic staining pattern of neogenin and UNC5A/B that also increased following activation. Using a novel microfluidic assay, we found that Netrin-1 stimulated bidirectional migration and enhanced the size of migratory subpopulations of mitogen-activated CD4(+) T cells, but it had no demonstrable effects on the migration of purified CD4(+)CD25(+)CD127(dim) T regulatory cells. Furthermore, using a short hairpin RNA knockdown approach, we observed that the promigratory effects of Netrin-1 on T effectors is dependent on its interactions with neogenin. In the humanized SCID mouse, local injection of Netrin-1 into skin enhanced inflammation and the number of neogenin-expressing CD3(+) T cell infiltrates. Neogenin was also observed on CD3(+) T cell infiltrates within human cardiac allograft biopsies with evidence of rejection. Collectively, our findings demonstrate that Netrin-1/neogenin interactions augment CD4(+) T cell chemokinesis and promote cellular infiltration in association with acute inflammation in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiotaxis de Leucocito/fisiología , Factores de Crecimiento Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , Células Cultivadas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Ratones , Ratones SCID , Técnicas Analíticas Microfluídicas , Receptores de Netrina , Netrina-1 , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Curr Opin Organ Transplant ; 22(1): 55-63, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27898465

RESUMEN

PURPOSE OF REVIEW: Chronic rejection is associated with persistent mononuclear cell recruitment, endothelial activation and proliferation, local tissue hypoxia and related biology that enhance effector immune responses. In contrast, the tumor microenvironment elicits signals/factors that inhibit effector T cell responses and rather promote immunoregulation locally within the tissue itself. The identification of immunoregulatory check points and/or secreted factors that are deficient within allografts is of great importance in the understanding and prevention of chronic rejection. RECENT FINDINGS: The relative deficiency of immunomodulatory molecules (cell surface and secreted) on microvascular endothelial cells within the intragraft microenvironment, is of functional importance in shaping the phenotype of rejection. These regulatory molecules include coinhibitory and/or intracellular regulatory signals/factors that enhance local activation of T regulatory cells. For example, semaphorins may interact with endothelial cells and CD4 T cells to promote local tolerance. Additionally, metabolites and electrolytes within the allograft microenvironment may regulate local effector and regulatory cell responses. SUMMARY: Multiple factors within allografts shape the microenvironment either towards local immunoregulation or proinflammation. Promoting the expression of intragraft cell surface or secreted molecules that support immunoregulation will be critical for long-term graft survival and/or alloimmune tolerance.


Asunto(s)
Rechazo de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Humanos , Microambiente Tumoral
8.
Pediatr Nephrol ; 31(1): 41-51, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25903640

RESUMEN

Advances in therapeutics have dramatically improved short-term graft survival, but the incidence of chronic rejection has not changed in the past 20 years. New insights into mechanism are sorely needed at this time and it is hoped that the development of predictive biomarkers will pave the way for the emergence of preventative therapeutics. In this review, we discuss a paradigm suggesting that sequential changes within graft endothelial cells (EC) lead to an intragraft microenvironment that favors the development of chronic rejection. Key initial events include EC injury, activation and uncontrolled leukocyte-induced angiogenesis. We propose that all of these early changes in the microvasculature lead to abnormal blood flow patterns, local tissue hypoxia, and an associated overexpression of HIF-1α-inducible genes, including vascular endothelial growth factor. We also discuss how cell intrinsic regulators of mTOR-mediated signaling within EC are of critical importance in microvascular stability and may thus have a role in the inhibition of chronic rejection. Finally, we discuss recent findings indicating that miRNAs may regulate EC stability, and we review their potential as novel non-invasive biomarkers of allograft rejection. Overall, this review provides insights into molecular events, genes, and signals that promote chronic rejection and their potential as biomarkers that serve to support the future development of interruption therapeutics.


Asunto(s)
Células Endoteliales/metabolismo , Rechazo de Injerto/etiología , Trasplante de Riñón/efectos adversos , Riñón/irrigación sanguínea , Microvasos/metabolismo , Investigación Biomédica Traslacional , Aloinjertos , Animales , Biomarcadores/metabolismo , Microambiente Celular , Enfermedad Crónica , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Microvasos/inmunología , Microvasos/fisiopatología , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
9.
Curr Opin Organ Transplant ; 20(1): 13-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25563987

RESUMEN

PURPOSE OF REVIEW: New developments suggest that the graft itself and molecules expressed within the graft microenvironment dictate the phenotype and evolution of chronic rejection. RECENT FINDINGS: Once ischemia-reperfusion injury, cellular and humoral immune responses target the microvasculature, the associated local tissue hypoxia results in hypoxia-inducible factor 1α-dependent expression of pro-inflammatory and proangiogenic growth factors including vascular endothelial growth factor (VEGF) as a physiological response to injury. Local expression of VEGF can promote the recruitment of alloimune T cells into the graft. mTOR/Akt signaling within endothelial cells regulates cytokine- and alloantibody-induced activation and proliferation and their proinflammatory phenotype. Inhibition of mTOR and/or Akt results in an anti-inflammatory phenotype and enables the expression of coinhibitory molecules that limit local T cell reactivation and promotes immunoregulation. Semaphorin family molecules may bind to neuropilin-1 on regulatory T cell subsets to stabilize functional responses. Ligation of neuropilin-1 on Tregs also inhibits Akt-induced responses suggesting common theme for enhancing local immunoregulation and long-term graft survival. SUMMARY: Events within the graft initiated by mTOR/Akt-induced signaling promote the development of chronic rejection. Semaphorin-neuropilin biology represents a novel avenue for targeting this biology and warrants further investigation.


Asunto(s)
Rechazo de Injerto , Trasplante de Órganos , Transducción de Señal , Aloinjertos , Animales , Enfermedad Crónica , Humanos
10.
Platelets ; 24(6): 428-34, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22916829

RESUMEN

Dopamine (DA) is a co-agonist for platelet activation; yet, donor DA treatment is associated with improved transplantation outcome in renal and heart recipients. Recently, N-octanoyl-dopamine (NOD) was developed which displays superior effects compared to DA in terms of graft protecting properties. Whereas DA is a known platelet co-agonist, the effect of NOD on platelet function is unknown. This is a hypothesis generating study with the aim to assess the effects and molecular mechanisms of NOD and NOD-like compounds on platelet function. The influence of DA, NOD, and NOD-like compounds on platelet responses to classical agonists (adenosine 5'-diphosphate (ADP), U46619) was investigated in six healthy donors by applying whole blood aggregometry (Multiplate®) and flow cytometry for Pac-1, CD62P, and CD63 expression. Changes in platelet cAMP concentrations were assessed by ELISA. While DA showed synergy in platelet activation by ADP and U46619, NOD caused significant inhibition of platelet function both in whole blood aggregometry and flow cytometry. The inhibitory effect of NOD was not mediated via cAMP levels. The nonredox-active NOD-analog N-octanoyl-tyramine had no effects on platelet function. Acetylated NOD conferred to NOD by intracellular esterases showed similar inhibitory effects as NOD. In contrast to DA, NOD is a potent inhibitor of platelet function most likely through intracellular redox-active processes. This adds to the overall protective effect of NOD on pre-transplantation injury and makes NOD an attractive candidate compound for donor or organ conditioning prior to transplantation.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Dopamina/análogos & derivados , Degranulación de la Célula/efectos de los fármacos , AMP Cíclico/biosíntesis , Dopamina/farmacología , Humanos , Agregación Plaquetaria/efectos de los fármacos , Receptores Fibrinógenos/agonistas , Receptores Fibrinógenos/metabolismo
11.
Sci Rep ; 7: 44403, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28303937

RESUMEN

Excessive lymphangiogenesis is associated with cancer progression and renal disease. Attenuation of lymphangiogenesis might represent a novel strategy to target disease progression although clinically approved anti-lymphangiogenic drugs are not available yet. VitaminD(VitD)-deficiency is associated with increased cancer risk and chronic kidney disease. Presently, effects of VitD on lymphangiogenesis are unknown. Given the apparently protective effects of VitD and the deleterious associations of lymphangiogenesis with renal disease, we here tested the hypothesis that VitD has direct anti-lymphangiogenic effects in vitro and is able to attenuate lymphangiogenesis in vivo. In vitro cultured mouse lymphatic endothelial cells (LECs) expressed VitD Receptor (VDR), both on mRNA and protein levels. Active VitD (calcitriol) blocked LEC tube formation, reduced LEC proliferation, and induced LEC apoptosis. siRNA-mediated VDR knock-down reversed the inhibitory effect of calcitriol on LEC tube formation, demonstrating how such inhibition is VDR-dependent. In vivo, proteinuric rats were treated with vehicle or paricalcitol for 6 consecutive weeks. Compared with vehicle-treated proteinuric rats, paricalcitol showed markedly reduced renal lymphangiogenesis. In conclusion, our data show that VitD is anti-lymphangiogenic through VDR-dependent anti-proliferative and pro-apoptotic mechanisms. Our findings highlight an important novel function of VitD demonstrating how it may have therapeutic value in diseases accompanied by pathological lymphangiogenesis.


Asunto(s)
Ergocalciferoles/farmacología , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Proteinuria/tratamiento farmacológico , Receptores de Calcitriol/genética , Vitamina D/farmacología , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Doxorrubicina/toxicidad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Expresión Génica , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ratones , Proteinuria/inducido químicamente , Proteinuria/metabolismo , Proteinuria/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/metabolismo
12.
Transplantation ; 100(1): 80-90, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26674731

RESUMEN

BACKGROUND: Transplant vasculopathy (TV) is a major cause for late graft loss after cardiac transplantation. Endothelial damage and T cell infiltration play a pivotal role in the development of TV. Because N-octanoyl dopamine (NOD) inhibits vascular inflammation and suppresses T cell activation in vitro, we here tested the hypothesis that NOD treatment ameliorates TV. METHODS: Aortic grafts were orthotopically transplanted in the Dark Agouti to Brown Norway strain combination. Recipient rats were treated with NOD or vehicle administered via osmotic minipumps. Histology and quantitative polymerase chain reaction (qPCR) were performed on nontransplanted aortas and grafts explanted 2 and 4 weeks after transplantation to assess the degree of TV, inflammation, apoptosis, and number of (proliferating) α smooth muscle actin (αSMA) neointimal cells. In vitro analyses of human aortic smooth muscle cells were performed to test the effect of NOD on proliferation (WST-1 assay), cell cycle (flow cytometry and qPCR), and cytokine-induced apoptosis (flow cytometry). RESULTS: Allografts from vehicle-treated recipients developed neointimal lesions predominantly consisting of αSMA-expressing cells. NOD treatment significantly reduced neointima formation and neointimal αSMA cells. In situ, smooth muscle cell proliferation (Ki67) was not influenced by NOD. Macrophage (CD68), T (CD3), and Natural Killer (ANK61) cell infiltration as well as intragraft TNFα and IFNγ mRNA expression were similar in both groups. Medial apoptosis (cleaved caspase-3) was significantly reduced by NOD. In vitro, NOD inhibited proliferation of human aortic smooth muscle cells by causing a G1-arrest and protected from TNFα-induced apoptosis. CONCLUSIONS: This study identified NOD as potential treatment modality to attenuate TV. Our data clearly support a vasculoprotective effect of NOD by reducing smooth muscle cell proliferation and inflammation-induced apoptosis.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades de la Aorta/prevención & control , Dopamina/análogos & derivados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/trasplante , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/trasplante , Actinas/metabolismo , Aloinjertos , Animales , Antiinflamatorios/administración & dosificación , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aorta/trasplante , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Citoprotección , Dopamina/administración & dosificación , Dopamina/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Bombas de Infusión Implantables , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima , Ratas Endogámicas BN , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo
13.
Transplantation ; 100(8): 1656-66, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27014792

RESUMEN

BACKGROUND: Development of transplant vasculopathy is a major cause of graft loss and mortality in solid organ transplant recipients. Previous studies in mice have indicated that vanin-1, a member of the vanin protein family with pantetheinase activity, is possibly involved in neointima formation. Here, we investigated if RR6, a recently developed vanin inhibitor, could attenuate development of transplant vasculopathy. METHODS: Abdominal allogeneic aorta transplantation from Dark Agouti to Brown Norway rats was performed. Surface neointima was quantified 2 and 4 weeks after transplantation. Systemic vanin activity was measured, and allograft leukocyte infiltration, glutathione-synthesizing capacity, matrix metalloproteinase 9 expression and neointimal smooth muscle cell (SMC) proliferation were assessed by immunohistochemistry. In vitro, the effects of RR6 on SMC proliferation (water-soluble tetrazolium-1 assay) and cytokine-induced apoptosis (flow cytometry) were investigated. RESULTS: RR6 treatment significantly reduced systemic pantetheinase activity during the 4-week follow-up period. RR6 attenuated neointima formation 4 weeks after transplantation. Neointimal SMC proliferation and medial SMC matrix metalloproteinase 9 expression were not altered by RR6. However, RR6 significantly reduced neointimal macrophage influx that was accompanied by increased GCLC messenger RNA expression. In vitro, RR6 inhibited platelet-derived growth factor-induced SMC proliferation and protected SMCs from TNF-α-induced apoptosis. CONCLUSIONS: Pharmacological inhibition of vanin activity attenuates development of transplant vasculopathy. This was accompanied by reduced macrophage infiltration and increased glutathione-synthesizing capacity. In vitro, RR6 reduced SMC proliferation and apoptosis that was not confirmed in vivo. Further in-depth studies are warranted to reveal the underlying mechanism(s) of RR6-induced attenuation of transplant vasculopathy in vivo.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/trasplante , Enfermedades de la Aorta/prevención & control , Inhibidores Enzimáticos/farmacología , Supervivencia de Injerto/efectos de los fármacos , Amidohidrolasas/metabolismo , Animales , Aorta Abdominal/enzimología , Aorta Abdominal/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/patología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Glutatión/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Músculo Liso Vascular/trasplante , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Neointima , Ratas Endogámicas BN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Trasplante Homólogo
14.
Transplant Rev (Orlando) ; 29(3): 109-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25576467

RESUMEN

Conjugates of fatty acids with ethanolamine, amino acids or monoamine neurotransmitters occur widely in nature giving rise to so-called endocannabinoids. Anandamide and 2-arachidonoyl glycerol are the best characterized endocannabinoids activating both cannabinoid receptors (CB1 and CB2) and transient receptor potential vanilloid type 1 (TRPV1) channels (anandamide) or activating cannabinoid receptors only (2-arachidonoyl glycerol). TRPV1 is also activated by vanilloids, such as capsaicin, and endogenous neurolipins, e.g. N-arachidonoyl dopamine (NADA) and N-oleoyl dopamine (OLDA). Because donor dopamine treatment has shown to improve transplantation outcome in renal and heart recipients, this review will mainly focus on the biological activities of N-acyl dopamine derivates (NADD) as potential non-hemodynamic alternative for implementation in transplantation medicine. Hence the influence of NADD on transplantation relevant entities, i.e. cold inflicted injury, cytoprotection, I/R-injury, immune-modulation and inflammation will be summarized. The cytoprotective properties of endogenous endocannabinoids in this context will be briefly touched upon.


Asunto(s)
Capsaicina/metabolismo , Dopamina/metabolismo , Endocannabinoides/metabolismo , Trasplante de Órganos , Animales , Rechazo de Injerto , Supervivencia de Injerto , Trasplante de Corazón/efectos adversos , Trasplante de Corazón/métodos , Humanos , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Rol , Sensibilidad y Especificidad , Inmunología del Trasplante
15.
Lab Anim ; 48(4): 338-41, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25002205

RESUMEN

Subcutaneous osmotic pump implantation connected to a venous catheter is a well-established method for delivering compounds intravenously for an intermediate duration (approximately two weeks). When prolonged release is desired (approximately four weeks) reduced flow rate is needed with a similar pump volume. With a fixed intra-pump compound concentration, reduced flow rate results in unwanted reduced bioavailability of the compound. Prolonged intravenous delivery would therefore need a pump replacement, resulting in increased discomfort and confounding effects on experimental outcome. To overcome this, we describe a method to double the compound infusion rate for four weeks by implanting two low-flow rate osmotic pumps (2.5 µL/h for 28 days) connected to a jugular vein catheter in a single rat. Rats implanted with a single high-flow rate pump (5 µL/h for 14 days) served as controls. Double pump-implanted rats displayed similar post-operative weight gain and physical activity indicating similar levels of discomfort when compared with single pump-implanted rats. Double pump-implanted rats had an increased risk of pump-related complications (four delivery failures [double pump] versus one delivery failure [single pump]). Our data show that double pump implantation is a feasible alternative to changing pumps or the use of extracorporeal pump systems connected via a long wire to partly restrained animals.


Asunto(s)
Cateterismo Venoso Central/instrumentación , Sistemas de Liberación de Medicamentos/instrumentación , Bombas de Infusión Implantables/normas , Venas Yugulares , Animales , Cateterismo Venoso Central/efectos adversos , Sistemas de Liberación de Medicamentos/efectos adversos , Femenino , Bombas de Infusión Implantables/efectos adversos , Masculino , Ratas , Ratas Wistar
16.
J Leukoc Biol ; 96(3): 453-62, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24929005

RESUMEN

Recently, we developed a nonhemodynamic dopamine derivative, NOD, which has profound anti-inflammatory effects in vitro. As NOD also protects rats from ischemic AKI, the present study tested whether NOD is able to modulate cellular immunity for potential use as a T cell-suppressive agent. To this end, T cells were stimulated by anti-CD3/CD28 or PMA/ionomycin in the presence or absence of different concentrations of NOD. T cell proliferation, activation markers, intracellular cytokine expression, and activation of transcription factors were assessed. Whereas T cell proliferation was inhibited significantly by NOD at Day 3, proliferation was restored at Day 7 or later depending on the NOD concentration used. Inhibition of proliferation was reflected by a diminished CD25 expression and switch from naive to memory T cells. Early TCR activation events were unaffected, yet NF-κB and AP-1 were strongly inhibited by NOD. The inhibitory effect of NOD seemed to be dependent on its redox activity, as NOT, a redox-inactive NOD derivate, did not influence proliferation. NOD displayed synergistic effects with CNIs on T cell proliferation. Our data demonstrate that NOD displays T cell-suppressive activity. In keeping with its anti-inflammatory action and its beneficial effect on ischemia-induced AKI, NOD may be an interesting drug candidate to prevent CNI-related side-effects.


Asunto(s)
Dopamina/análogos & derivados , Fase G1/efectos de los fármacos , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Inhibidores de la Calcineurina/farmacología , División Celular/efectos de los fármacos , Dopamina/farmacología , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Humanos , Memoria Inmunológica , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/efectos de los fármacos , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Linfocitos T/citología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Proteína Tirosina Quinasa ZAP-70/metabolismo
17.
PLoS One ; 9(6): e99298, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24926788

RESUMEN

AIM: N-acyl dopamines (NADD) are gaining attention in the field of inflammatory and neurological disorders. Due to their hydrophobicity, NADD may have access to the endoplasmic reticulum (ER). We therefore investigated if NADD induce the unfolded protein response (UPR) and if this in turn influences cell behaviour. METHODS: Genome wide gene expression profiling, confirmatory qPCR and reporter assays were employed on human umbilical vein endothelial cells (HUVEC) to validate induction of UPR target genes and UPR sensor activation by N-octanoyl dopamine (NOD). Intracellular ATP, apoptosis and induction of thermotolerance were used as functional parameters to assess adaptation of HUVEC. RESULTS: NOD, but not dopamine dose dependently induces the UPR. This was also found for other synthetic NADD. Induction of the UPR was dependent on the redox activity of NADD and was not caused by selective activation of a particular UPR sensor. UPR induction did not result in cell apoptosis, yet NOD strongly impaired cell proliferation by attenuation of cells in the S-G2/M phase. Long-term treatment of HUVEC with low NOD concentration showed decreased intracellular ATP concentration paralleled with activation of AMPK. These cells were significantly more resistant to cold inflicted injury. CONCLUSIONS: We provide for the first time evidence that NADD induce the UPR in vitro. It remains to be assessed if UPR induction is causally associated with hypometabolism and thermotolerance. Further pharmacokinetic studies are warranted to address if the NADD concentrations used in vitro can be obtained in vivo and if this in turn shows therapeutic efficacy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Dopamina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dopamina/farmacología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro
18.
PLoS One ; 8(9): e73122, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023820

RESUMEN

BACKGROUND AND PURPOSE: Catechol containing compounds have anti-inflammatory properties, yet for catecholamines these properties are modest. Since we have previously demonstrated that the synthetic dopamine derivative N-octanoyl dopamine (NOD) has superior anti-inflammatory properties compared to dopamine, we tested NOD in more detail and sought to elucidate the molecular entities and underlying mechanism by which NOD down-regulates inflammation. EXPERIMENTAL APPROACH: Genome wide gene expression profiling of human umbilical vein endothelial cells (HUVECs) was performed after stimulation with TNF-α or in the combination with NOD. Confirmation of these differences, NFκB activation and the molecular entities that were required for the anti-inflammatory properties were assessed in subsequent experiments. KEY RESULTS: Down regulation of inflammatory genes by NOD occurred predominantly for κB regulated genes, however not all κB regulated genes were affected. These findings were explained by inhibition of RelA phosphorylation at Ser276. Leukocyte adherence to TNF-α stimulated HUVECs was inhibited by NOD and was reflected by a diminished expression of adhesion molecules on HUVECs. NOD induced HO-1 expression, but this was not required for inhibition of NFκB. The anti-inflammatory effect of NOD seems to involve the redox active catechol structure, although the redox active para-dihydroxy benzene containing compounds also displayed anti-inflammatory effects, provided that they were sufficiently hydrophobic. CONCLUSIONS AND IMPLICATIONS: The present study highlighted important mechanisms and molecular entities by which dihydroxy benzene compounds exert their potential anti-inflammatory action. Since NOD does not have hemodynamic properties, NOD seems to be a promising candidate drug for the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Dopamina/análogos & derivados , Regulación de la Expresión Génica/efectos de los fármacos , Serina/metabolismo , Factor de Transcripción ReIA/química , Factor de Transcripción ReIA/metabolismo , Antiinflamatorios no Esteroideos/química , Adhesión Celular/efectos de los fármacos , Dopamina/química , Dopamina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
PLoS One ; 7(8): e43525, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916273

RESUMEN

Since stimulation of transient receptor potential channels of the vanilloid receptor subtype 1 (TRPV1) mitigates acute kidney injury (AKI) and endogenous N-acyl dopamine derivatives are able to activate TRPV1, we tested if synthetic N-octanoyl-dopamine (NOD) activates TRPV1 and if it improves AKI. These properties of NOD and its intrinsic anti-inflammatory character were compared with those of dopamine (DA). TRPV1 activation and anti-inflammatory properties of NOD and DA were tested using primary cell cultures in vitro. The influence of NOD and DA on AKI was tested in a prospective, randomized, controlled animal study with 42 inbred male Lewis rats (LEW, RT1), treated intravenously with equimolar concentrations of DA or NOD one hour before the onset of warm ischemia and immediately before clamp release. NOD, but not DA, activates TRPV1 channels in isolated dorsal root ganglion neurons (DRG) that innervate several tissues including kidney. In TNFα stimulated proximal tubular epithelial cells, inhibition of NFκB and subsequent inhibition of VCAM1 expression by NOD was significantly stronger than by DA. NOD improved renal function compared to DA and saline controls. Histology revealed protective effects of NOD on tubular epithelium at day 5 and a reduced number of monocytes in renal tissue of DA and NOD treated rats. Our data demonstrate that NOD but not DA activates TRPV1 and that NOD has superior anti-inflammatory properties in vitro. Although NOD mitigates deterioration in renal function after AKI, further studies are required to assess to what extend this is causally related to TRPV1 activation and/or desensitization.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Dopamina/análogos & derivados , Dopamina/uso terapéutico , Canales Catiónicos TRPV/agonistas , Animales , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Inmunohistoquímica , Masculino , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Molécula 1 de Adhesión Celular Vascular/metabolismo
20.
Clin Chim Acta ; 412(3-4): 263-7, 2011 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-20971102

RESUMEN

BACKGROUND: We reported an association of a particular allele of the carnosinase (CNDP1 Mannheim) gene with reduced serum carnosinase (CN1) activity and absence of nephropathy in diabetic patients. Carnosine protects against the adverse effects of high glucose levels but serum carnosine concentration was generally low. METHODS: We measured the concentration of two further histidine dipeptides, anserine and homocarnosine, via HPLC. CN1 activity was measured fluorometically and for concentration we developed a capture ELISA. RESULTS: We found an association between the CNDP1 Mannheim allele and reduced serum CN1 activity for all three dipeptides but no correlation to serum concentrations although anserine and homocarnosine inhibited carnosinase activity. Patients with liver cirrhosis have low CN1 activity (0.24 ± 0.17 µmol/ml/h, n=7 males; normal range: 3.2 ± 1.1, n=104; p<0.05) and CN1 concentrations (2.3 ± 1.5 µg/ml; normal range: 24.9 ± 8.9, p<0.05) but surprisingly, histidine dipeptide concentrations in serum are not increased compared to controls. CONCLUSIONS: Serum histidine dipeptide concentrations are not correlated to CN1 activity. The protective effect of low CN1 activity might be related either to turnover of CN1 substrates or a protective function of dipeptides might be localized in other tissues.


Asunto(s)
Anserina/metabolismo , Carnosina/metabolismo , Dipeptidasas/metabolismo , Histidina/metabolismo , Adulto , Anserina/sangre , Carnosina/sangre , Femenino , Histidina/sangre , Humanos , Cirrosis Hepática/sangre , Cirrosis Hepática/enzimología , Cirrosis Hepática/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA