Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS One ; 13(7): e0200697, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30024944

RESUMEN

The purpose of this study was to determine whether blocking of G protein ßγ (Gßγ) signaling halts heart failure (HF) progression by macrophage phenotype manipulation. Cardiac Gßγ signaling plays a crucial role in HF pathogenesis. Previous data suggested that inhibiting Gßγ signaling reprograms T helper cell 1 (Th1) and Th2 cytokines, suggesting that Gßγ might be a useful drug target for treating HF. We investigated the efficacy of a small molecule Gßγ inhibitor, gallein, in a clinically relevant, experimental autoimmune myocarditis (EAM) model of HF as well as in human macrophage phenotypes in vitro. In the myocardium of HF patients, we observed that G protein coupled receptor kinase (GRK)2 levels were down-regulated compared with healthy controls. In rat EAM, treatment with gallein effectively improved survival and cardiac function, suppressed cardiac remodeling, and further attenuated myocardial protein expression of GRK2 as well as high mobility group box (HMGB)1 and its cascade signaling proteins. Furthermore, gallein effectively inhibited M1 polarization and promoted M2 polarization in vivo in the EAM heart and in vitro in human monocyte-derived macrophages. Taken together, these data suggest that the small molecule Gßγ inhibitor, gallein, could be an important pharmacologic therapy for HF as it can switch the phenotypic reprogramming from M1 to M2 phenotype in a rat model of EAM heart and in human macrophages.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Macrófagos/efectos de los fármacos , Miocarditis/prevención & control , Transducción de Señal/efectos de los fármacos , Xantenos/farmacología , Animales , Enfermedades Autoinmunes/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteína HMGB1/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Humanos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/clasificación , Macrófagos/metabolismo , Masculino , Miocarditis/metabolismo , Ratas Endogámicas Lew
2.
Biomed Microdevices ; 20(2): 49, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29916059

RESUMEN

With nearly 40% of U.S. adults obese, and childhood and adolescent rates rising, obesity and associated comorbidities are serious public health concerns with massive societal costs. Often, lifestyle interventions do not offer sufficient weight loss to improve health, requiring surgery and medications as adjunct management strategies. Here, we present a 4-month case study in which the sustained, low-dose, and constant administration of the thyroid receptor ß selective agonist GC-1 (sobetirome) from a novel nanochannel membrane implant was assessed in an obese, pre-diabetic rhesus macaque. Dramatic loss of white adipose tissue in the abdomen from 36 to 18% was observed via magnetic resonance imaging in conjunction with normalized serum insulin and glycemia, with no signs of cardiotoxicity shown. The non-human primate study highlights sustained low-dose delivery of GC-1 from our minimally invasive subcutaneous implant as a valuable approach to induce weight loss and manage obesity and comorbidities, including type 2 diabetes.


Asunto(s)
Acetatos/metabolismo , Sistemas de Liberación de Medicamentos/instrumentación , Nanotecnología/instrumentación , Obesidad/metabolismo , Fenoles/metabolismo , Animales , Macaca mulatta
3.
mBio ; 9(3)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29764948

RESUMEN

Viral pneumonias cause profound worldwide morbidity, necessitating novel strategies to prevent and treat these potentially lethal infections. Stimulation of intrinsic lung defenses via inhalation of synergistically acting Toll-like receptor (TLR) agonists protects mice broadly against pneumonia, including otherwise-lethal viral infections, providing a potential opportunity to mitigate infectious threats. As intact lung epithelial TLR signaling is required for the inducible resistance and as these cells are the principal targets of many respiratory viruses, the capacity of lung epithelial cells to be therapeutically manipulated to function as autonomous antiviral effectors was investigated. Our work revealed that mouse and human lung epithelial cells could be stimulated to generate robust antiviral responses that both reduce viral burden and enhance survival of isolated cells and intact animals. The antiviral protection required concurrent induction of epithelial reactive oxygen species (ROS) from both mitochondrial and dual oxidase sources, although neither type I interferon enrichment nor type I interferon signaling was required for the inducible protection. Taken together, these findings establish the sufficiency of lung epithelial cells to generate therapeutically inducible antiviral responses, reveal novel antiviral roles for ROS, provide mechanistic insights into inducible resistance, and may provide an opportunity to protect patients from viral pneumonia during periods of peak vulnerability.IMPORTANCE Viruses are the most commonly identified causes of pneumonia and inflict unacceptable morbidity, despite currently available therapies. While lung epithelial cells are principal targets of respiratory viruses, they have also been recently shown to contribute importantly to therapeutically inducible antimicrobial responses. This work finds that lung cells can be stimulated to protect themselves against viral challenges, even in the absence of leukocytes, both reducing viral burden and improving survival. Further, it was found that the protection occurs via unexpected induction of reactive oxygen species (ROS) from spatially segregated sources without reliance on type I interferon signaling. Coordinated multisource ROS generation has not previously been described against viruses, nor has ROS generation been reported for epithelial cells against any pathogen. Thus, these findings extend the potential clinical applications for the strategy of inducible resistance to protect vulnerable people against viral infections and also provide new insights into the capacity of lung cells to protect against infections via novel ROS-dependent mechanisms.


Asunto(s)
Células Epiteliales/inmunología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Especies Reactivas de Oxígeno/inmunología , Animales , Células Epiteliales/virología , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/virología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
4.
Sci Rep ; 6: 36207, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27827458

RESUMEN

Efferocytosis, a process of clearance of apoptotic cells by phagocytes, is essential for successful resolution of inflammation and maintenance of tissue homeostasis. Diabetes compromises the function of macrophages leading to adverse inflammatory response during wound healing, myocardial injury, atherosclerosis and autoimmune disorders. However, the effect of diabetes on macrophage-mediated efferocytosis of apoptotic cardiomyocytes (ACM) and the molecular mechanisms involved are not understood so far. In the present study we found that invitro efferocytosis of ACM was impaired in macrophages from db/db (diabetic) mice. Macrophages exposed to high glucose (HG) decreases microRNA-126 (miR-126) expression with a corresponding increase in ADAM9 expression. Dual-luciferase reporter assay confirms that ADAM9 3'UTR contains miR-126 target site. ADAM9 inhibition reduces HG-induced proteolytic cleavage of Mer tyrosine receptor kinase (MerTK, a proto-oncogene that plays a critical role in phagocytosis), resulting in shedding of soluble-Mer (sMER) and loss of MERTK function. Over-expression of miR-126 attenuates HG-induced impairment of efferocytosis. Furthermore, human diabetic hearts show lower miR-126 expression with a corresponding increase in ADAM9 expression vs. normal counterparts. These data suggests that diabetes impairs efferocytosis of ACM and that strategies to enhance efferocytosis might attenuate diabetes-induced impairment in inflammation resolution and cardiac repair after injury.


Asunto(s)
Proteínas ADAM/genética , Diabetes Mellitus Experimental/genética , Macrófagos/citología , Proteínas de la Membrana/genética , MicroARNs/genética , Miocitos Cardíacos/citología , Regiones no Traducidas 3' , Animales , Apoptosis , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Humanos , Macrófagos/efectos de los fármacos , Ratones , Fagocitosis , Proto-Oncogenes Mas , Células RAW 264.7 , Células THP-1 , Tirosina Quinasa c-Mer/metabolismo
5.
Biochem Biophys Res Commun ; 471(4): 423-9, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26898797

RESUMEN

Diabetic cardiomyopathy is a common complication in patients with diabetes and is associated with underlying chronic inflammation and cardiac cell death, subsequently leading to heart failure (HF). ELAV-like protein 1 (ELAVL1) plays a critical role in the progression of inflammation and HF. However the role of ELAVL-1 in inflammation induced cardiac cell death (pyroptosis) under hyperglycemic condition remains elusive. Our data demonstrates that ELAVL1 expression augmented with a concomitant increase in caspase-1 and IL-1 beta expression in human hearts and human ventricular cardiomyocytes under hyperglycemic condition. Furthermore, ELAVL1 knockdown abrogates TNF-α induced canonical pyroptosis via NLRP3, caspase-1 and IL-1beta suppression. Bioinformatics analysis and target validation assays showed that miR-9 directly targets ELAVL1. Interestingly, miRNA-9 expression significantly reduced in high glucose treated cardiomyocytes and in human diabetic hearts. Inhibition of miR-9 upregulates ELAVL1 expression and activates caspase-1. Alternatively, treatment with miR-9 mimics attenuates hyperglycemia-induced ELAVL1 and inhibits cardiomyocyte pyroptosis. Taken together our study highlights the potential therapeutic implications of targeting miR-9/ELAVL1 in preventing cardiomyocyte cell loss during HF in diabetics.


Asunto(s)
Proteína 1 Similar a ELAV/genética , Hiperglucemia/genética , MicroARNs/genética , Miocitos Cardíacos/patología , Piroptosis/genética , Animales , Línea Celular , Células Cultivadas , Cardiomiopatías Diabéticas/patología , Proteína 1 Similar a ELAV/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ventrículos Cardíacos/patología , Humanos , Hiperglucemia/metabolismo , Ratones , MicroARNs/metabolismo , Miocitos Cardíacos/fisiología
6.
Hypertension ; 67(3): 597-605, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26781283

RESUMEN

The insufficiency of compensatory angiogenesis in the heart of patients with hypertension contributes to heart failure transition. The hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling cascade controls responsive angiogenesis. One of the challenges in reprograming the insufficient angiogenesis is to achieve a sustainable tissue exposure to the proangiogenic factors, such as HIF1α stabilization. In this study, we identified Rnd3, a small Rho GTPase, as a proangiogenic factor participating in the regulation of the HIF1α-VEGF signaling cascade. Rnd3 physically interacted with and stabilized HIF1α, and consequently promoted VEGFA expression and endothelial cell tube formation. To demonstrate this proangiogenic role of Rnd3 in vivo, we generated Rnd3 knockout mice. Rnd3 haploinsufficient (Rnd3(+/-)) mice were viable, yet developed dilated cardiomyopathy with heart failure after transverse aortic constriction stress. The poststress Rnd3(+/-) hearts showed significantly impaired angiogenesis and decreased HIF1α and VEGFA expression. The angiogenesis defect and heart failure phenotype were partially rescued by cobalt chloride treatment, a HIF1α stabilizer, confirming a critical role of Rnd3 in stress-responsive angiogenesis. Furthermore, we generated Rnd3 transgenic mice and demonstrated that Rnd3 overexpression in heart had a cardioprotective effect through reserved cardiac function and preserved responsive angiogenesis after pressure overload. Finally, we assessed the expression levels of Rnd3 in the human heart and detected significant downregulation of Rnd3 in patients with end-stage heart failure. We concluded that Rnd3 acted as a novel proangiogenic factor involved in cardiac responsive angiogenesis through HIF1α-VEGFA signaling promotion. Rnd3 downregulation observed in patients with heart failure may explain the insufficient compensatory angiogenesis involved in the transition to heart failure.


Asunto(s)
Vasos Coronarios/patología , Regulación de la Expresión Génica , Hipertensión/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Unión al GTP rho/genética , Animales , Western Blotting , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Neovascularización Patológica/metabolismo , ARN/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis
7.
J Am Coll Cardiol ; 66(20): 2214-2226, 2015 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-26564600

RESUMEN

BACKGROUND: MicroRNA (miR) dysregulation in the myocardium has been implicated in cardiac remodeling after injury or stress. OBJECTIVES: The aim of this study was to explore the role of miR in human CD34(+) cell (hCD34(+)) dysfunction in vivo after transplantation into the myocardium under ischemia-reperfusion (I-R) conditions. METHODS: In response to inflammatory stimuli, the miR array profile of endothelial progenitor cells was analyzed using a polymerase chain reaction-based miR microarray. miR-377 expression was assessed in myocardial tissue from human patients with heart failure (HF). We investigated the effect of miR-377 inhibition on an hCD34(+) cell angiogenic proteome profile in vitro and on cardiac repair and function after I-R injury in immunodeficient mice. RESULTS: The miR array data from endothelial progenitor cells in response to inflammatory stimuli indicated changes in numerous miR, with a robust decrease in the levels of miR-377. Human cardiac biopsies from patients with HF showed significant increases in miR-377 expression compared with nonfailing control hearts. The proteome profile of hCD34(+) cells transfected with miR-377 mimics showed significant decrease in the levels of proangiogenic proteins versus nonspecific control-transfected cells. We also validated that serine/threonine kinase 35 is a target of miR-377 using a dual luciferase reporter assay. In a mouse model of myocardial I-R, intramyocardial transplantation of miR-377 silenced hCD34(+) cells in immunodeficient mice, promoting neovascularization (at 28 days, post-I-R) and lower interstitial fibrosis, leading to improved left ventricular function. CONCLUSIONS: These findings indicate that HF increased miR-377 expression in the myocardium, which is detrimental to stem cell function, and transplantation of miR-377 knockdown hCD34(+) cells into ischemic myocardium promoted their angiogenic ability, attenuating left ventricular remodeling and cardiac fibrosis.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Insuficiencia Cardíaca/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Daño por Reperfusión/metabolismo , Adulto , Animales , Antígenos CD34 , Femenino , Corazón , Humanos , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Miocardio/patología , Neovascularización Fisiológica/fisiología , Daño por Reperfusión/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Circ Cardiovasc Genet ; 7(3): 266-76, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24825877

RESUMEN

BACKGROUND: Impaired bioenergetics is a prominent feature of the failing heart, but the underlying metabolic perturbations are poorly understood. METHODS AND RESULTS: We compared metabolomic, gene transcript, and protein data from 6 paired samples of failing human left ventricular tissue obtained during left ventricular assist device insertion (heart failure samples) and at heart transplant (post-left ventricular assist device samples). Nonfailing left ventricular wall samples procured from explanted hearts of patients with right heart failure served as novel comparison samples. Metabolomic analyses uncovered a distinct pattern in heart failure tissue: 2.6-fold increased pyruvate concentrations coupled with reduced Krebs cycle intermediates and short-chain acylcarnitines, suggesting a global reduction in substrate oxidation. These findings were associated with decreased transcript levels for enzymes that catalyze fatty acid oxidation and pyruvate metabolism and for key transcriptional regulators of mitochondrial metabolism and biogenesis, peroxisome proliferator-activated receptor γ coactivator 1α (PGC1A, 1.3-fold) and estrogen-related receptor α (ERRA, 1.2-fold) and γ (ERRG, 2.2-fold). Thus, parallel decreases in key transcription factors and their target metabolic enzyme genes can explain the decreases in associated metabolic intermediates. Mechanical support with left ventricular assist device improved all of these metabolic and transcriptional defects. CONCLUSIONS: These observations underscore an important pathophysiologic role for severely defective metabolism in heart failure, while the reversibility of these defects by left ventricular assist device suggests metabolic resilience of the human heart.


Asunto(s)
Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/metabolismo , Adulto , Anciano , Femenino , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética
9.
Eur Heart J ; 35(16): 1061-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24375073

RESUMEN

AIMS: Various reports have raised the possibility of humoral immune responses as contributors for the progression of heart failure. Previous studies, however, have focused on the analysis of serum and documented circulating antibodies against a variety of cardiac proteins. However, there is little evidence on whether anti-cardiac antibodies are deposited in end-stage failing myocardium. Our objective was to determine whether or not there was evidence of deposition of anti-cardiac antibodies and/or activated complement components in end-stage failing human myocardium. METHODS AND RESULTS: Myocardial samples were obtained from 100 end-stage heart failure patients and 40 donor control biopsies. Sections were cut and stained using standard fluorescent immunohistochemistry techniques with anti-human immunoglobulin G (IgG), IgG3, and C3c. Gel electrophoresis and protein identification by mass spectrometry were used to confirm the presence of IgG and its antigen. Immunoglobulin G was localized to the sarcolemma in 71% of patients, 48% of those being positive for the subtype IgG3. The proportion of patients with ischaemic heart disease that was positive for IgG was 65% and among those with non-ischaemic aetiologies was 76%. In a subgroup analysis, the presence of IgG and its subunits were confirmed by mass spectrometry and adenosine triphosphate synthase ß subunit identified as an antigen. Complement was activated in 31% of all patients. The presence of IgG, IgG3, and C3c was directly correlated with the length of disease (r = 0.451, P = 0.006). CONCLUSION: Evidence of anti-cardiac antibodies and complement activation was found in a large number of patients with end-stage cardiomyopathy regardless of the aetiology. Adenosine triphosphate synthase appears to be a new prominent antigenic stimulus; but more interestingly, the simultaneous co-existence of activated complement components suggests that this humoral mechanism may participate in disease progression.


Asunto(s)
Anticuerpos/metabolismo , Insuficiencia Cardíaca/inmunología , Miocardio/inmunología , Adenosina Trifosfatasas/inmunología , Antígenos/metabolismo , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Inmunidad Humoral/inmunología , Inmunoglobulina G/metabolismo , Masculino , Persona de Mediana Edad
10.
Congest Heart Fail ; 17(3): 140-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21609388

RESUMEN

Left ventricular assist devices (LVADs) induce reverse cardiac remodeling by reducing myocyte size and collagen deposition. On the other hand, cardiac resynchronization therapy (CRT) induces reverse cardiac remodeling by improving electromechanical synchronization. The clinical and structural changes produced by CRT in failing myocardium are known, but whether these changes are accompanied by reverse cellular remodeling is unknown. A total of 12 patients with chronic heart failure (CHF) who underwent CRT and 15 patients who had LVAD therapy as clinically indicated and 8 healthy controls were compared. Demographics, echocardiographic data, and histologic samples from myocardial biopsies were analyzed and compared among groups. The authors found significant increases in myocyte size, myocardial fibrosis, and inflammation in both CHF groups who underwent CRT or LVAD, compared with healthy controls. After CRT or LVAD therapy, a significant decrease in myocyte size and tumor necrosis factor α (TNF-α) expression compared with healthy controls (P < .05) was found. In the CRT group, 6 of 8 patients demonstrated reduction in myocyte size and interstitial fibrosis. In addition, there was a decrease in myocyte size by 13%, total collagen by 27% and TNF-α by 49% in the CRT group vs 28%, 45%, and 45% in the LVAD group. CRT produces cellular reverse remodeling in failing human hearts that are comparable with those produced by LVAD therapy.


Asunto(s)
Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Remodelación Ventricular/fisiología , Anciano , Biopsia , Enfermedad Crónica , Ecocardiografía , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Estudios Prospectivos , Factor de Necrosis Tumoral alfa/metabolismo
11.
Pathol Res Pract ; 205(9): 634-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19446406

RESUMEN

The mast cell has been associated with fibrosis in many different tissues, organs, and different disease processes including hematopoietic malignancies. Mast cells are often increased in the bone marrow of patients with primary bone marrow disorders, and patients with systemic mastocytosis often have a second concomitant neoplastic disease of the bone marrow. The goals of the current study were to determine the role the mast cell has in the pathogenesis of myeloproliferative neoplasms (MPN) and to correlate the mast cell burden with the degree of reticulin fibrosis. We used computer-assisted image analysis of bone marrow core biopsies stained for mast cell tryptase from patients with myeloproliferative neoplasms [31 cases: 12 chronic myelogenous leukemia (CML), 6 primary myelofibrosis (PMF), 4 essential thrombocythemia (ET), 4 polycythemia vera (PV), and 5 chronic myeloproliferative disorder, unclassifiable (CMPD-U)]. Although the number of cases of some subtypes of MPN was small, the results suggested that PMF and ET each had significantly more mast cells than both CML and control cases (P<0.01 and 0.05, respectively, Mann-Whitney test). CMPD-U and PV showed no significant differences from the control cases, but the CML cases had significantly fewer mast cells than our control cases (P=0.02, Mann-Whitney test). In addition, the quantity of mast cells seen in the bone marrows of MPN patients correlated with reticulin fibrosis (P=0.04, Mann-Whitney test). Our studies highlight the different mast cell quantities in different myeloproliferative neoplasms and suggest a direct role for the mast cell in intramedullary fibrosis. Further studies are warranted to confirm our observation and to study the mechanisms by which mast cells contribute to fibrosis in the MPN setting.


Asunto(s)
Mastocitos/patología , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/patología , Reticulina/metabolismo , Fibrosis , Humanos , Interpretación de Imagen Asistida por Computador , Inmunohistoquímica
12.
J Heart Lung Transplant ; 27(7): 701-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18582797

RESUMEN

BACKGROUND: Left ventricular assist devices (LVADs) cause an influx of mast cells into the failing heart, but the underlying mechanism is unknown. This study investigates the potential role of stem cell factor (SCF) and its receptor (c-Kit) in promoting the recruitment of mast cells during heart failure and after LVAD support. METHODS: Myocardial samples were collected from 10 end-stage heart failure patients undergoing LVAD implantation (pre-LVAD) and paired with samples taken at the time of orthotopic heart transplantation (post-LVAD). Biopsies of normal hearts served as controls. We assessed gene expression of SCF and c-Kit. In addition, we stained for SCF, c-Kit, tryptase and chymase, and utilized in situ hybridization to determine the origin of SCF. RESULTS: SCF mRNA and overall mast cell numbers were significantly increased (p < 0.01/p < 0.001) after LVAD support as compared with paired heart failure tissues. c-Kit mRNA was significantly increased post-LVAD compared with normal tissues (p < 0.05). The c-Kit protein was expressed only in cardiac mast cells. SCF mRNA was found in endothelial cells, myocytes and interstitial cells, as confirmed by antibody staining. CONCLUSIONS: LVADs cause an increase of SCF and c-Kit gene expression, which coincides with a surge of mast cells after ventricular unloading. This suggests that SCF functions as an important mediator for the recruitment of mast cells to the mechanically unloaded human heart.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Factor de Células Madre/biosíntesis , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Expresión Génica , Insuficiencia Cardíaca/terapia , Trasplante de Corazón , Corazón Auxiliar , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Transplantation ; 85(10): 1365-71, 2008 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-18497671

RESUMEN

Mast cells are best known as primary responders in allergic reactions, including anaphylaxis and asthma. However, recent studies have shown that mast cells are functionally diverse cells with immunoregulatory properties that influence both the innate and adaptive immunities. Mast cells are capable of producing an array of both proinflammatory and anti-inflammatory mediators, acting as antigen-presenting cells, and expressing a spectrum of costimulatory molecules. Moreover, mast cells seem to confer a certain degree of immune privilege to tissues in concert with T-regulatory cells and are essential players in fibrotic conditions. The following review of the literature serves to further define the role of mast cells in the immunologic reactions affecting transplanted solid organ grafts.


Asunto(s)
Mastocitos/fisiología , Trasplante de Órganos/fisiología , Enfermedad Aguda , Enfermedad Crónica , Rechazo de Injerto , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunosupresores/uso terapéutico , Inmunología del Trasplante , Trasplante Homólogo/fisiología
14.
Cardiovasc Res ; 74(1): 133-9, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17276423

RESUMEN

OBJECTIVE: Unloading of the rodent heart activates the fetal gene program, decreases peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARalpha-regulated gene expression (MCAD), and induces cardiomyocyte atrophy. NF-kappaB regulates the fetal gene program and PPARalpha-regulated gene expression during cardiac hypertrophy and induces atrophy in skeletal muscle. Our objective was to test the hypothesis that NF-kappaB is the regulator for activation of the fetal gene program, for downregulation of PPARalpha and PPARalpha-regulated gene expression, and for cardiomyocyte atrophy in the heart subjected to mechanical unloading. METHODS: Activation of the inhibitory kappa B kinase beta (IKKbeta)/NF-kappaB pathways were measured in the heterotopically transplanted rat heart using Western blotting of total and phospho-IKKbeta and using transcription factor ELISA's for the five members of the NF-kappaB family (p65 (Rel A), p105/p50, c-Rel, RelB, and p100/p52). In loss of function experiments, we transplanted hearts of p105/p50 knockout mice into wildtype mice and compared changes in gene expression and cardiomyocyte size with wildtype hearts transplanted into wildtype mice. RESULTS: Total and phospho-IKKbeta levels significantly increased in the transplanted heart seven days after surgery. The activation of IKKbeta was paralleled by increased DNA binding activity of p65 and p105/p50. Mechanical unloading induced myosin heavy chain beta expression and decreased cardiomyocyte size in hearts of both wildtype and p105/p050 knockout animals. In contrast, the downregulation of PPARalpha and MCAD was significantly attenuated or prevented in the hearts of p105/p50 knockout mice. CONCLUSIONS: The IKKbeta/p65/p50 pathway is activated in the unloaded rodent heart and a likely regulator for the downregulation of PPARalpha and PPARalpha-regulated gene expression.


Asunto(s)
Regulación hacia Abajo , Trasplante de Corazón , Miocardio/metabolismo , FN-kappa B/genética , PPAR alfa/metabolismo , Animales , Fenómenos Biomecánicos , Tamaño de la Célula , ADN/metabolismo , Regulación de la Expresión Génica , Genes del Desarrollo , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Unión Proteica , Ratas , Ratas Wistar
15.
J Heart Lung Transplant ; 24(4): 454-61, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15797748

RESUMEN

BACKGROUND: The heart undergoes repair and initiates protective mechanisms via ventricular unloading. We examined the presence of 2 markers in pre-unloaded and post-unloaded human cardiac tissue that are important indicators of cardiac failure, tumor necrosis factor-alpha and inducible nitric oxide synthase. We also measured 2 nuclear transcription factors, NFkappaB50 and NFkappaB65, comparing quantities and localizations to determine if mechanical unloading reduced their presence, as these markers are also thought to be indicators of impending heart failure. Amounts and localizations in patients that had been diagnosed with either ischemic or non-ischemic cardiomyopathy were compared after mechanical unloading with a left ventricular assist device. To establish that unloading had been achieved, levels of atrial natriuretic protein were determined. METHODS: Core biopsies were harvested at assist device implantation and removal. Fluorescence deconvolution microscopy image reconstructions of fluorescence probes were correlated with data obtained by western Blot and electrobility shift assays. RESULTS: Statistically significant differences in localization and amounts of tumor necrosis factor and nitric oxide synthase were seen between pre- and post-assist device samples. Amounts of tumor necrosis factor and nitric oxide synthase in ischemic tissue were increased at the time of assist device removal, but decreased in dilated or idiomyopathic samples. Ventricular unloading resulted in reduced levels of natriuretic protein, with the greatest reduction being seen in ischemic tissue. Both NFkappaB50 and NFkappaB65 increased in ischemic tissue, but only NFkappaB50 in non-ischemic samples. CONCLUSIONS: Changes in localization of the factors and altered levels of cytokine and nitric oxide synthase indicate that the heart switches to a "protective and repair" mode, and mechanical unloading allows this transition to occur. Observed changes were dependent on the etiology of the disease.


Asunto(s)
Factor Natriurético Atrial/ultraestructura , Cardiomiopatía Dilatada/metabolismo , Corazón Auxiliar , Isquemia Miocárdica/metabolismo , Miocardio/ultraestructura , Óxido Nítrico Sintasa/ultraestructura , Factor de Necrosis Tumoral alfa/ultraestructura , Factor Natriurético Atrial/metabolismo , Biomarcadores/metabolismo , Biopsia , Western Blotting , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/terapia , Remoción de Dispositivos , Electroforesis , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/ultraestructura , Humanos , Microscopía Fluorescente , Isquemia Miocárdica/patología , Isquemia Miocárdica/terapia , Miocardio/metabolismo , Óxido Nítrico Sintasa/metabolismo , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Surg Res ; 123(1): 118-25, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15652959

RESUMEN

BACKGROUND: Nonspecific inflammatory events following brain death may increase the intensity of the immunological host response. The present study investigated the course of pro-inflammatory molecules in heart, lung, kidney, and plasma after brain death induction. MATERIALS AND METHODS: Brain death was induced in five pigs by inflation of an intracranial Foley catheter and five pigs were sham-operated as controls. Each experiment was terminated 6 h after brain death/sham operation and the organs were harvested. We measured the mRNA and protein levels for TNF-alpha, IL-1beta, and IL-6 in heart, lung, kidney, and plasma. Additionally, the mRNA expression for IL-6R, ICAM-1, MCP-1, and TGF-beta was determined in each organ. RESULTS: After 6 h, the plasma cytokine levels were higher in the brain-dead animals than in the sham-operated. In heart, lung, and kidney there was an increase in IL-6 and IL-1beta following brain death, while TNF-alpha was up-regulated in lung only (P < 0.05). MCP-1 and TGF-beta were significantly higher in heart and lung and IL-6R increased in heart after brain death (P < 0.05). CONCLUSIONS: Brain death was associated with non-uniform cytokine expression patterns in the investigated organs. These expression patterns may cause variable pro-inflammatory priming resulting in different degrees of damage and explain the organ-specific variation in outcomes after transplantations.


Asunto(s)
Muerte Encefálica/metabolismo , Citocinas/genética , Riñón/metabolismo , Pulmón/metabolismo , Miocardio/metabolismo , Animales , Citocinas/análisis , Femenino , Perfilación de la Expresión Génica , Molécula 1 de Adhesión Intercelular/genética , Interleucina-1/genética , Interleucina-6/genética , Masculino , Especificidad de Órganos , ARN Mensajero/análisis , Receptores de Interleucina-6/genética , Porcinos , Factor de Necrosis Tumoral alfa/genética
18.
J Surg Res ; 115(1): 41-7, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14572771

RESUMEN

BACKGROUND: Most human pancreatic adenocarcinoma cells do not express somatostatin receptors, and somatostatin does not inhibit the growth of these cancers. We have demonstrated previously that somatostatin inhibits the growth of pancreatic cancers expressing somatostatin receptor subtype-2 (SSTR2), but not receptor-negative cancers. SSTR2 expression may be an important tumor-suppressor pathway that is lost in human pancreatic cancer. We hypothesized that SSTR2 gene transfer would restore the growth-inhibitory response of human pancreatic cancer to somatostatin. MATERIALS AND METHODS: Palpable human pancreatic adenocarcinoma tumors were established on the backs of nude mice by subcutaneous injection of cultured cells (Panc-1). The animals were divided into 5 groups (n = 10/group). Group I served as an untreated control. Group II received an intramuscular injection of the long-acting somatostatin analogue Sandostatin LAR. Group III received Lac-Z expressing adenovirus via intraperitoneal injection. Group IV received SSTR2 expressing adenovirus via intraperitoneal injection. Group V received SSTR2 expressing adenovirus via intraperitoneal injection and an intramuscular injection of Sandostatin LAR. The rate of tumor growth was assessed with calipers. After 28 days, the animals were anesthetized and exsanguanated, and the tumors were excised and weighed. Plasma somatostatin and octreotide levels were measured by radioimmunoassay. Expression of cell-surface somatostatin-receptor protein and known tumor-suppressor proteins was determined by reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry. RESULTS: Systemic delivery of SSTR2-expressing adenovirus by intraperitoneal injection resulted in expression of SSTR2 protein in the subcutaneous human pancreatic cancers. Final tumor weight was significantly decreased in the groups expressing SSTR2 receptors compared to the other 3 groups. Treatment with Sandostatin LAR increased plasma octreotide levels as determined by radioimmunoassay, but had no significant effect on tumor growth. Western blot analysis revealed an up-regulation of the cyclin-dependent kinase inhibitors p27 and p16 in the SSTR2 transfected tumors. CONCLUSIONS: Expression of SSTR2 by human pancreatic cancer causes significant slowing of tumor growth by a mechanism independent of exogenous somatostatin. The mechanism may involve up-regulation of known tumor-suppressor proteins. Restoration of SSTR2 gene expression deserves further study as a potential gene-therapy strategy in human pancreatic cancer.


Asunto(s)
Adenocarcinoma/patología , Expresión Génica , Neoplasias Pancreáticas/patología , Receptores de Somatostatina/genética , Receptores de Somatostatina/fisiología , Transfección , Adenoviridae/genética , Animales , División Celular/efectos de los fármacos , Vectores Genéticos , Humanos , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Octreótido/administración & dosificación , Octreótido/sangre , ARN Mensajero/análisis , Somatostatina/sangre , Somatostatina/farmacología , Trasplante Heterólogo , Células Tumorales Cultivadas
19.
Cardiology ; 100(1): 17-22, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12975541

RESUMEN

BACKGROUND: Left ventricular assist device (LVAD) support of the failing human heart improves myocyte function and increases cell survival. One potential mechanism underlying this phenomenon is activation of the protein kinase B (PKB)/Akt/glycogen synthase kinase-3beta (GSK-3beta) survival pathway. METHODS AND RESULTS: Left ventricular tissue was obtained both at the time of implantation and explantation of the LVAD (n = 11). Six patients were diagnosed with idiopathic dilated cardiomyopathy, 4 patients with ischemic cardiomyopathy and 1 patient with peripartum cardiomyopathy. The mean duration of LVAD support was 205 +/- 35 days. Myocyte diameter and phosphorylation of ERK were used as indices for reverse remodeling. Transcript levels of genes required for the activation of PKB/Akt (insulin-like growth factor-1, insulin receptor substrate-1) were measured by quantitative RT-PCR. In addition, we measured the relative activity of PKB/Akt and GSK-3beta, and assayed for molecular and histological indices of PKB/Akt activation (cyclooxygenase mRNA levels and glycogen levels). Myocyte diameter and phosphorylation of ERK decreased with LVAD support. In contrast, none of the components of the PKB/Akt/GSK-3beta pathway changed significantly with mechanical unloading. CONCLUSION: The PKB/Akt/GSK-3beta pathway is not activated during LVAD support. Other signaling pathways must be responsible for the improvement of cellular function and cell survival during LVAD support.


Asunto(s)
Glucógeno Sintasa Quinasa 3/genética , Insuficiencia Cardíaca/enzimología , Corazón Auxiliar , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/genética , Actinas/genética , Adulto , Supervivencia Celular , Ciclooxigenasa 2 , Activación Enzimática , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Ventrículos Cardíacos/patología , Humanos , Proteínas Sustrato del Receptor de Insulina , Isoenzimas/genética , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Fosfoproteínas/genética , Prostaglandina-Endoperóxido Sintasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptor IGF Tipo 1/genética , Transcripción Genética
20.
J Mol Cell Cardiol ; 34(1): 51-62, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11812164

RESUMEN

Coronary microembolization is a frequent complication of atherosclerotic plaque rupture in acute coronary syndromes and during coronary interventions. Experimental coronary microembolization results in progressive contractile dysfunction associated with a local inflammation. We studied the causal role of tumor necrosis factor-alpha (TNF-alpha) in the progressive contractile dysfunction resulting from coronary microembolization. Anesthetized dogs were subjected to either coronary microembolization with infusion of 3.000 microspheres (42 microm diameter) per ml coronary inflow into the left circumflex coronary artery (n=9), or to intracoronary infusion of recombinant human TNF-alpha without microembolization (n=4), or to treatment with anti-murine TNF-alpha sheep antibodies prior to microembolization (n=4). Posterior systolic wall thickening (PWT; sonomicrometry) decreased from 21.1+/-5.3% (s.d.) at baseline to 5.5+/-2.2% (P<0.05) at 8 h after microembolization. Infarct size (1.8+/-1.9%; TTC and histology) and the amount of apoptosis (<0.1%; TUNEL and DNA-laddering) were small. TNF-alpha at the protein level (WEHI cytolytic assay) was increased and localized to leukocytes (immunostaining), which were increased in number (quantitative histology). In situ hybridization for TNF-alpha mRNA identified viable cardiomyocytes surrounding the microinfarcts as the major source of TNF-alpha. Supporting the role of TNF-alpha, infusion of TNF-alpha without microembolization decreased PWT from 27.3+/-6.9% at baseline to 10.1+/-4.9% after 8 h (P<0.05); in contrast, in the presence of TNF-alpha antibodies, microembolization no longer reduced PWT (19.3+/-7.0% at baseline v 16.9+/-5.0% at 8 h). In conclusion, TNF-alpha is the mediator responsible for the profound contractile dysfunction following coronary microembolization.


Asunto(s)
Arteriosclerosis/fisiopatología , Circulación Coronaria/fisiología , Contracción Miocárdica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Animales , Apoptosis , Circulación Coronaria/efectos de los fármacos , Perros , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Leucocitos/metabolismo , Microcirculación/fisiología , Microscopía Fluorescente , Infarto del Miocardio , Necrosis , ARN Mensajero/metabolismo , Flujo Sanguíneo Regional , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA