Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Biol Rep ; 49(5): 3481-3490, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35083615

RESUMEN

BACKGROUND: Pulmonary fibrosis is a debilitating condition with limited therapeutic avenues. The pathogenicity of pulmonary fibrosis constitutes involvement of cellular proliferation, activation, and transformational changes of fibroblast to myofibroblasts. It is a progressive lung disease and is primarily characterized by aberrant accumulation of extracellular matrix proteins in the lungs with poor prognosis. The inflammatory response in the pathogenesis of lung fibrosis is suggested because of release of several cytokines; however, the underlying mechanism remains undefined. A genetic model is the appropriate way to delineate the underlying mechanism of pulmonary fibrosis. METHODS AND RESULTS: In this report, we have used cc-10 promoter based IκBα mutant mice (IKBM, an inhibitor of NF-κB) which were challenged with bleomycin (BLM). Compared to wild-type (WT) mice, the IKBM mice showed significant reduction in several fibrotic, vascular, and inflammatory genes. Moreover, we have identified a new set of dysregulated microRNAs (miRNAs) by miRNA array analysis in BLM-induced WT mice. Among these miRNAs, let-7a-5p and miR-503-5p were further analyzed. Our data showed that these two miRNAs were upregulated in WT-BLM and were reduced in IKBM-BLM mice. Bioinformatic analyses showed that let-7a-5p and miR-503-5p target for endothelin1 and bone morphogenic receptor 1A (BMPR1A), respectively, and were downregulated in WT-BLM mice indicating a link in pulmonary fibrosis. CONCLUSION: We concluded that inhibition of NF-κB and modulation of let-7a-5p and miR-503-5p contribute a pivotal role in pulmonary fibrosis and may be considered as possible therapeutic target for the clinical management of lung fibrosis.


Asunto(s)
MicroARNs , FN-kappa B , Fibrosis Pulmonar , Animales , Bleomicina , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo
2.
J Dual Diagn ; 16(3): 322-335, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32493131

RESUMEN

Objective: Alcohol use and alcohol use disorders (AUDs) are an increasing concern among veterans, particularly those from recent conflicts in Iraq and Afghanistan. The study of biomarkers in alcohol use and AUD has moved to enhancing the understanding of the development and maintenance of AUDs, as well as investigating its association with clinical severity and potential predictors of treatment response. Cortisol, a glucocorticoid known as a stress hormone, has been linked with both stress and trauma, as well as increased alcohol suppression effects. Method/Results: The present review summarizes existing literature and presents suggestions for future research to evaluate whether cortisol may be a possible biomarker of alcohol use disorder risk in combat veterans. Specifically, aspects of combat deployments and high levels of PTSD, coupled with the stress of reintegration may dysregulate cortisol and increase risk to AUD. There may also be bidirectional impacts, such that alcohol is used as a coping mechanism and can dysregulate hypothalamic pituitary adrenal (HPA) axis functioning and cortisol. Conclusions: In the context of this framework, cortisol may serve as a biomarker for the development of AUD, as well as a biomarker of risk or relapse. This review ends with both theoretical and clinical implications, as well as directions for future research.


Asunto(s)
Consumo de Bebidas Alcohólicas/sangre , Alcoholismo/sangre , Alcoholismo/diagnóstico , Biomarcadores/sangre , Trastornos de Combate/sangre , Hidrocortisona/sangre , Trastornos por Estrés Postraumático/sangre , Veteranos , Humanos , Proyectos de Investigación
3.
J Mol Cell Cardiol ; 99: 100-112, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27539860

RESUMEN

Retinoic acid receptor (RAR) has been implicated in pathological stimuli-induced cardiac remodeling. To determine whether the impairment of RARα signaling directly contributes to the development of heart dysfunction and the involved mechanisms, tamoxifen-induced myocardial specific RARα deletion (RARαKO) mice were utilized. Echocardiographic and cardiac catheterization studies showed significant diastolic dysfunction after 16wks of gene deletion. However, no significant differences were observed in left ventricular ejection fraction (LVEF), between RARαKO and wild type (WT) control mice. DHE staining showed increased intracellular reactive oxygen species (ROS) generation in the hearts of RARαKO mice. Significantly increased NOX2 (NADPH oxidase 2) and NOX4 levels and decreased SOD1 and SOD2 levels were observed in RARαKO mouse hearts, which were rescued by overexpression of RARα in cardiomyocytes. Decreased SERCA2a expression and phosphorylation of phospholamban (PLB), along with decreased phosphorylation of Akt and Ca2+/calmodulin-dependent protein kinase II δ (CaMKII δ) was observed in RARαKO mouse hearts. Ca2+ reuptake and cardiomyocyte relaxation were delayed by RARα deletion. Overexpression of RARα or inhibition of ROS generation or NOX activation prevented RARα deletion-induced decrease in SERCA2a expression/activation and delayed Ca2+ reuptake. Moreover, the gene and protein expression of RARα was significantly decreased in aged or metabolic stressed mouse hearts. RARα deletion accelerated the development of diastolic dysfunction in streptozotocin (STZ)-induced type 1 diabetic mice or in high fat diet fed mice. In conclusion, myocardial RARα deletion promoted diastolic dysfunction, with a relative preserved LVEF. Increased oxidative stress have an important role in the decreased expression/activation of SERCA2a and Ca2+ mishandling in RARαKO mice, which are major contributing factors in the development of diastolic dysfunction. These data suggest that impairment of cardiac RARα signaling may be a novel mechanism that is directly linked to pathological stimuli-induced diastolic dysfunction.


Asunto(s)
Calcio/metabolismo , Diástole , Miocardio/metabolismo , Estrés Oxidativo , Receptor alfa de Ácido Retinoico/deficiencia , Disfunción Ventricular/genética , Disfunción Ventricular/metabolismo , Disfunción Ventricular/fisiopatología , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Activación Enzimática , Fibrosis , Eliminación de Gen , Expresión Génica , Masculino , Ratones , Miocitos Cardíacos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Disfunción Ventricular/diagnóstico por imagen
4.
J Cell Physiol ; 231(8): 1737-44, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26627308

RESUMEN

Thymosin beta-4 (Tß4) is a ubiquitous protein with many properties relating to cell proliferation and differentiation that promotes wound healing and modulates inflammatory mediators. However, the role of Tß4 in cardiomyocyte hypertrophy is currently unknown. The purpose of this study was to determine the cardio-protective effect of Tß4 in angiotensin II (Ang II)-induced cardiomyocyte growth. Neonatal rat ventricular cardiomyocytes (NRVM) were pretreated with Tß4 followed by Ang II stimulation. Cell size, hypertrophy marker gene expression and Wnt signaling components, ß-catenin, and Wnt-induced secreted protein-1 (WISP-1) were evaluated by quantitative real-time PCR, Western blotting and fluorescent microscopy. Pre-treatment of Tß4 resulted in reduction of cell size, hypertrophy marker genes and Wnt-associated gene expression, and protein levels; induced by Ang II in cardiomyocyte. WISP-1 was overexpressed in NRVM and, the effect of Tß4 in Ang II-induced cardiomyocyte growth was evaluated. WISP-1 overexpression promoted cardiomyocytes growth and was reversed by pretreatment with Tß4. This is the first report which demonstrates that Tß4 targets Wnt/WISP-1 to protect Ang II-induced cardiomyocyte growth. J. Cell. Physiol. 231: 1737-1744, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Angiotensina II/toxicidad , Proteínas CCN de Señalización Intercelular/metabolismo , Cardiomegalia/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Timosina/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Transporte Activo de Núcleo Celular , Animales , Animales Recién Nacidos , Proteínas CCN de Señalización Intercelular/genética , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas/genética , Ratas Sprague-Dawley , Transfección , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
J Mol Cell Cardiol ; 57: 106-18, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23395853

RESUMEN

Diabetic cardiomyopathy (DCM) is a significant contributor to the morbidity and mortality associated with diabetes and metabolic syndrome. Retinoids, through activation of retinoic acid receptor (RAR) and retinoid x receptor (RXR), have been linked to control glucose and lipid homeostasis, with effects on obesity and diabetes. However, the functional role of RAR and RXR in the development of DCM remains unclear. Zucker diabetic fatty (ZDF) and lean rats were treated with Am580 (RARα agonist) or LGD1069 (RXR agonist) for 16 weeks, and cardiac function and metabolic alterations were determined. Hyperglycemia, hyperlipidemia and insulin resistance were observed in ZDF rats. Diabetic cardiomyopathy was characterized in ZDF rats by increased oxidative stress, apoptosis, fibrosis, inflammation, activation of MAP kinases and NF-κB signaling and diminished Akt phosphorylation, along with decreased glucose transport and increased cardiac lipid accumulation, and ultimately diastolic dysfunction. Am580 and LGD1069 attenuated diabetes-induced cardiac dysfunction and the pathological alterations, by improving glucose tolerance and insulin resistance; facilitating Akt activation and glucose utilization, and attenuating oxidative stress and interrelated MAP kinase and NF-κB signaling pathways. Am580 inhibited body weight gain, attenuated the increased cardiac fatty acid uptake, ß-oxidation and lipid accumulation in the hearts of ZDF rats. However, LGD1069 promoted body weight gain, hyperlipidemia and cardiac lipid accumulation. In conclusion, our data suggest that activation of RAR and RXR may have therapeutic potential in the treatment of diabetic cardiomyopathy. However, further studies are necessary to clarify the role of RAR and RXR in the regulation of lipid metabolism and homeostasis.


Asunto(s)
Benzoatos/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Receptores de Ácido Retinoico/agonistas , Receptores X Retinoide/agonistas , Tetrahidronaftalenos/farmacología , Animales , Benzoatos/uso terapéutico , Bexaroteno , Glucemia , Colágeno/genética , Colágeno/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/metabolismo , Evaluación Preclínica de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Insulina/sangre , Metabolismo de los Lípidos , Masculino , Miocardio/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Zucker , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/metabolismo , Transducción de Señal , Tetrahidronaftalenos/uso terapéutico
6.
J Cell Physiol ; 228(2): 380-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22718360

RESUMEN

We have previously shown that retinoic acid (RA) has protective effects on high glucose (HG)-induced cardiomyocyte apoptosis. To further elucidate the molecular mechanisms of RA effects, we determined the interaction between nuclear factor (NF)-κB and RA signaling. HG induced a sustained phosphorylation of IKK/IκBα and transcriptional activation of NF-κB in cardiomyocytes. Activated NF-κB signaling has an important role in HG-induced cardiomyocyte apoptosis and gene expression of interleukin-6 (IL-6), tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein-1 (MCP-1). All-trans RA (ATRA) and LGD1069, through activation of RAR/RXR-mediated signaling, inhibited the HG-mediated effects in cardiomyocytes. The inhibitory effect of RA on NF-κB activation was mediated through inhibition of IKK/IκBα phosphorylation. ATRA and LGD1069 treatment promoted protein phosphatase 2A (PP2A) activity, which was significantly suppressed by HG stimulation. The RA effects on IKK and IκBα were blocked by okadaic acid or silencing the expression of PP2Ac-subunit, indicating that the inhibitory effect of RA on NF-κB is regulated through activation of PP2A and subsequent dephosphorylation of IKK/IκBα. Moreover, ATRA and LGD1069 reversed the decreased PP2A activity and inhibited the activation of IKK/IκBα and gene expression of MCP-1, IL-6, and TNF-α in the hearts of Zucker diabetic fatty rats. In summary, our findings suggest that the suppressed activation of PP2A contributed to sustained activation of NF-κB in HG-stimulated cardiomyocytes; and that the protective effect of RA on hyperglycemia-induced cardiomyocyte apoptosis and inflammatory responses is partially regulated through activation of PP2A and suppression of NF-κB-mediated signaling and downstream targets.


Asunto(s)
Apoptosis/efectos de los fármacos , Citoprotección , Hiperglucemia/complicaciones , Miocitos Cardíacos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Animales , Bexaroteno , Citocinas/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Masculino , Fosforilación , Proteína Fosfatasa 2/efectos de los fármacos , Ratas , Ratas Zucker , Tetrahidronaftalenos/farmacología
7.
Exp Ther Med ; 26(4): 468, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37664684

RESUMEN

War veterans, in particular, are more prone to mental illness as they are more likely to have encountered multiple traumatic brain injuries (TBIs) whilst serving on active duty in war zone areas. A TBI is known to cause mortality or serious neurological disabilities among survivors and elicits a number of pathological processes, including neuroinflammation and blood brain barrier (BBB) disruption, leading to secondary brain damage and subsequent impairment of the neurovascular unit. Although several drugs exhibit promising effects for TBI, the repertoire of currently available therapeutic strategies remains limited. Thymosin 4 (Tß4) is a 43-amino acid G-acting sequestering peptide that confers neuroprotective potential in TBI models. However, its role in BBB function remains unclear. Further research into the mechanism of BBB disruption induced by TBI and its specific role in neurovascular pathophysiology is necessary. In the present study, the protective effects of Tß4 in lipopolysaccharide (LPS)-stimulated gene expression of several tight junction proteins, inflammatory genes, apoptotic genes, and adhesion genes in human brain microvascular endothelial cells (hBMVECs), one of the pivotal cell types in the BBB, were reported. The results suggested that pretreatment with Tß4 reversed the LPS-induced damage of BBB components in hBMVECs. Furthermore, these results identified neuregulin 1 as a possible target for Tß4. Therefore, it is proposed that Tß4-mediated cellular signaling in hBMVEC may be vital for understanding the association between the BBB and TBI pathophysiology, which warrants further investigation.

8.
J Cell Physiol ; 227(6): 2632-44, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21882190

RESUMEN

The biological actions of retinoids are mediated by nuclear retinoic acid receptors (RARs) and retinoid X receptors (RXRs). We have recently reported that decreased expression of RARα and RXRα has an important role in high glucose (HG)-induced cardiomyocyte apoptosis. However, the regulatory mechanisms of HG effects on RARα and RXRα remain unclear. Using neonatal cardiomyocytes, we found that ligand-induced promoter activity of RAR and RXR was significantly suppressed by HG. HG promoted protein destabilization and serine-phosphorylation of RARα and RXRα. Proteasome inhibitor MG132 blocked the inhibitory effect of HG on RARα and RXRα. Inhibition of intracellular reactive oxidative species (ROS) abolished the HG effect. In contrast, H(2)O(2) stimulation suppressed the expression and ligand-induced promoter activity of RARα and RXRα. HG promoted phosphorylation of ERK1/2, JNK and p38 MAP kinases, which was abrogated by an ROS inhibitor. Inhibition of JNK, but not ERK and p38 activity, reversed HG effects on RARα and RXRα. Activation of JNK by over expressing MKK7 and MEKK1, resulted in significant downregulation of RARα and RXRα. Ligand-induced promoter activity of RARα and RXRα was also suppressed by overexpression of MEKK1. HG-induced cardiomyocyte apoptosis was potentiated by activation of JNK, and prevented by all-trans retinoic acid and inhibition of JNK. Silencing the expression of RARα and RXRα activated the JNK pathway. In conclusion, HG-induced oxidative stress and activation of the JNK pathway negatively regulated expression/activation of RAR and RXR. The impaired RAR/RXR signaling and oxidative stress/JNK pathway forms a vicious circle, which significantly contributes to hyperglycemia induced cardiomyocyte apoptosis.


Asunto(s)
Glucosa/metabolismo , Hiperglucemia/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/enzimología , Estrés Oxidativo , Receptores de Ácido Retinoico/metabolismo , Receptor alfa X Retinoide/metabolismo , Transducción de Señal , Alitretinoína , Animales , Animales Recién Nacidos , Apoptosis , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hiperglucemia/genética , Hiperglucemia/patología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Receptor alfa X Retinoide/agonistas , Receptor alfa X Retinoide/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Activación Transcripcional , Transfección , Tretinoina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Cells ; 11(13)2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35805118

RESUMEN

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric condition which develops either due to stress or witnessing a traumatic situation. PTSD is characterized by acute and chronic stress response exhibit anxiety, fear, and an increased inflammatory etiology. Inflammation contributes a critical role in several parts of the brain that control fear and flashback cognatic function. It is known that impairment of the neurological circuit leads to the development of PTSD. Evidence has suggested that dysregulation of the sympathetic nervous system and hypothalamic-pituitary adrenal (HPA) axis and inflammatory responsiveness are pivotal and a greater risk in PTSD. NF-κB, a master regulator for inflammation, has been showed to modulate memory reconsolidation and synaptic plasticity; however, NF-κB's association with PTSD remain elusive. In this review, we provide relevant findings regarding NF-κB activity in various components of brain and describe a potential mechanism linking PTSD using preclinical and clinical models. We envisage NF-κB signaling as a crucial mediator for inflammation, cognitive function, memory restoration and behavioral actions of stress and suggest that it could be used for therapeutic intervention in PTSD.


Asunto(s)
Trastornos por Estrés Postraumático , Humanos , Inflamación/complicaciones , FN-kappa B , Plasticidad Neuronal , Sistema Hipófiso-Suprarrenal , Trastornos por Estrés Postraumático/etiología , Trastornos por Estrés Postraumático/psicología
10.
J Cell Physiol ; 226(5): 1292-307, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20945395

RESUMEN

Diabetes mellitus (DM) is a primary risk factor for cardiovascular diseases and heart failure. Activation of the retinoic acid receptor (RAR) and retinoid X receptor (RXR) has an anti-diabetic effect; but, a role in diabetic cardiomyopathy remains unclear. Using neonatal and adult cardiomyocytes, we determined the role of RAR and RXR in hyperglycemia-induced apoptosis and expression of renin-angiotensin system (RAS) components. Decreased nuclear expression of RARα and RXRα, activation of apoptotic signaling and cell apoptosis was observed in high glucose (HG) treated neonatal and adult cardiomyocytes and diabetic hearts in Zucker diabetic fatty (ZDF) rats. HG-induced apoptosis and reactive oxygen species (ROS) generation was prevented by both RAR and RXR agonists. Silencing expression of RARα and RXRα, by small interference RNA, promoted apoptosis under normal conditions and significantly enhanced HG-induced apoptosis, indicating that RARα and RXRα are required in regulating cell apoptotic signaling. Blocking angiotensin type 1 receptor (AT(1) R); but, not AT(2) R, attenuated HG-induced apoptosis and ROS generation. Moreover, HG induced gene expression of angiotensinogen, renin, AT(1) R, and angiotensin II (Ang II) synthesis were inhibited by RARα agonists and promoted by silencing RARα. Activation of RXRα, downregulated the expression of AT(1) R; and RXRα silencing accelerated HG induced expression of angiotensinogen and Ang II synthesis, whereas there was no significant effect on renin gene expression. These results indicate that reduction in the expression of RARα and RXRα has an important role in hyperglycemia mediated apoptosis and expression of RAS components. Activation of RAR/RXR signaling protects cardiomyocytes from hyperglycemia, by reducing oxidative stress and inhibition of the RAS.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Ácido Retinoico/agonistas , Sistema Renina-Angiotensina/efectos de los fármacos , Receptor alfa X Retinoide/agonistas , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Angiotensina II/metabolismo , Antagonistas de Receptores de Angiotensina/farmacología , Animales , Animales Recién Nacidos , Glucemia/metabolismo , Células Cultivadas , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/patología , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Sistema Renina-Angiotensina/genética , Receptor alfa de Ácido Retinoico , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Transducción de Señal/genética , Factores de Tiempo
11.
Psychiatry Res ; 305: 114252, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34739954

RESUMEN

Posttraumatic stress disorder (PTSD) is a common psychiatric disorder for military Veterans, characterized by hyperarousal, intrusive thoughts, flashbacks, hypervigilance, and distress after experiencing traumatic events. Some of the known physiological effects of PTSD include hypothalamic-pituitary-adrenal (HPA)-axis imbalance, a cortical function resulting in neuronal deficit and changes in behavior. Moreover, excessive discharge of inflammatory molecules and a dysregulated immune system are implicated in the pathophysiology of PTSD. Due to complex nature of this disorder, the biological underpinnings of PTSD remain inexplicable. Investigating novel biomarkers to understanding the pathogenesis of PTSD may reflect the underlying molecular network for therapeutic use and treatment. Circulatory microRNAs (miRNAs) and exosomes are evolving biomarkers that have shown a key role in psychiatric and neurological disorders including PTSD. Given the unique nature of combat trauma, as well as evidence that a large portion of Veterans do not benefit from frontline treatments, focus on veterans specifically is warranted. In the present review, we delineate the identification and role of several miRNAs in PTSD among veterans. An association of miRNA with HPA-axis regulation through FKBP5, a key modulator in PTSD is discussed as an emerging molecule in psychiatric diseases. We conclude that miRNAs may be used as circulatory biomarker detection in Veterans with PTSD.


Asunto(s)
MicroARNs , Trastornos por Estrés Postraumático , Veteranos , Biomarcadores , Humanos , Sistema Hipófiso-Suprarrenal , Trastornos por Estrés Postraumático/diagnóstico por imagen , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/terapia , Veteranos/psicología
12.
J Am Heart Assoc ; 8(21): e012880, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31640463

RESUMEN

Background Cardiac fibrosis occurs because of disruption of the extracellular matrix network leading to myocardial dysfunction. Angiotensin II (AngII) has been implicated in the development of cardiac fibrosis. Recently, microRNAs have been identified as an attractive target for therapeutic intervention in cardiac pathologies; however, the underlying mechanism of microRNAs in cardiac fibrosis remains unclear. Next-generation sequencing analysis identified a novel characterized microRNA, miR-1954, that was significantly reduced in AngII-infused mice. The finding led us to hypothesize that deficiency of miR-1954 triggers cardiac fibrosis. Methods and Results A transgenic mouse was created using α-MHC (α-myosin heavy chain) promoter and was challenged with AngII infusion. AngII induced cardiac hypertrophy and remodeling. The in vivo overexpression of miR-1954 showed significant reduction in cardiac mass and blood pressure in AngII-infused mice. Further analysis showed significant reduction in cardiac fibrotic genes, hypertrophy marker genes, and an inflammatory gene and restoration of a calcium-regulated gene (Atp2a2 [ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2]; also known as SERCA2), but no changes were observed in apoptotic genes. THBS1 (thrombospondin 1) is indicated as a target gene for miR-1954. Conclusions Our findings provide evidence, for the first time, that miR-1954 plays a critical role in cardiac fibrosis by targeting THBS1. We conclude that promoting the level of miR-1954 would be a promising strategy for the treatment of cardiac fibrosis.


Asunto(s)
Fibrosis/genética , MicroARNs/genética , Miocardio/patología , Remodelación Ventricular/genética , Actinas/metabolismo , Angiotensina II/farmacología , Animales , Cardiomegalia/genética , Cardiomegalia/terapia , Caspasa 3/metabolismo , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Modelos Animales de Enfermedad , Fibrosis/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , Interleucina-6/metabolismo , Ratones Transgénicos , Tamaño de los Órganos , Proteína de Unión al Calcio S100A4/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
13.
Biochim Biophys Acta ; 1772(9): 1093-102, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17611083

RESUMEN

Causes of retinoid resistance often observed in neuroblastomas are unknown. We studied all trans-retinoic acid (RA) signaling in neuroblastoma cells differing in N-myc levels in terms of neurite formation, expression of tissue transglutaminase, neuronal marker proteins, matrix metalloproteinases (MMPs), and activation of Rac1 and Cdc42. Poor invasiveness observed in SH-SY5Y, LA-N-5, and SMS-KCNR cells was associated with RA-induced neurite formation, Cdc42 activation and N-myc down regulation; expression of constitutively active Cdc42 down regulated N-myc expression and reduced invasion in RA-resistant SK-N-BE(2) and IMR32 cells. RA treatment for 24 h transiently increased invasion and expression of MMP9 in SH-SY5Y, LA-N-5 and MMP2 in SMS-KCNR cells. MMP inhibition prevented RA-induced neurite formation indicating a role in differentiation. Variation in RA signaling thus follows a defined pattern and relates to invasive potential. A defective RA signaling might result in retinoid resistance and unpredictable clinical outcome observed in some neuroblastomas.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Metaloproteinasas de la Matriz/fisiología , Neuroblastoma/patología , Transducción de Señal , Tretinoina/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc , Humanos , Neuritas/efectos de los fármacos , Neuroblastoma/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
14.
Diabetes ; 55(12): 3326-34, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130476

RESUMEN

Diabetes is a risk factor for neuronal dysfunction. Impairment in signaling mechanisms that regulate differentiation of neurons is hypothesized to be one of the main causes of neuronal dysfunction. Retinoic acid, a physiologically active retinoid synthesized from vitamin A, regulates neuronal differentiation during embryonic development and is required for maintenance of plasticity in differentiated neurons. To date, little is known about the molecular events underlying hyperglycemia-induced complications in the central nervous system (CNS). Here, we provide evidence, in a diabetes rat model, of hyperglycemia-induced oxidative stress along with apoptotic stress in developing cortical neurons isolated from 16-day-old rat embryos. We also demonstrate impaired retinoic acid signaling that is involved in neuronal differentiation. Retinoic acid-induced neurite outgrowth and expression of neuronal markers were reduced in this model. The activation of small-molecular weight G-protein, Rac1, that mediates these effects was also reduced. Retinoic acid applied at a physiological concentration significantly decreased hyperglycemia-induced oxidative stress and thus supported the antioxidant defense system. These results suggest that diabetes-induced neuronal complications during pregnancy might be due to impaired retinoic acid signaling, and exogenously administered retinoic acid may be useful against CNS complications associated with diabetes.


Asunto(s)
Corteza Cerebral/patología , Diabetes Mellitus Experimental/patología , Hiperglucemia/fisiopatología , Neuronas/patología , Estrés Oxidativo/fisiología , Complicaciones del Embarazo/patología , Tretinoina/farmacología , Proteína de Unión al GTP rac1/fisiología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Femenino , Hiperglucemia/patología , Estrés Oxidativo/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Proteína de Unión al GTP rac1/efectos de los fármacos
15.
J Clin Med ; 3(2): 566-94, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26237391

RESUMEN

Diabetic cardiomyopathy (DCM), a significant contributor to morbidity and mortality in diabetic patients, is characterized by ventricular dysfunction, in the absence of coronary atherosclerosis and hypertension. There is no specific therapeutic strategy to effectively treat patients with DCM, due to a lack of a mechanistic understanding of the disease process. Retinoic acid, the active metabolite of vitamin A, is involved in a wide range of biological processes, through binding and activation of nuclear receptors: retinoic acid receptors (RAR) and retinoid X receptors (RXR). RAR/RXR-mediated signaling has been implicated in the regulation of glucose and lipid metabolism. Recently, it has been reported that activation of RAR/RXR has an important role in preventing the development of diabetic cardiomyopathy, through improving cardiac insulin resistance, inhibition of intracellular oxidative stress, NF-κB-mediated inflammatory responses and the renin-angiotensin system. Moreover, downregulated RAR/RXR signaling has been demonstrated in diabetic myocardium, suggesting that impaired RAR/RXR signaling may be a trigger to accelerate diabetes-induced development of DCM. Understanding the molecular mechanisms of retinoid receptors in the regulation of cardiac metabolism and remodeling under diabetic conditions is important in providing the impetus for generating novel therapeutic approaches for the prevention and treatment of diabetes-induced cardiac complications and heart failure.

16.
Exp Biol Med (Maywood) ; 236(12): 1389-401, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22087020

RESUMEN

It is well recognized by orthopedic surgeons that fractures of alcoholics are more difficult to heal successfully and have a higher incidence of non-union, but the mechanism of alcohol's effect on fracture healing is unknown. In order to give direction for the study of the effects of alcohol on fracture healing, we propose to identify gene expression and microRNA changes during the early stages of fracture healing that might be attributable to alcohol consumption. As the inflammatory stage appears to be the most critical for successful fracture healing, this paper focuses on the events at day three following fracture or the stage of inflammation. Sprague-Dawley rats were placed on an ethanol-containing or pair-fed Lieber and DeCarli diet for four weeks prior to surgical fracture. Following insertion of a medullary pin, a closed mid-diaphyseal fracture was induced using a Bonnarens and Einhorn fracture device. At three days' post-fracture, the region of the fracture calluses was harvested from the right hind-limb. RNA was extracted and microarray analysis was conducted against the entire rat genome. There were 35 genes that demonstrated significant increased expression due to alcohol consumption and 20 that decreased due to alcohol. In addition, the expression of 20 microRNAs was increased and six decreased. In summary, while it is recognized that mRNA levels may or may not represent protein levels successfully produced by the cell, these studies reveal changes in gene expression that support the hypothesis that alcohol consumption affects events involved with inflammation. MicroRNAs are known to modulate mRNA and these findings were consistent with much of what was seen with mRNA microarray analysis, especially the involvement of smad4 which was demonstrated by mRNA microarray, microRNA and polymerase chain reaction.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Etanol/farmacología , Curación de Fractura/efectos de los fármacos , Animales , Etanol/sangre , Curación de Fractura/genética , Genoma , Inflamación/genética , Masculino , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero , Ratas , Ratas Sprague-Dawley
17.
Cardiovasc Res ; 90(1): 88-96, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21131638

RESUMEN

AIMS: Angiotensin II (Ang II) stimulates cardiac remodelling and fibrosis in the mechanically overloaded myocardium. Although Rho GTPases regulate several cellular processes, including myocardial remodelling, involvement in mediating mechanical stretch-induced regulation of angiotensinogen (Ao), the precursor to Ang II, remains to be determined. We, therefore, examined the role and associated signalling mechanisms of Rho GTPases (Rac1 and RhoA) in regulation of Ao gene expression in a stretch model of neonatal rat cardiac fibroblasts (CFs). METHODS AND RESULTS: CFs were plated on deformable stretch membranes. Equiaxial mechanical stretch caused significant activation of both Rac1 and RhoA within 2-5 min. Rac1 activity returned to control levels after 4 h, whereas RhoA remained at a high level of activity until the end of the stretch period (24 h). Mechanical stretch initially caused a moderate decrease in Ao gene expression, but was significantly increased at 8-24 h. RhoA had a major role in mediating both the stretch-induced inhibition of Ao at 4 h and the subsequent upregulation of Ao expression at 24 h. ß1 integrin receptor blockade by Tac ß1 expression impaired acute (2 and 15 min) stretch-induced Rac1 activation, but increased RhoA activity. Molecular experiments revealed that Ao gene expression was inhibited by Rac1 through both JNK-dependent and independent mechanisms, and stimulated by RhoA through a p38-dependent mechanism. CONCLUSION: These results indicate that stretch-induced activation of Rac1 and RhoA differentially regulates Ao gene expression by modulating p38 and JNK activation.


Asunto(s)
Angiotensinógeno/metabolismo , Forma de la Célula , Fibroblastos/enzimología , Mecanotransducción Celular , Miocardio/enzimología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Angiotensinógeno/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática , Regulación de la Expresión Génica , Integrina beta1/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocardio/citología , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Estrés Mecánico , Factores de Tiempo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/genética
18.
Am J Physiol Heart Circ Physiol ; 294(2): H633-44, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18156191

RESUMEN

This study was designed to determine the effect of all-trans retinoic acid (RA) on the development of cardiac remodeling in a pressure overload rat model. Male Sprague-Dawley rats were subjected to sham operation and the aortic constriction procedure. A subgroup of sham control and aortic constricted rats were treated with RA for 5 mo after surgery. Pressure-overloaded rats showed significantly increased interstitial and perivascular fibrosis, heart weight-to-body weight ratio, and gene expression of atrial natriuretic peptide and brain natriuretic peptide. Echocardiographic analysis showed that pressure overload induced systolic and diastolic dysfunction, as evidenced by decreased fractional shortening, ejection fraction, stroke volume, and increased E-to-E(a) ratio and isovolumic relaxation time. RA treatment prevented the above changes in cardiac structure and function and hypertrophic gene expression in pressure-overloaded rats. RA restored the ratio of Bcl-2 to Bax, inhibited cleavage of caspase-3 and -9, and prevented the decreases in the levels of SOD-1 and SOD-2. Pressure overload-induced phosphorylation of ERK1/2, JNK, and p38 was inhibited by RA, via upregulation of mitogen-activated protein kinase phosphatase (MKP)-1 and MKP-2. The pressure overload-induced production of angiotensin II was inhibited by RA via upregulation of expression of angiotensin-converting enzyme (ACE)2 and through inhibition of the expression of cardiac and renal renin, angiotensinogen, ACE, and angiotensin type 1 receptor. Similar results were observed in cultured neonatal cardiomyocytes in response to static stretch. These results demonstrate that RA has a significant inhibitory effect on pressure overload-induced cardiac remodeling, through inhibition of the expression of renin-angiotensin system components.


Asunto(s)
Aorta Torácica/fisiología , Corazón/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Tretinoina/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Factor Natriurético Atrial/farmacología , Presión Sanguínea/efectos de los fármacos , Western Blotting , Células Cultivadas , Fibrosis/prevención & control , Frecuencia Cardíaca/efectos de los fármacos , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/prevención & control , Ligadura , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Péptido Natriurético Encefálico/farmacología , Estimulación Física , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/uso terapéutico , Ultrasonografía
19.
Cardiovasc Hematol Agents Med Chem ; 5(2): 109-32, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17430135

RESUMEN

Integrins are the principle mediators of molecular dialog between a cell and its extracellular matrix environment. The unique combinations of integrin subunits determine which extracellular matrix molecules are recognized by a cell. Recent studies have demonstrated that remodeling in heart and vasculature is linked to alterations in extracellular matrix and integrin expression. The roles of integrins in controlling cellular behavior have made these molecules highly attractive drug targets. New insights into mechanisms whereby the extracellular matrix takes part in the control of smooth muscle cell proliferation and cardiac growth suggest a number of putative targets for future therapies that can be applied to increase plaque stability, prevent the clinical consequences of atherosclerosis and improve outcomes after interventional procedures such as cardiac transplantation. Therapeutic candidates include antibodies, cyclic peptides, peptidomimetics and small molecules. The integrin inhibitors Integrilin and ReoPro have been approved as blood thinners in cardiovascular disease, and newer agents are undergoing testing. Although integrin function is important in the cardiovascular system, there are wide gaps in knowledge. In this review, we discuss the primary mechanisms of action and signaling of integrins in the cardiac and vascular system in normal and pathological states, as well as therapeutic strategies for targeting these molecules in the cardiovascular system.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Integrinas , Fármacos Cardiovasculares/química , Fenómenos Fisiológicos Cardiovasculares , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/crecimiento & desarrollo , Matriz Extracelular/efectos de los fármacos , Humanos , Integrinas/antagonistas & inhibidores , Integrinas/efectos de los fármacos , Integrinas/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA