Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Curr Opin Nephrol Hypertens ; 33(2): 174-180, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38164877

RESUMEN

PURPOSE OF REVIEW: Interleukin 15 (IL-15) is a member of the IL-2 family of common gamma chain receptor cytokines with well described anti-inflammatory, pro-survival and pro-proliferative signaling properties. The cytoprotective role of IL-15 in the kidney is now coming into focus with recent reports of its beneficial actions in various forms of kidney disease. This review will summarize what is currently known about IL-15 signaling in the kidney and highlight recent evidence of its beneficial effects on kidney physiology. RECENT FINDINGS: IL-15 and its heterotrimeric receptor are expressed throughout the kidney. Like all IL-2 family cytokines, IL-15 can activate signaling through the Janus Kinase (JAK)/Signal transducer of activated T-cells (STAT), phosphoinositol-3 kinase (PI-3K)/AKT and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways and recent evidence suggests that STAT5B is an essential transcriptional mediator of prosurvival signaling in glomerular visceral epithelial cells (i.e. podocytes). IL-15 has also been shown to suppress pro-apoptotic signaling in models of acute kidney injury and pro-fibrotic signaling in models of chronic kidney disease. SUMMARY: The cytoprotective properties of IL-15 suggest that it may have potential as a nonimmunosuppresive therapeutic for kidney disease. A novel class of IL-15 immunotherapies has emerged for the treatment cancer and some have demonstrated efficacy in clinical trials. These well tolerated IL-15 agonists could possibly be repurposed for the treatment of kidney disease and warrant further exploration.


Asunto(s)
Interleucina-15 , Enfermedades Renales , Humanos , Interleucina-2 , Transducción de Señal , Citocinas , Enfermedades Renales/tratamiento farmacológico
2.
BMC Nephrol ; 24(1): 30, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759756

RESUMEN

BACKGROUND: Tobacco exposure has been recognized as a risk factor for cardiovascular disease (CVD) and progression of kidney disease. Patients with proteinuric glomerulopathies are at increased risk for cardiovascular morbidity and mortality. Multiple studies have linked tobacco exposure to CVD and chronic kidney disease, but the relationships between smoking and proteinuric glomerulopathies in adults and children have not been previously explored. METHODS: Data from the Nephrotic Syndrome Study Network (NEPTUNE), a multi-center prospective observational study of participants with proteinuric glomerulopathies, was analyzed. 371 adults and 192 children enrolled in NEPTUNE were included in the analysis. Self-reported tobacco exposure was classified as non-smoker, active smoker, former smoker, or exclusive passive smoker. Baseline serum cotinine levels were measured in a sub-cohort of 178 participants. RESULTS: The prevalence of active smokers, former smokers and exclusive passive smoking among adults at baseline was 14.6%, 29.1% and 4.9%, respectively. Passive smoke exposure was 16.7% among children. Active smoking (reference non-smoking) was significantly associated with greater total cholesterol among adults (ß 17.91 95% CI 0.06, 35.76, p = 0.049) while passive smoking (reference non-smoking) was significantly associated with greater proteinuria over time among children (ß 1.23 95% CI 0.13, 2.33, p = 0.03). Higher cotinine levels were associated with higher baseline eGFR (r = 0.17, p = 0.03). CONCLUSION: Tobacco exposure is associated with greater risk for CVD and worse kidney disease outcomes in adults and children with proteinuric glomerulopathies. Preventive strategies to reduce tobacco exposure may help protect against future cardiovascular and kidney morbidity and mortality in patients with proteinuric glomerulopathies.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Renales , Contaminación por Humo de Tabaco , Humanos , Adulto , Niño , Estudios de Cohortes , Cotinina , Nicotiana , Contaminación por Humo de Tabaco/efectos adversos , Neptuno , Enfermedades Renales/inducido químicamente
3.
JCI Insight ; 7(11)2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35472001

RESUMEN

COVID-19 infection causes collapse of glomerular capillaries and loss of podocytes, culminating in a severe kidney disease called COVID-19-associated nephropathy (COVAN). The underlying mechanism of COVAN is unknown. We hypothesized that cytokines induced by COVID-19 trigger expression of pathogenic APOL1 via JAK/STAT signaling, resulting in podocyte loss and COVAN phenotype. Here, based on 9 biopsy-proven COVAN cases, we demonstrated for the first time, to the best of our knowledge, that APOL1 protein was abundantly expressed in podocytes and glomerular endothelial cells (GECs) of COVAN kidneys but not in controls. Moreover, a majority of patients with COVAN carried 2 APOL1 risk alleles. We show that recombinant cytokines induced by SARS-CoV-2 acted synergistically to drive APOL1 expression through the JAK/STAT pathway in primary human podocytes, GECs, and kidney micro-organoids derived from a carrier of 2 APOL1 risk alleles, but expression was blocked by a JAK1/2 inhibitor, baricitinib. We demonstrate that cytokine-induced JAK/STAT/APOL1 signaling reduced the viability of kidney organoid podocytes but was rescued by baricitinib. Together, our results support the conclusion that COVID-19-induced cytokines are sufficient to drive COVAN-associated podocytopathy via JAK/STAT/APOL1 signaling and that JAK inhibitors could block this pathogenic process. These findings suggest JAK inhibitors may have therapeutic benefits for managing cytokine-induced, APOL1-mediated podocytopathy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Citocinas , Inhibidores de las Cinasas Janus , Enfermedades Renales , Apolipoproteína L1/genética , Azetidinas/farmacología , COVID-19/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/virología , Organoides/metabolismo , Purinas/farmacología , Pirazoles/farmacología , SARS-CoV-2/aislamiento & purificación , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
4.
Am J Physiol Renal Physiol ; 322(2): F164-F174, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894725

RESUMEN

Interleukin (IL)-1 receptor type 1 (IL-1R1) activation triggers a proinflammatory signaling cascade that can exacerbate kidney injury. However, the functions of podocyte IL-1R1 in glomerular disease remain unclear. To study the role of IL-1R1 signaling in podocytes, we selectively ablated podocyte IL-1R1 in mice (PKO mice). We then subjected PKO mice and wild-type controls to two glomerular injury models: nephrotoxic serum (NTS)- and adriamycin-induced nephropathy. Surprisingly, we found that IL-1R1 activation in podocytes limited albuminuria and podocyte injury during NTS- and adriamycin-induced nephropathy. Moreover, deletion of IL-1R1 in podocytes drove podocyte apoptosis and glomerular injury through diminishing Akt activation. Activation of Akt signaling abrogated the differences in albuminuria and podocyte injury between wild-type and PKO mice during NTS. Thus, IL-1R1 signaling in podocytes limits susceptibility to glomerular injury via an Akt-dependent signaling pathway. These data identify an unexpected protective role for IL-1R1 signaling in podocytes in the pathogenesis of glomerular disease.NEW & NOTEWORTHY The present study establishes that activation of the receptor for interleukin-1 limits susceptibility to damage to the kidney glomerulus in preclinical mouse models by stimulating Akt signaling cascades inside the podocyte.


Asunto(s)
Glomerulonefritis/metabolismo , Podocitos/metabolismo , Proteinuria/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Doxorrubicina , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Glomerulonefritis/prevención & control , Humanos , Interleucina-1beta/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones de la Cepa 129 , Ratones Noqueados , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/inducido químicamente , Proteinuria/patología , Proteinuria/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Tipo I de Interleucina-1/agonistas , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal
5.
JCI Insight ; 6(15)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34369383

RESUMEN

The transcription factor Twist1 regulates several processes that could impact kidney disease progression, including epithelial cell differentiation and inflammatory cytokine induction. Podocytes are specialized epithelia that exhibit features of immune cells and could therefore mediate unique effects of Twist1 on glomerular disease. To study Twist1 functions in podocytes during proteinuric kidney disease, we employed a conditional mutant mouse in which Twist1 was selectively ablated in podocytes (Twist1-PKO). Deletion of Twist1 in podocytes augmented proteinuria, podocyte injury, and foot process effacement in glomerular injury models. Twist1 in podocytes constrained renal accumulation of monocytes/macrophages and glomerular expression of CCL2 and the macrophage cytokine TNF-α after injury. Deletion of TNF-α selectively from podocytes had no impact on the progression of proteinuric nephropathy. By contrast, the inhibition of CCL2 abrogated the exaggeration in proteinuria and podocyte injury accruing from podocyte Twist1 deletion. Collectively, Twist1 in podocytes mitigated urine albumin excretion and podocyte injury in proteinuric kidney diseases by limiting CCL2 induction that drove monocyte/macrophage infiltration into injured glomeruli. Myeloid cells, rather than podocytes, further promoted podocyte injury and glomerular disease by secreting TNF-α. These data highlight the capacity of Twist1 in the podocyte to mitigate glomerular injury by curtailing the local myeloid immune response.


Asunto(s)
Quimiocina CCL2/metabolismo , Células Mieloides/inmunología , Podocitos/metabolismo , Insuficiencia Renal Crónica , Factor de Necrosis Tumoral alfa/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Diferenciación Celular , Silenciador del Gen , Inmunidad/inmunología , Glomérulos Renales/inmunología , Glomérulos Renales/lesiones , Glomérulos Renales/metabolismo , Macrófagos , Ratones , Proteinuria/metabolismo , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
6.
Front Pediatr ; 6: 307, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30406062

RESUMEN

Background: Steroid-resistant nephrotic syndrome (SRNS) is a leading cause of end-stage kidney disease in children and young adults. Despite advances in genomic science that have led to the discovery of >50 monogenic causes of SRNS, there are no clear guidelines for genetic testing in clinical practice. Methods: Using high throughput sequencing, we evaluated 492 individuals from 181 families for mutations in 40 known SRNS genes. Causative mutations were defined as missense, truncating, and obligatory splice site variants with a minor allele frequency <1% in controls. Non-synonymous variants were considered pathogenic if determined to be deleterious by at least two in silico models. We further evaluated for differences in age at disease onset, family history of SRNS or chronic kidney disease, race, sex, renal biopsy findings, and extra-renal manifestations in subgroups with and without disease causing variants. Results: We identified causative variants in 40 of 181 families (22.1%) with SRNS. Variants in INF2, COL4A3, and WT1 were the most common, accounting for over half of all causative variants. Causative variants were identified in 34 of 86 families (39.5%) with familial disease and 6 of 95 individuals (6.3%) with sporadic disease (χ2 p < 0.00001). Family history was the only significant clinical predictor of genetic SRNS. Conclusion: We identified causative mutations in almost 40% of all families with hereditary SRNS and 6% of individuals with sporadic disease, making family history the single most important clinical predictors of monogenic SRNS. We recommend genetic testing in all patients with SRNS and a positive family history, but only selective testing in those with sporadic disease.

7.
J Am Soc Nephrol ; 29(8): 2110-2122, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30002222

RESUMEN

BACKGROUND: We previously reported that mutations in the anillin (ANLN) gene cause familial forms of FSGS. ANLN is an F-actin binding protein that modulates podocyte cell motility and interacts with the phosphoinositide 3-kinase (PI3K) pathway through the slit diaphragm adaptor protein CD2-associated protein (CD2AP). However, it is unclear how the ANLN mutations cause the FSGS phenotype. We hypothesized that the R431C mutation exerts its pathogenic effects by uncoupling ANLN from CD2AP. METHODS: We conducted in vivo complementation assays in zebrafish to determine the effect of the previously identified missense ANLN variants, ANLNR431C and ANLNG618C during development. We also performed in vitro functional assays using human podocyte cell lines stably expressing wild-type ANLN (ANLNWT ) or ANLNR431C . RESULTS: Experiments in anln-deficient zebrafish embryos showed a loss-of-function effect for each ANLN variant. In human podocyte lines, expression of ANLNR431C increased cell migration, proliferation, and apoptosis. Biochemical characterization of ANLNR431C -expressing podocytes revealed hyperactivation of the PI3K/AKT/mTOR/p70S6K/Rac1 signaling axis and activation of mTOR-driven endoplasmic reticulum stress in ANLNR431C -expressing podocytes. Inhibition of mTOR, GSK-3ß, Rac1, or calcineurin ameliorated the effects of ANLNR431C . Additionally, inhibition of the calcineurin/NFAT pathway reduced the expression of endogenous ANLN and mTOR. CONCLUSIONS: The ANLNR431C mutation causes multiple derangements in podocyte function through hyperactivation of PI3K/AKT/mTOR/p70S6K/Rac1 signaling. Our findings suggest that the benefits of calcineurin inhibition in FSGS may be due, in part, to the suppression of ANLN and mTOR. Moreover, these studies illustrate that rational therapeutic targets for familial FSGS can be identified through biochemical characterization of dysregulated podocyte phenotypes.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/genética , Movimiento Celular/genética , Células Cultivadas , Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Humanos , Mutación Missense , Podocitos/metabolismo , Sensibilidad y Especificidad , Transducción de Señal , Pez Cebra , Proteína de Unión al GTP rac1/genética
8.
Kidney Int ; 92(2): 283-285, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28709595

RESUMEN

Selective modulation of Rho GTPase activity in podocytes recapitulates characteristic features of human nephrosis. Using a mouse model, Robins et al. found that high levels of Rac1 activation in podocytes caused podocyte detachment and glomerulosclerosis. Podocyte Rac1 activity was enhanced in biopsy specimens from patients with nephrosis, and serum from this patient population activated Rac1 in cultured podocytes. These data provide a causal link between podocyte Rac1 activation and human nephrotic diseases.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Nefrosis , Síndrome Nefrótico , Podocitos , Humanos , Proteína de Unión al GTP rac1
9.
J Am Soc Nephrol ; 26(4): 831-43, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25145932

RESUMEN

FSGS is a clinical disorder characterized by focal scarring of the glomerular capillary tuft, podocyte injury, and nephrotic syndrome. Although idiopathic forms of FSGS predominate, recent insights into the molecular and genetic causes of FSGS have enhanced our understanding of disease pathogenesis. Here, we report a novel missense mutation of the transcriptional regulator Wilms' Tumor 1 (WT1) as the cause of nonsyndromic, autosomal dominant FSGS in two Northern European kindreds from the United States. We performed sequential genome-wide linkage analysis and whole-exome sequencing to evaluate participants from family DUK6524. Subsequently, whole-exome sequencing and direct sequencing were performed on proband DNA from family DUK6975. We identified multiple suggestive loci on chromosomes 6, 11, and 13 in family DUK6524 and identified a segregating missense mutation (R458Q) in WT1 isoform D as the cause of FSGS in this family. The identical mutation was found in family DUK6975. The R458Q mutation was not found in 1600 control chromosomes and was predicted as damaging by in silico simulation. We depleted wt1a in zebrafish embryos and observed glomerular injury and filtration defects, both of which were rescued with wild-type but not mutant human WT1D mRNA. Finally, we explored the subcellular mechanism of the mutation in vitro. WT1(R458Q) overexpression significantly downregulated nephrin and synaptopodin expression, promoted apoptosis in HEK293 cells and impaired focal contact formation in podocytes. Taken together, these data suggest that the WT1(R458Q) mutation alters the regulation of podocyte homeostasis and causes nonsyndromic FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/metabolismo , Proteínas WT1/genética , Adolescente , Adulto , Animales , Movimiento Celular , Supervivencia Celular , Exoma , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Células HEK293 , Humanos , Masculino , Mutación Missense , Nefrosis/etiología , Nefrosis/metabolismo , Podocitos/fisiología , Análisis de Secuencia de ADN , Proteínas WT1/deficiencia , Adulto Joven , Pez Cebra , Proteínas de Pez Cebra/deficiencia
10.
J Am Soc Nephrol ; 25(9): 1991-2002, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24676636

RESUMEN

FSGS is characterized by segmental scarring of the glomerulus and is a leading cause of kidney failure. Identification of genes causing FSGS has improved our understanding of disease mechanisms and points to defects in the glomerular epithelial cell, the podocyte, as a major factor in disease pathogenesis. Using a combination of genome-wide linkage studies and whole-exome sequencing in a kindred with familial FSGS, we identified a missense mutation R431C in anillin (ANLN), an F-actin binding cell cycle gene, as a cause of FSGS. We screened 250 additional families with FSGS and found another variant, G618C, that segregates with disease in a second family with FSGS. We demonstrate upregulation of anillin in podocytes in kidney biopsy specimens from individuals with FSGS and kidney samples from a murine model of HIV-1-associated nephropathy. Overexpression of R431C mutant ANLN in immortalized human podocytes results in enhanced podocyte motility. The mutant anillin displays reduced binding to the slit diaphragm-associated scaffold protein CD2AP. Knockdown of the ANLN gene in zebrafish morphants caused a loss of glomerular filtration barrier integrity, podocyte foot process effacement, and an edematous phenotype. Collectively, these findings suggest that anillin is important in maintaining the integrity of the podocyte actin cytoskeleton.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Movimiento Celular/genética , Secuencia Conservada , Proteínas Contráctiles/genética , Proteínas del Citoesqueleto/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Barrera de Filtración Glomerular/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Linaje , Podocitos/metabolismo , Homología de Secuencia de Aminoácido , Regulación hacia Arriba , Pez Cebra , Proteínas de Pez Cebra/genética
11.
Mol Ther ; 19(11): 1961-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21730973

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) is caused by the deficiency of glucose-6-phosphatase (G6Pase). Long-term complications of GSD-Ia include life-threatening hypoglycemia and proteinuria progressing to renal failure. A double-stranded (ds) adeno-associated virus serotype 2 (AAV2) vector encoding human G6Pase was pseudotyped with four serotypes, AAV2, AAV7, AAV8, and AAV9, and we evaluated efficacy in 12-day-old G6pase (-/-) mice. Hypoglycemia during fasting (plasma glucose <100 mg/dl) was prevented for >6 months by the dsAAV2/7, dsAAV2/8, and dsAAV2/9 vectors. Prolonged fasting for 8 hours revealed normalization of blood glucose following dsAAV2/9 vector administration at the higher dose. The glycogen content of kidney was reduced by >65% with both the dsAAV2/7 and dsAAV2/9 vectors, and renal glycogen content was stably reduced between 7 and 12 months of age for the dsAAV2/9 vector-treated mice. Every vector-treated group had significantly reduced glycogen content in the liver, in comparison with untreated G6pase (-/-) mice. G6Pase was expressed in many renal epithelial cells of with the dsAAV2/9 vector for up to 12 months. Albuminuria and renal fibrosis were reduced by the dsAAV2/9 vector. Hepatorenal correction in G6pase (-/-) mice demonstrates the potential of AAV vectors for the correction of inherited diseases of metabolism.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/mortalidad , Humanos , Hipoglucemia/genética , Hipoglucemia/terapia , Estimación de Kaplan-Meier , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados
12.
J Mol Cell Cardiol ; 50(2): 346-56, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20837021

RESUMEN

Since massive irreversible loss of cardiac myocytes occurs following myocardial injury, injection of human mesenchymal stem cells (hMSCs) has emerged as a promising therapeutic intervention. Despite the growing enthusiasm for this approach, the understanding of how hMSCs evoke cardiac improvement is ever more controversial. The present study critically tests hypothesis that hMSCs provide specific benefit directly to damaged ventricular myocytes. Cultures of neonatal mouse ventricular cardiac myocytes (nMCM) were subjected to two distinct acute stress protocols; incubations with either endotoxin, lipopolysaccharide (LPS) or toxic cytokine, IL-1ß. Myocyte injury was assessed in intracellular Ca(2+) signaling assays in fluo-3-loaded nMCMs that were imaged with high temporal resolution by fluorescent microscopy. Following LPS or IL-1ß treatment there was profound myocyte injury, manifest by chaotic [Ca(2+)](i) handling, quantified as a 3- to 5-fold increase in spontaneous [Ca(2+)](i) transients. Antibody neutralization experiments reveal such damage is mediated in part by interleukin-18 and not by tumor necrosis factor-α (TNF-α). Importantly, normal [Ca(2+)](i) signaling was preserved when cardiomyocytes were co-cultured with hMSCs. Since normal [Ca(2+)](i) handling was maintained in transwell cultures, where nMCMs and hMSCs were separated by a permeable membrane, a protective paracrine signaling cascade is operable. hMSCs provoke a genetic reprogramming of cardiomyocytes. LPS provokes release of TNFα from nMCMs which is blocked by hMSCs grown in co- or transwell cultures. Consistent with cytokine release, flow cytometry analyses reveal that hMSCs also block the LPS- and IL-1ß-dependent activation of cardiac transcription factor, NF-κB. Importantly, hMSC-conditioned medium restores normal Ca(2+) signaling in LPS- and IL-1ß-damaged nMCMs. These results reveal new evidence that hMSCs elicit protective and reparative effects on cardiac tissue through molecular reprogramming of the cardiac myocytes themselves. Thus these studies provide novel new insight into the cellular and molecular mechanisms that underlie the therapeutic benefit of hMSCs in the setting of heart failure. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Asunto(s)
Reprogramación Celular/fisiología , Ventrículos Cardíacos/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Adyuvantes Inmunológicos/farmacología , Animales , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Interleucina-18/antagonistas & inhibidores , Interleucina-1beta/farmacología , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , FN-kappa B/metabolismo , Comunicación Paracrina , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
13.
Am J Obstet Gynecol ; 195(1): 275-87, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16681987

RESUMEN

OBJECTIVE: Coupled to hundreds of receptors, G-proteins modulate signal transduction pathways and are important hormonal targets. The first objective was to determine the effect of pregnancy and estradiol on myometrial guanosine triphosphatase activity. The second objective was to begin dissecting the molecular mechanism(s) underlying alterations in guanosine triphosphatase activity. STUDY DESIGN: Myometrial tissue was obtained from pregnant, nonpregnant, and ovariectomized untreated and estradiol-treated guinea pigs. Myometrial membranes were prepared by homogenization and differential centrifugation. Basal high-affinity specific guanosine triphosphatase activity was quantitated by enzymatic assay and expressed in rhomol 32Pi per milligram protein per minute. Guanosine triphosphatase activity was stimulated using oxytocin, isoproterenol, and prostaglandin F2alpha. Specific G-protein subunits were quantitated using Western blots. G-protein associated gene expression was semiquantitated using HGU133A gene array chips from Affymetrix. RESULTS: Basal myometrial guanosine triphosphatase activity was increased in pregnant compared with nonpregnant animals. Estradiol increased basal myometrial guanosine triphosphatase activity, compared with untreated controls. The effect of estradiol on stimulated activity was agonist dependent. Both Galphas and Galphai isoform 1 protein levels were increased in myometrium from late pregnant compared with nonpregnant animals. By late gestation, the messenger ribonucleic acid levels of those genes were unaltered, compared with the nonpregnant animal. In general, the impact of pregnancy on G-protein family member gene messenger ribonucleic acid expression was modest. Only the small guanosine triphosphatase Rap1b demonstrated altered expression more than 2-fold during either myometrial quiescence (midpregnancy) or activation (term pregnancy) (up 3-fold during quiescence). Genomic network analyses revealed that the expression of another small guanosine triphosphatase, Rab7, was exclusively up-regulated (80%) during quiescence. During late pregnancy, network analysis showed that only G-protein beta was exclusively altered (up-regulated). Estradiol mimicked the pregnancy effect on both transcription and translation of G-protein family members for some but not all potentially relevant genes. CONCLUSION: The increase in functional myometrial guanosine triphosphatase activity during pregnancy may reflect increased synthesis of 1 or more small guanosine triphosphatase.


Asunto(s)
Estradiol/fisiología , Proteínas de Unión al GTP/metabolismo , Miometrio/metabolismo , Transducción de Señal/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Western Blotting , Regulación hacia Abajo/fisiología , Electroforesis en Gel de Poliacrilamida , Femenino , Perfilación de la Expresión Génica , Guanosina Trifosfato/metabolismo , Cobayas , Isoproterenol/farmacología , Miometrio/enzimología , Embarazo , Biosíntesis de Proteínas/fisiología , Regulación hacia Arriba/fisiología , Contracción Uterina/metabolismo
14.
J Exp Med ; 203(5): 1235-47, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16636132

RESUMEN

Emerging evidence suggests that both human stem cells and mature stromal cells can play an important role in the development and growth of human malignancies. In contrast to these tumor-promoting properties, we observed that in an in vivo model of Kaposi's sarcoma (KS), intravenously (i.v.) injected human mesenchymal stem cells (MSCs) home to sites of tumorigenesis and potently inhibit tumor growth. We further show that human MSCs can inhibit the in vitro activation of the Akt protein kinase within some but not all tumor and primary cell lines. The inhibition of Akt activity requires the MSCs to make direct cell-cell contact and can be inhibited by a neutralizing antibody against E-cadherin. We further demonstrate that in vivo, Akt activation within KS cells is potently down-regulated in areas adjacent to MSC infiltration. Finally, the in vivo tumor-suppressive effects of MSCs correlates with their ability to inhibit target cell Akt activity, and KS tumors engineered to express a constitutively activated Akt construct are no longer sensitive to i.v. MSC administration. These results suggest that in contrast to other stem cells or normal stromal cells, MSCs possess intrinsic antineoplastic properties and that this stem cell population might be of particular utility for treating those human malignancies characterized by dysregulated Akt.


Asunto(s)
Efecto Injerto vs Tumor/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Sarcoma de Kaposi/inmunología , Animales , Modelos Animales de Enfermedad , Activación Enzimática/inmunología , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Proteína Oncogénica v-akt/inmunología , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/terapia , Células del Estroma/inmunología , Células del Estroma/trasplante , Trasplante Heterólogo , Células Tumorales Cultivadas
15.
Am J Physiol Heart Circ Physiol ; 289(5): H2103-11, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15980040

RESUMEN

TNF-alpha is recognized as a significant contributor to myocardial dysfunction. Although several studies suggest that members of the NF-kappaB family of transcription factors are essential regulators of myocardial TNF-alpha gene expression, recent developments in our understanding of the modulation of NF-kappaB activity through posttranslational modification of NF-kappaB subunits suggest that the present view of NF-kappaB-dependent cytokine expression in heart is incomplete. Therefore, the goal of the present study was to examine the role of p65 subunit phosphorylation in the regulation of TNF-alpha production in cultured neonatal ventricular myocytes. Bacterial LPS-induced TNF-alpha production is accompanied by a 12-fold increase in phosphorylation of p65 at Ser536, a modification associated with enhancement of p65 transactivation potential. Pharmacological inhibition of IKK-beta reduced LPS-induced TNF-alpha production 38-fold, TNF-alpha mRNA levels 6-fold, and IkappaB-alpha phosphorylation 5-fold and degraded IkappaB-alpha 2-fold and p65 phosphorylation 6-fold. Overexpression of dominant-negative p65 reduced TNF-alpha production 3.5-fold, whereas overexpression of dominant-negative IKK-beta reduced LPS-induced TNF-alpha production 2-fold and p65 phosphorylation 2-fold. Overexpression of dominant-negative IKK-alpha had no effect on p65 phosphorylation or TNF-alpha production, revealing that IKK-beta, not IKK-alpha, plays a central role in regulation of p65 phosphorylation at Ser536 and TNF-alpha production in heart. Finally, we demonstrated, using a chromatin immunoprecipitation assay, that LPS stimulates recruitment of Ser536-phosphorylated p65 to the TNF-alpha gene promoter in cardiac myocytes. Taken together, these data provide compelling evidence for the role of NF-kappaB signaling in TNF-alpha gene expression in heart and highlight the importance of this proinflammatory gene-regulatory pathway as a potential therapeutic target in the management of cytokine-induced myocardial dysfunction.


Asunto(s)
Lipopolisacáridos/farmacología , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Animales Recién Nacidos , Western Blotting , Células Cultivadas , Cromatina/metabolismo , Citocinas/biosíntesis , Inmunoprecipitación , Ratones , Mutagénesis Sitio-Dirigida/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Fosforilación , Transducción de Señal/efectos de los fármacos , Quinasa de Factor Nuclear kappa B
16.
Am J Physiol Heart Circ Physiol ; 282(3): H872-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11834481

RESUMEN

Although tumor necrosis factor (TNF)-alpha is implicated in numerous cardiac pathologies, the intracellular events leading to its production by heart cells are largely unknown. The goal of the present study was to identify the role of the transcription factor nuclear factor (NF)-kappaB in this process. Among the many inducers of TNF-alpha expression in myeloid cells, only lipopolysaccharide (LPS) led to its induction in cultured neonatal myocytes. LPS also activated the NF-kappaB pathway, as evidenced by the degradation of the inhibitory protein IkappaB and the appearance of NF-kappaB-binding complexes in nuclear extracts. Furthermore, inhibitors of NF-kappaB activation, such as lactacystin, MG132, and pyrrolidine dithiocarbamate, were found to completely block the production of TNF-alpha in response to LPS stimulation, indicating a requirement of NF-kappaB for TNF-alpha expression. However, interleukin-1beta and phorbol 12-myristate 13-acetate also activated NF-kappaB but did not evoke TNF-alpha expression, revealing that this factor is not sufficient for cytokine production. Detailed examination of the NF-kappaB cascade revealed that cardiac cells displayed a unique pattern of IkappaB degradation in response to LPS, with IkappaBbeta but not IkappaBalpha being degraded upon stimulation. Additionally, two specific p65-containing DNA-binding complexes were observed in the nuclear extracts of neonatal cardiomyocytes: an inducible complex that is necessary for TNF-alpha expression and a constitutive species. Taken together, these results reveal that NF-kappaB is not only involved in cytokine production but also may be linked to other pathways that subserve a constitutive, protective mechanism for the heart cell.


Asunto(s)
Acetilcisteína/análogos & derivados , Endotoxinas/toxicidad , Corazón/fisiología , Lipopolisacáridos/toxicidad , FN-kappa B/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Factor de Necrosis Tumoral alfa/genética , Acetilcisteína/farmacología , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Western Blotting , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Corazón/efectos de los fármacos , Ratones , Complejos Multienzimáticos/metabolismo , Miocardio/citología , Miocardio/metabolismo , Complejo de la Endopetidasa Proteasomal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA