Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Aesthet Surg J ; 34(2): 227-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24353247

RESUMEN

BACKGROUND: Light and electron microscopy have not identified a distinct anatomical structure associated with either skin wrinkles or creases, and a histological difference between wrinkled and adjacent skin has not been identified. OBJECTIVES: The authors investigate whether facial wrinkles are related to underlying lymphatic vessels and perilymphatic fat. METHODS: Lymphatic vessels with a specialized tube of perilymphatic fat were identified beneath palmar creases. Sections of skin, adipose tissue, and muscle were harvested from each of 13 cadavers. Three sites were investigated: the transverse forehead crease, lateral orbicularis oculi wrinkle (crow's feet), and the nasojugal crease. The tissue was paraffin embedded and processed. Two-step indirect immunohistochemistry was performed, and images were examined using laser confocal microscopy. Measurements were taken with software. RESULTS: Every wrinkle examined was found above and within ±1 mm of a major lymphatic vessel and its surrounding tube of adipose tissue. The results satisfied our null hypothesis and were statistically significant. Lymphatic vessels were identified by positive immunofluorescence as well as histological criteria. These findings have been further validated by fluorochrome tracer studies. CONCLUSIONS: An anatomical basis for wrinkles was identified among the specimens studied. Lymphatic vessels, along with the surrounding distinct perilymphatic fat, traveled directly beneath wrinkles and creases. Lymphatic dysregulation leads to inflammation, scarring, and fibrosis, but inadvertent injection of these vessels can be avoided with anatomical knowledge.


Asunto(s)
Tejido Adiposo/anatomía & histología , Cara/anatomía & histología , Vasos Linfáticos/anatomía & histología , Músculo Esquelético/anatomía & histología , Envejecimiento de la Piel/fisiología , Piel/anatomía & histología , Tejido Adiposo/cirugía , Anciano , Anciano de 80 o más Años , Cadáver , Disección , Cara/cirugía , Femenino , Colorantes Fluorescentes , Mano/anatomía & histología , Mano/cirugía , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Persona de Mediana Edad , Músculo Esquelético/cirugía
2.
J Biol Chem ; 286(10): 8655-8665, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21209102

RESUMEN

Fibrosis is a pathological process characterized by infiltration and proliferation of mesenchymal cells in interstitial space. A substantial portion of these cells is derived from residing non-epithelial and/or epithelial cells that have acquired the ability to migrate and proliferate. The mesenchymal transition is also observed in cancer cells to confer the ability to metastasize. Here, we show that renal fibrosis induced by unilateral ureteral obstruction and metastasis of human cancer xenografts are suppressed by administration of secreted Klotho protein to mice. Klotho is a single-pass transmembrane protein expressed in renal tubular epithelial cells. The extracellular domain of Klotho is secreted by ectodomain shedding. Secreted Klotho protein directly binds to the type-II TGF-ß receptor and inhibits TGF-ß1 binding to cell surface receptors, thereby inhibiting TGF-ß1 signaling. Klotho suppresses TGF-ß1-induced epithelial-to-mesenchymal transition (EMT) responses in cultured cells, including decreased epithelial marker expression, increased mesenchymal marker expression, and/or increased cell migration. In addition to TGF-ß1 signaling, secreted Klotho has been shown to inhibit Wnt and IGF-1 signaling that can promote EMT. These results have raised the possibility that secreted Klotho may function as an endogenous anti-EMT factor by inhibiting multiple growth factor signaling pathways simultaneously.


Asunto(s)
Glucuronidasa/metabolismo , Neoplasias Renales/metabolismo , Riñón/metabolismo , Neoplasias Experimentales/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Regulación Neoplásica de la Expresión Génica/genética , Glucuronidasa/genética , Células HEK293 , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , Proteínas Klotho , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Trasplante Heterólogo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
3.
Am J Kidney Dis ; 58(1): 127-34, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21496980

RESUMEN

The klotho gene (KL) was identified first as a putative aging-suppressor gene that extended life span when overexpressed and accelerated aging-like phenotypes when disrupted in mice. It encodes a single-pass transmembrane protein and is expressed predominantly in kidney, where it functions as an obligate coreceptor for fibroblast growth factor 23 (FGF-23). FGF-23 is a bone-derived hormone that suppresses phosphate reabsorption and 1,25 dihydroxyvitamin D(3) (vitamin D) synthesis in the kidney. Klotho also is expressed in the parathyroid gland, where FGF-23 decreases parathyroid hormone expression and secretion, further suppressing vitamin D synthesis in kidney. Thus, FGF-23 functions as a phosphaturic hormone and a counter-regulatory hormone for vitamin D, thereby inducing negative phosphate balance. Mice lacking either FGF-23 or Klotho show hyperphosphatemia in addition to developing multiple aging-like phenotypes, which can be rescued by resolving phosphate retention. These findings have unveiled an unexpected link between aging and phosphate. In patients with chronic kidney disease (CKD), phosphate retention is seen universally and has been associated with increased mortality risk. Patients with CKD have high serum FGF-23 levels with decreased klotho expression in the kidney and parathyroid, rendering FGF-23 and Klotho as potential biomarkers and therapeutic targets for CKD. The Klotho protein not only serves as a coreceptor for FGF-23, but also functions as a humoral factor. Klotho's extracellular domain is released into blood and urine by ectodomain shedding and exerts various functions independently of FGF-23, including regulation of multiple ion channels and transporters. Decreased urinary Klotho protein level has been identified as one of the earliest biomarkers of CKD progression. This review focuses on the current understanding of Klotho protein function, with emphasis on its potential involvement in the pathophysiologic process of CKD.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/fisiología , Fosfatos/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Anciano , Envejecimiento/fisiología , Animales , Biomarcadores/orina , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/genética , Glucuronidasa/orina , Humanos , Proteínas Klotho , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo
4.
Dev Biol ; 331(2): 292-9, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19447101

RESUMEN

In mammals, primordial follicles are generated early in life and remain dormant for prolonged intervals. Their growth resumes via a process known as primordial follicle activation. Recent genetic studies have demonstrated that phosphoinositide 3-kinase (PI3K) is the essential signaling pathway controlling this process throughout life, acting via Akt to regulate nucleocytoplasmic shuttling of Foxo3, which functions as a downstream molecular switch. The receptor tyrosine kinase Kit has been implicated by numerous studies as the critical upstream regulator of primordial follicle activation via PI3K/Akt. Here we present a genetic analysis of the contribution of Kit in regulating primordial follicle activation and early follicle growth, employing a knock-in mutation (Kit(Y719F)) that completely abrogates signaling via PI3K. Surprisingly, homozygous Kit(Y719F) female mice undergo primordial follicle activation and are fertile, demonstrating that Kit signaling via PI3K is dispensable for this process. However, other abnormalities were identified in Kit(Y719F) ovaries, including accelerated primordial follicle depletion and accumulation of morphologically abnormal primary/secondary follicles with persistent nuclear Foxo3 localization. These findings reveal specific roles of Kit in the maintenance of the primordial follicle reserve and in the primary to secondary follicle transition, but argue that Kit is dispensable in primordial follicle activation.


Asunto(s)
Folículo Ovárico/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Femenino , Técnicas de Sustitución del Gen , Ratones , Mutación , Folículo Ovárico/citología , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/fisiología , Proteínas Proto-Oncogénicas c-kit/genética
5.
Dev Biol ; 321(1): 197-204, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18601916

RESUMEN

In mammals, oocytes are packaged into compact structures-primordial follicles-which remain inert for prolonged intervals until individual follicles resume growth via a process known as primordial follicle activation. Here we show that the phosphoinositide 3-kinase (PI3K) signalling pathway controls primordial follicle activation through the forkhead transcription factor Foxo3. Within oocytes, Foxo3 is regulated by nucleocytoplasmic shuttling. Foxo3 is imported into the nucleus during primordial follicle assembly, and is exported upon activation. Oocyte-specific ablation of Pten resulted in PI3K-induced Akt activation, Foxo3 hyperphosphorylation, and Foxo3 nuclear export, thereby triggering primordial follicle activation, defining the steps by which the PI3K pathway and Foxo3 control this process. Inducible ablation of Pten and Foxo3 in adult oocytes using a new tool for genetic analysis of the germline, Vasa-Cre(ERT2), showed that this pathway functions throughout life. Thus, a principal physiologic role of the PI3K pathway is to control primordial follicle activation via Foxo3.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Oocitos/crecimiento & desarrollo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Proteína Forkhead Box O3 , Ratones , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal
6.
Genetics ; 177(1): 179-94, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17660561

RESUMEN

Female infertility syndromes are among the most prevalent chronic health disorders in women, but their genetic basis remains unknown because of uncertainty regarding the number and identity of ovarian factors controlling the assembly, preservation, and maturation of ovarian follicles. To systematically discover ovarian fertility genes en masse, we employed a mouse model (Foxo3) in which follicles are assembled normally but then undergo synchronous activation. We developed a microarray-based approach for the systematic discovery of tissue-specific genes and, by applying it to Foxo3 ovaries and other samples, defined a surprisingly large set of ovarian factors (n = 348, approximately 1% of the mouse genome). This set included the vast majority of known ovarian factors, 44% of which when mutated produce female sterility phenotypes, but most were novel. Comparative profiling of other tissues, including microdissected oocytes and somatic cells, revealed distinct gene classes and provided new insights into oogenesis and ovarian function, demonstrating the utility of our approach for tissue-specific gene discovery. This study will thus facilitate comprehensive analyses of follicle development, ovarian function, and female infertility.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Genes/fisiología , Genoma , Infertilidad Femenina/metabolismo , Folículo Ovárico/fisiología , Animales , Northern Blotting , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Hibridación in Situ , Infertilidad Femenina/patología , Rayos Láser , Meiosis , Ciclo Menstrual , Ratones , Ratones Noqueados , Microdisección , Análisis de Secuencia por Matrices de Oligonucleótidos , Oocitos/fisiología , Oocitos/ultraestructura , Oogénesis/fisiología , Folículo Ovárico/ultraestructura , Sondas ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Hum Reprod ; 23(1): 216-21, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17959613

RESUMEN

BACKGROUND: The forkhead transcription factor Foxo3 is a master regulator and potent suppressor of primordial follicle activation. Loss of Foxo3 function in the mouse leads to premature ovarian failure (POF) due to global follicle activation. METHODS AND RESULTS: Here, we show that the mouse Foxo3 locus is haploinsufficient, and that Foxo3-/+ females undergo early reproductive senescence consistent with an increased rate of primordial follicle utilization. Then, to determine if heterozygous or homozygous polymorphisms or mutations of the human orthologue FOXO3 contribute to POF or idiopathic primary amenorrhea (PA), we sequenced the exons and flanking splice sequences of the gene in a large number of women with idiopathic POF (n = 273) or PA (n = 29). A total of eight single-nucleotide polymorphisms (SNPs) were identified, revealing a substantial amount of genetic variation at this locus. Allelic frequencies in control samples excluded several of these variants as causal. For the remaining variants, site-directed mutagenesis was performed to assess their functional impact. However, these rare sequence variants were not associated with significant decreases in FOXO3 activity. CONCLUSIONS: Taken together, our findings suggest that, despite the potential for FOXO3 haploinsufficiency to cause ovarian failure, FOXO3 mutations or common SNPs are not a common cause of either POF or PA.


Asunto(s)
Amenorrea/genética , Factores de Transcripción Forkhead/genética , Variación Genética , Insuficiencia Ovárica Primaria/genética , Adulto , Animales , Secuencia de Bases , Exones , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Frecuencia de los Genes , Haplotipos , Heterocigoto , Humanos , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa/métodos , Polimorfismo de Nucleótido Simple
8.
Mol Biol Cell ; 16(3): 1543-54, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15647377

RESUMEN

Mitochondria are complex organelles with a highly dynamic distribution and internal organization. Here, we demonstrate that mitofilin, a previously identified mitochondrial protein of unknown function, controls mitochondrial cristae morphology. Mitofilin is enriched in the narrow space between the inner boundary and the outer membranes, where it forms a homotypic interaction and assembles into a large multimeric protein complex. Down-regulation of mitofilin in HeLa cells by using specific small interfering RNA lead to decreased cellular proliferation and increased apoptosis, suggesting abnormal mitochondrial function. Although gross mitochondrial fission and fusion seemed normal, ultrastructural studies revealed disorganized mitochondrial inner membrane. Inner membranes failed to form tubular or vesicular cristae and showed as closely packed stacks of membrane sheets that fused intermittently, resulting in a complex maze of membranous network. Electron microscopic tomography estimated a substantial increase in inner:outer membrane ratio, whereas no cristae junctions were detected. In addition, mitochondria subsequently exhibited increased reactive oxygen species production and membrane potential. Although metabolic flux increased due to mitofilin deficiency, mitochondrial oxidative phosphorylation was not increased accordingly. We propose that mitofilin is a critical organizer of the mitochondrial cristae morphology and thus indispensable for normal mitochondrial function.


Asunto(s)
Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Proteínas Musculares/química , Proteínas Musculares/fisiología , Animales , Apoptosis , Western Blotting , Proliferación Celular , Centrifugación por Gradiente de Densidad , Electroforesis en Gel de Poliacrilamida , Glicerol/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Hígado/metabolismo , Sustancias Macromoleculares/metabolismo , Ratones , Microscopía Electrónica , Proteínas Mitocondriales/fisiología , Plásmidos/metabolismo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Retroviridae/genética , Relación Estructura-Actividad , Transfección , Técnicas del Sistema de Dos Híbridos
9.
Neoplasia ; 16(3): 247-56, 256.e2, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24784001

RESUMEN

Despite evidence that kinesin family member 14 (KIF14) can serve as a prognostic biomarker in various solid tumors, how it contributes to tumorigenesis remains unclear. We observed that experimental decrease in KIF14 expression increases docetaxel chemosensitivity in estrogen receptor-negative/progesterone receptor-negative/human epidermal growth factor receptor 2-negative, "triple-negative" breast cancers (TNBC). To investigate the oncogenic role of KIF14, we used noncancerous human mammary epithelial cells and ectopically expressed KIF14 and found increased proliferative capacity, increased anchorage-independent grown in vitro, and increased resistance to docetaxel but not to doxorubicin, carboplatin, or gemcitabine. Seventeen benign breast biopsies of BRCA1 or BRCA2 mutation carriers showed increased KIF14 mRNA expression by fluorescence in situ hybridization compared to controls with no known mutations in BRCA1 or BRCA2, suggesting increased KIF14 expression as a biomarker of high-risk breast tissue. Evaluation of 34 cases of locally advanced TNBC showed that KIF14 expression significantly correlates with chemotherapy-resistant breast cancer. KIF14 knockdown also correlates with decreased AKT phosphorylation and activity. Live-cell imaging confirmed an insulin-induced temporal colocalization of KIF14 and AKT at the plasma membrane, suggesting a potential role of KIF14 in promoting activation of AKT. An experimental small-molecule inhibitor of KIF14 was then used to evaluate the potential anticancer benefits of downregulating KIF14 activity. Inhibition of KIF14 shows a chemosensitizing effect and correlates with decreasing activation of AKT. Together, these findings show an early and critical role for KIF14 in the tumorigenic potential of TNBC, and therapeutic targeting of KIF14 is feasible and effective for TNBC.


Asunto(s)
Resistencia a Antineoplásicos , Cinesinas/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Proteína BRCA1/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Heterocigoto , Humanos , Cinesinas/genética , Terapia Neoadyuvante , Proteínas Oncogénicas/genética , Fosforilación , Neoplasias de la Mama Triple Negativas/metabolismo
10.
Mol Endocrinol ; 24(10): 2050-64, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20667984

RESUMEN

Although members of the fibroblast growth factor (FGF) family and their receptors have well-established roles in embryogenesis, their contributions to adult physiology remain relatively unexplored. Here, we use real-time quantitative PCR to determine the mRNA expression patterns of all 22 FGFs, the seven principal FGF receptors (FGFRs), and the three members of the Klotho family of coreceptors in 39 different mouse tissues. Unsupervised hierarchical cluster analysis of the mRNA expression data reveals that most FGFs and FGFRs fall into two groups the expression of which is enriched in either the central nervous system or reproductive and gastrointestinal tissues. Interestingly, the FGFs that can act as endocrine hormones, including FGF15/19, FGF21, and FGF23, cluster in a third group that does not include any FGFRs, underscoring their roles in signaling between tissues. We further show that the most recently identified Klotho family member, Lactase-like, is highly and selectively expressed in brown adipose tissue and eye and can function as an additional coreceptor for FGF19. This FGF atlas provides an important resource for guiding future studies to elucidate the physiological functions of FGFs in adult animals.


Asunto(s)
Atlas como Asunto , Factores de Crecimiento de Fibroblastos/fisiología , Ratones/fisiología , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Animales , Análisis por Conglomerados , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Perfilación de la Expresión Génica , Humanos , Ratones/embriología , Ratones Endogámicos C57BL , Análisis por Micromatrices , ARN Mensajero/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Distribución Tisular
11.
Genesis ; 45(6): 413-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17551945

RESUMEN

Cell type-specific genetic modification using the Cre/loxP system is a powerful tool for genetic analysis of distinct cell lineages. Because of the exquisite specificity of Vasa expression (confined to the germ cell lineage in invertebrate and vertebrate species), we hypothesized that a Vasa promoter-driven transgenic Cre line would prove useful for the germ cell lineage-specific inactivation of genes. Here we describe a transgenic mouse line, Vasa-Cre, where Cre is efficiently and specifically expressed in germ cells. Northern analysis showed that transgene expression was confined to the gonads. Cre-mediated recombination with the Rosa26-lacZ reporter was observed beginning at approximately e15, and was >95% efficient in male and female germ cells by birth. Although there was a potent maternal effect with some animals showing more widespread recombination, there was no ectopic activity in most adults. This Vasa-Cre transgenic line should thus prove useful for genetic analysis of diverse aspects of gametogenesis and as a general deletor line.


Asunto(s)
ARN Helicasas DEAD-box/genética , Gametogénesis/genética , Células Germinativas/enzimología , Integrasas/genética , Ratones Transgénicos/genética , Animales , Genotipo , Ratones , Regiones Promotoras Genéticas , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
12.
Reproduction ; 133(5): 855-63, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17616716

RESUMEN

Primordial follicles are long-lived structures assembled early in life. The mechanisms that control the balance between the conservation and the activation of primordial follicles are critically important for fertility and dictate the onset of menopause. The forkhead transcription factor Foxo3 serves an essential role in these processes by suppressing the growth of primordial follicles, thereby preserving them until later in life. While other factors regulating primordial follicle growth have been described, most serve multiple functions at several stages of female germ cell or follicle development, and corresponding mouse mutants exhibit pleiotropic phenotypes with disruption of multiple stages of follicle assembly, development, or survival. To investigate the possibility that Foxo3 also functions in other aspects of ovarian development beyond its known role in primordial follicle activation (PFA), we performed detailed analyses of mouse ovaries including electron microscopy to study primordial follicle structure, assembly, and early growth. These analyses revealed that the timing of primordial follicle assembly, early oocyte survival, and the expression of early germ line markers were unaffected in early Foxo3 ovaries. Taken together, these studies demonstrate that the phenotype associated with Foxo3 deficiency is remarkably specific for PFA and further support the placement of Foxo3 in a unique phenotypic class among mammalian female sterile mutants. Lastly, we discuss the implications of the specificity of this mutant phenotype with regard to the hypothesis that oocyte regeneration may occur in adults and serves as a means to replenish oocytes lost via natural physiological processes.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Infertilidad Femenina/metabolismo , Folículo Ovárico/fisiología , Animales , Apoptosis , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/análisis , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Infertilidad Femenina/patología , Ciclo Menstrual , Ratones , Ratones Noqueados , Oocitos/fisiología , Oocitos/ultraestructura , Folículo Ovárico/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA