Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Nephrol ; 27(2): 110-121, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36264415

RESUMEN

BACKGROUND: Anti-glomerular basement membrane (anti-GBM) nephritis, characterized by glomerular crescent formation, requires early treatment because of poor prognosis. Hydroxychloroquine (HCQ) is an antimalarial drug with known immunomodulatory, anti-inflammatory, and autophagy inhibitory effects; it is recognized in the treatment of autoimmune diseases such as systemic lupus erythematosus. However, its effect on anti-GBM nephritis remains unknown. In this study, we investigated the effect of HCQ on anti-GBM nephritis in rats. METHODS: Seven-weeks-old male WKY rats were administered anti-GBM serum to induce anti-GBM nephritis. Either HCQ or vehicle control was administered from day 0 to day 7 after the induction of nephritis. Renal function was assessed by measuring serum creatinine, proteinuria, and hematuria. Renal histological changes were assessed by PAS staining and Masson trichrome staining, and infiltration of macrophages was assessed by ED-1 staining. Mitogen-activated protein kinase (MAPK) was evaluated by western blotting, while chemokine and inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay using urine sample. RESULTS: HCQ treatment suppressed the decline in renal function. Histologically, extracapillary and intracapillary proliferations were observed from day 1, while fibrinoid necrosis and ED-1 positive cells were observed from day 3. Rats with anti-GBM nephritis showed high levels of monocyte chemotactic protein-1 and tumor necrosis factor-α. These changes were significantly suppressed following HCQ treatment. In addition, HCQ suppressed JNK/p38 MAPK phosphorylation. CONCLUSION: HCQ attenuates anti-GBM nephritis by exerting its anti-inflammatory effects via the inhibition of JNK/p38 MAPK activation, indicating its therapeutic potential against anti-GBM nephritis.


Asunto(s)
Glomerulonefritis , Nefritis , Ratas , Masculino , Animales , Proteínas Quinasas p38 Activadas por Mitógenos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Ratas Endogámicas WKY , Nefritis/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Glomerulonefritis/patología
2.
Int J Mol Sci ; 24(8)2023 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-37108115

RESUMEN

Growing evidence indicates that hepatocyte growth factor (HGF) possesses potent antifibrotic activity. Furthermore, macrophages migrate to inflamed sites and have been linked to the progression of fibrosis. In this study, we utilized macrophages as vehicles to express and deliver the HGF gene and investigated whether macrophages carrying the HGF expression vector (HGF-M) could suppress peritoneal fibrosis development in mice. We obtained macrophages from the peritoneal cavity of mice stimulated with 3% thioglycollate and used cationized gelatin microspheres (CGMs) to produce HGF expression vector-gelatin complexes. Macrophages phagocytosed these CGMs, and gene transfer into macrophages was confirmed in vitro. Peritoneal fibrosis was induced by intraperitoneal injection of chlorhexidine gluconate (CG) for three weeks; seven days after the first CG injection, HGF-M was administered intravenously. Transplantation of HGF-M significantly suppressed submesothelial thickening and reduced type III collagen expression. Moreover, in the HGF-M-treated group, the number of α-smooth muscle actin- and TGF-ß-positive cells were significantly lower in the peritoneum, and ultrafiltration was preserved. Our results indicated that the transplantation of HGF-M prevented the progression of peritoneal fibrosis and indicated that this novel gene therapy using macrophages may have potential for treating peritoneal fibrosis.


Asunto(s)
Fibrosis Peritoneal , Ratones , Animales , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/terapia , Fibrosis Peritoneal/metabolismo , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Gelatina/metabolismo , Modelos Animales de Enfermedad , Actinas/metabolismo , Peritoneo/patología , Fibrosis , Macrófagos/metabolismo
3.
Med Mol Morphol ; 55(1): 27-40, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34622315

RESUMEN

Peritoneal fibrosis is a serious complication of long-term peritoneal dialysis, attributable to inflammation and mitochondrial dysfunction. Mitochonic acid-5 (MA-5), an indole-3-acetic acid derivative, improves mitochondrial dysfunction and has therapeutic potential against various diseases including kidney diseases. However, whether MA-5 is effective against peritoneal fibrosis remains unclear. Therefore, we investigated the effect of MA-5 using a peritoneal fibrosis mouse model. Peritoneal fibrosis was induced in C57BL/6 mice via intraperitoneal injection of chlorhexidine gluconate (CG) every other day for 3 weeks. MA-5 was administered daily by oral gavage. The mice were divided into control, MA-5, CG, and CG + MA-5 groups. Following treatment, immunohistochemical analyses were performed. Fibrotic thickening of the parietal peritoneum induced by CG was substantially attenuated by MA-5. The number of α-smooth muscle actin-positive myofibroblasts, transforming growth factor ß-positive cells, F4/80-positive macrophages, monocyte chemotactic protein 1-positive cells, and 4-hydroxy-2-nonenal-positive cells was considerably decreased. In addition, reduced ATP5a1-positive and uncoupling protein 2-positive cells in the CG group were notably increased by MA-5. MA-5 may ameliorate peritoneal fibrosis by suppressing macrophage infiltration and oxidative stress, thus restoring mitochondrial function. Overall, MA-5 has therapeutic potential against peritoneal fibrosis.


Asunto(s)
Fibrosis Peritoneal , Animales , Clorhexidina/análogos & derivados , Modelos Animales de Enfermedad , Ácidos Indolacéticos , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/prevención & control , Peritoneo/metabolismo , Peritoneo/patología , Fenilbutiratos/química
4.
Genes Dev ; 27(2): 169-81, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23322300

RESUMEN

The liver is a unique organ with a remarkably high potential to regenerate upon injuries. In severely damaged livers where hepatocyte proliferation is impaired, facultative liver progenitor cells (LPCs) proliferate and are assumed to contribute to regeneration. An expansion of LPCs is often observed in patients with various types of liver diseases. However, the underlying mechanism of LPC activation still remains largely unknown. Here we show that a member of the fibroblast growth factor (FGF) family, FGF7, is a critical regulator of LPCs. Its expression was induced concomitantly with LPC response in the liver of mouse models as well as in the serum of patients with acute liver failure. Fgf7-deficient mice exhibited markedly depressed LPC expansion and higher mortality upon toxin-induced hepatic injury. Transgenic expression of FGF7 in vivo led to the induction of cells with characteristics of LPCs and ameliorated hepatic dysfunction. We revealed that Thy1(+) mesenchymal cells produced FGF7 and appeared in close proximity to LPCs, implicating a role for those cells as the functional LPC niche in the regenerating liver. These findings provide new insights into the cellular and molecular basis for LPC regulation and identify FGF7 as a potential therapeutic target for liver diseases.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/citología , Regeneración Hepática/fisiología , Transducción de Señal , Células Madre/citología , Animales , Proliferación Celular , Factor 7 de Crecimiento de Fibroblastos/genética , Hepatocitos/metabolismo , Hepatopatías/fisiopatología , Ratones , Células Madre/metabolismo , Antígenos Thy-1/metabolismo , Regulación hacia Arriba
5.
Histochem Cell Biol ; 154(2): 123-134, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32653936

RESUMEN

Mammalian spermatogenesis is characterized by disproportionate germ cell apoptosis. The high frequency of apoptosis is considered a safety mechanism that serves to avoid unfavorable transmission of paternal aberrant genetic information to the offspring as well as elimination mechanism for removal of overproduced immature or damaged spermatogenic cells. The molecular mechanisms involved in the induction of germ cell apoptosis include both intrinsic mitochondrial Bcl-2/Bax and extrinsic Fas/FasL pathways. However, little is known about the nuclear trigger of those systems. Recent studies indicate that epigenomes are essential in the regulation of gene expression through remodeling of the chromatin structure, and are genome-like transmission materials that reflect the effects of various environmental factors. In spermatogenesis, epigenetic errors can act as the trigger for elimination of germ cells with abnormal chromatin structure, abnormal gene expression and/or morphological defects (disordered differentiation). In this review, we focus on the relationship between global changes in epigenetic parameters and germ cell apoptosis in mice and other mammals.


Asunto(s)
Apoptosis/genética , Epigenoma/genética , Células Germinativas/metabolismo , Espermatogénesis/genética , Animales , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Células Germinativas/patología , Ratones
6.
Histochem Cell Biol ; 153(4): 287-288, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32206867

RESUMEN

The figure shown below is the correct version. We apologize for the mistake.

7.
Clin Exp Nephrol ; 24(5): 411-419, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31912273

RESUMEN

BACKGROUND: Prothymosin alpha (ProTα) is a nuclear protein expressed in virtually all mammalian tissues. Previous studies have shown that ProTα exhibits protective effects against ischemia-induced cell death in various cell types. Recently, the 6-residue peptide P6Q (NEVDQE), the modified form of the active 6-residue core (51-56) in ProTα, has also been shown to have protective effects against retinal ischemia. However, it remains to be elucidated whether P6Q is effective against acute kidney injury (AKI). Therefore, we investigated the renoprotective effect of P6Q on cisplatin-induced AKI. METHODS: Cultured HK-2 cells were treated with cisplatin for 24 h and pretreatment with ProTα or P6Q was carried out 30 min before cisplatin treatment. Cell viability was evaluated using the MTT assay. In an in vivo study, 8-week-old male Wistar rats were divided into control, cisplatin treated, and cisplatin treated with P6Q injection groups. In the last of these, P6Q was injected intravenously before cisplatin treatment. Then, we evaluated the renoprotective effect of P6Q. RESULTS: In the study on cultured cells, pretreatment with ProTα or P6Q prevented cisplatin-induced cell death. In the in vivo study, pretreatment with P6Q significantly attenuated cisplatin-induced increase in serum creatinine and blood urea nitrogen levels, renal tubular cell injury, and apoptosis. Moreover, P6Q attenuated the mitochondrial apoptotic pathway and accelerated Akt phosphorylation after cisplatin-induced renal damage. CONCLUSION: Taken together, our findings indicate that P6Q can attenuate cisplatin-induced AKI and suppress the mitochondrial apoptotic pathway via Akt phosphorylation. These data suggest that P6Q has potential as a preventative drug for cisplatin-induced AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Túbulos Renales Proximales/patología , Mitocondrias/metabolismo , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Animales , Antineoplásicos/farmacología , Nitrógeno de la Urea Sanguínea , Línea Celular , Cisplatino/farmacología , Creatinina/sangre , Humanos , Masculino , Péptidos/farmacología , Péptidos/uso terapéutico , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
8.
Histochem Cell Biol ; 151(4): 291-303, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30511269

RESUMEN

Diethylstilbestrol (DES), an estrogen agonist, increases prolactin (PRL) cells through transdifferentiation of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) cells to PRL cells as well as proliferation of PRL cells in adult male mouse pituitary. Since hyperacetylation of histone H3 is implicated in the regulation of activation of various genes, we examined the effect of DES on the state of histone H3 acetylation. DES significantly reduced the immunohistochemical signal for acetylated histone H3 at lysine 9 (H3K9ac) in PRL, LH and FSH cells, but not for H3K18ac or H3K23ac. DES-treated mice were injected intraperitoneally with HDAC inhibitors (HDACi), sodium phenylbutyrate (NaPB) or valproic acid (VPA), to mimic the acetylation level of histone H3. As expected, HDACi treatment restored the level of H3K9ac expression in these cells, and also inhibited DES-induced increase in PRL cells. Furthermore, NaPB and VPA also abrogated the effects of DES on the population density of both LH and FSH cells. Similarly, the numbers of proliferating and apoptotic cells in the pituitary in NaPB- or VPA-treated mice were comparable to those of the control mice. Considered together, these results indicated that the acetylation level of histone H3 plays an important role in DES-induced transdifferentiation of LH to PRL cells as well as proliferation of PRL cells.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Lactotrofos/efectos de los fármacos , Fenilbutiratos/farmacología , Hipófisis/efectos de los fármacos , Ácido Valproico/farmacología , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dietilestilbestrol/administración & dosificación , Dietilestilbestrol/farmacología , Gonadotrofos/citología , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/análisis , Histonas/biosíntesis , Inyecciones Intraperitoneales , Lactotrofos/citología , Masculino , Ratones , Ratones Endogámicos ICR , Fenilbutiratos/administración & dosificación , Hipófisis/metabolismo , Conejos , Ácido Valproico/administración & dosificación
11.
J Cell Sci ; 127(Pt 9): 2040-52, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610943

RESUMEN

Cdc42 is a key regulator of dynamic actin organization. However, little is known about how Cdc42-dependent actin regulation influences steady-state actin structures in differentiated epithelia. We employed inner ear hair-cell-specific conditional knockout to analyze the role of Cdc42 in hair cells possessing highly elaborate stable actin protrusions (stereocilia). Hair cells of Atoh1-Cre;Cdc42(flox/flox) mice developed normally but progressively degenerated after maturation, resulting in progressive hearing loss particularly at high frequencies. Cochlear hair cell degeneration was more robust in inner hair cells than in outer hair cells, and began as stereocilia fusion and depletion, accompanied by a thinning and waving circumferential actin belt at apical junctional complexes (AJCs). Adenovirus-encoded GFP-Cdc42 expression in hair cells and fluorescence resonance energy transfer (FRET) imaging of hair cells from transgenic mice expressing a Cdc42-FRET biosensor indicated Cdc42 presence and activation at stereociliary membranes and AJCs in cochlear hair cells. Cdc42-knockdown in MDCK cells produced phenotypes similar to those of Cdc42-deleted hair cells, including abnormal microvilli and disrupted AJCs, and downregulated actin turnover represented by enhanced levels of phosphorylated cofilin. Thus, Cdc42 influenced the maintenance of stable actin structures through elaborate tuning of actin turnover, and maintained function and viability of cochlear hair cells.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Actinas/metabolismo , Animales , Técnicas Biosensibles , Cóclea/citología , Cóclea/metabolismo , Perros , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunohistoquímica , Hibridación in Situ , Células de Riñón Canino Madin Darby , Ratones , Microscopía Electroquímica de Rastreo , Microscopía Electrónica de Transmisión , Técnicas de Cultivo de Órganos/métodos , Proteína de Unión al GTP cdc42/genética
12.
Nephrol Dial Transplant ; 31(10): 1615-23, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27190365

RESUMEN

BACKGROUND: Although histone acetylation, an epigenetic modification, has been reported to be related to the progression of various diseases, its involvement in nephrosclerosis is unclear. METHODS: Dahl salt-sensitive rats were used as a model of nephrosclerosis in this study. The rats were divided into three groups: (i) normal-salt diet group, (ii) high-salt diet group (HS), and (iii) HS administered daily with curcumin, a histone acetyltransferase inhibitor (HS+C). At 6 weeks after the treatment, the kidneys were dissected. Morphologic changes were assessed by Masson's trichrome staining. The number of macrophages, fibroblasts and the cells expressing acetylated histone H3 at Lys 9 (H3K9) were assessed by immunohistochemistry. RESULTS: Although both HS and HS+C rats revealed a marked increase in systolic blood pressure, serum creatinine was increased only in HS rats at 6 weeks. In the HS rats, nephrosclerosis was induced, accompanying a significant accumulation of macrophages and fibroblasts. The inflammation and fibrosis was markedly suppressed in the HS+C group. The level of histone acetylation at Lys 9 was enhanced in the HS rats, whereas curcumin administration suppressed the histone acetylation. Moreover, in the HS rats, interleukin-6 gene expression was associated with acetylated H3K9, as revealed by chromatin immunoprecipitation assay. CONCLUSIONS: Our results suggested that curcumin ameliorates nephrosclerosis via suppression of histone acetylation, independently of hypertension.


Asunto(s)
Curcumina/uso terapéutico , Histonas/metabolismo , Hipertensión/tratamiento farmacológico , Nefroesclerosis/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Acetilación , Animales , Presión Sanguínea , Curcumina/farmacología , Evaluación Preclínica de Medicamentos , Epigénesis Genética/efectos de los fármacos , Expresión Génica , Hipertensión/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Nefroesclerosis/metabolismo , Ratas , Ratas Endogámicas Dahl
13.
Acta Derm Venereol ; 96(6): 748-53, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-26912390

RESUMEN

There has been no previous systematic study of bullous skin diseases with granular basement membrane zone deposition exclusively of C3. In this study we collected 20 such patients, none of whom showed cutaneous vasculitis histopathologically. Oral dapsone and topical steroids were effective. Various serological tests detected no autoantibodies or autoantigens. Direct immunofluorescence for various complement components revealed deposition only of C3 and C5-C9, indicating that no known complement pathways were involved. Studies of in situ hybridization and micro-dissection with quantitative RT-PCR revealed a slight reduction in expression of C3 in patient epidermis. These patients may represent a new disease entity, for which we propose the term "granular C3 dermatosis". The mechanism for granular C3 deposition in these patients is unknown, but it is possible that the condition is caused by autoantibodies to skin or aberrant C3 expression in epidermal keratinocytes.


Asunto(s)
Membrana Basal/metabolismo , Complemento C3/metabolismo , Dermatitis Herpetiforme/metabolismo , Enfermedades Cutáneas Vesiculoampollosas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antiinfecciosos/uso terapéutico , Niño , Dapsona/uso terapéutico , Dermatitis Herpetiforme/tratamiento farmacológico , Dermatitis Herpetiforme/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente Directa , Humanos , Immunoblotting , Hibridación in Situ , Japón , Queratinocitos/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Enfermedades Cutáneas Vesiculoampollosas/tratamiento farmacológico , Enfermedades Cutáneas Vesiculoampollosas/patología , Esteroides/uso terapéutico
14.
Med Mol Morphol ; 49(3): 144-53, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26880269

RESUMEN

Long-term peritoneal dialysis causes peritoneal fibrosis, and previous reports suggest that inflammation plays a critical role in peritoneal fibrosis. Chondroitin sulfate (CS) suppresses the inflammatory response by preventing activation of nuclear factor (NF)-κB. We examined the effect of CS on the peritoneal fibrosis induced by chlorhexidine gluconate (CG) in mice. CS or water was administered daily. We divided mice into four groups: administered vehicle and water (control); administered vehicle and CS (CS); administered CG and water (CG); and administered CG and CS (CG+CS). Morphologic changes were assessed by Masson's trichrome staining. Inflammation- and fibrosis-associated factors were assessed by immunohistochemistry. Activation of NF-κB was examined by southwestern histochemistry. CS administration suppressed the progression of submesothelial thickening. The numbers of inflammation- and fibrosis-associated factors -positive cells were significantly decreased in the CG+CS group, compared to the CG group. Based on SWH, the CG+CS group contained significantly fewer NF-κB-activated cells than the CG group. Our results indicate that CS suppresses peritoneal fibrosis via suppression of NF-κB activation. These results suggest that CS has therapeutic potential for peritoneal fibrosis.


Asunto(s)
Sulfatos de Condroitina/uso terapéutico , FN-kappa B/metabolismo , Fibrosis Peritoneal/tratamiento farmacológico , Actinas/metabolismo , Animales , Quimiocina CCL2/metabolismo , Sulfatos de Condroitina/farmacología , Inmunohistoquímica , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos ICR , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fibrosis Peritoneal/enzimología , Fibrosis Peritoneal/patología , Fibrosis Peritoneal/prevención & control , Fenotipo , Fosforilación , Proteínas Smad/metabolismo , Coloración y Etiquetado
15.
Histochem Cell Biol ; 143(2): 209-24, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25326673

RESUMEN

Histone acetylation is involved in the regulation of chromatin structure and gene function. We reported previously that histone H3 acetylation pattern is subject to dynamic changes and limited to certain stages of germ cell differentiation during murine spermatogenesis, suggesting a crucial role for acetylation in the process. In the present study, we investigated the effects of hyper- and hypo-acetylation on spermatogenesis. Changes in acetylation level were induced by either in vivo administration of sodium phenylbutyrate, a histone deacetylase inhibitor, or by knockdown of histone acetyltransferases using short hairpin RNA plasmids transfection. Administration of sodium phenylbutyrate induced accumulation of acetylated histone H3 at lysine 9 and lysine 18 in round spermatids, together with spermatid morphological abnormalities and induction of apoptosis through a Bax-related pathway. Knockdown of steroid receptor coactivator 1, a member of histone acetyltransferases, but not general control of amino acid synthesis 5 nor elongator protein 3 by in vivo electroporation of shRNA plasmids, reduced acetylated histone H3 at lysine 9 in round spermatids, and induced morphological abnormalities. We concluded that the proper regulation of histone H3 acetylation levels is important for spermatid differentiation and complex chromatin remodeling during spermiogenesis.


Asunto(s)
Histonas/metabolismo , Espermátides/patología , Testículo/fisiopatología , Acetilación/efectos de los fármacos , Animales , Apoptosis , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Inmunohistoquímica , Masculino , Ratones , Tamaño de los Órganos , Fenilbutiratos/farmacología , Espermátides/efectos de los fármacos , Espermátides/enzimología , Espermátides/ultraestructura , Espermatogénesis/genética , Testículo/efectos de los fármacos , Testículo/enzimología , Testículo/ultraestructura
16.
FASEB J ; 28(2): 871-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24221084

RESUMEN

Although the roles of acids in bone metabolism are well characterized, the function of proton-sensing receptors in bone metabolism remains to be explored. In this study, we evaluated the role of proton-sensing receptor T-cell death-associated gene 8 (TDAG8) in osteoclastic activity during bone loss after ovariectomy. Through observations of bone mineral content, we found that pathological bone resorption was significantly exacerbated in mice homozygous for a gene trap mutation in the Tdag8 gene. Furthermore, osteoclasts from the homozygous mutant mice resorbed calcium in vitro more than the osteoclasts from the heterozygous mice did. Impaired osteoclast formation under acidic conditions was ameliorated in cultures of bone marrow cells by Tdag8 gene mutation. Extracellular acidification changed the cell morphology of osteoclasts via the TDAG8-Rho signaling pathway. These results suggest that the enhancement of TDAG8 function represents a new strategy for preventing bone resorption diseases, such as osteoporosis.


Asunto(s)
Resorción Ósea/metabolismo , Osteoclastos/metabolismo , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Hibridación in Situ , Ratones , Microscopía Confocal , Ovariectomía , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Biol Pharm Bull ; 38(2): 193-200, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25747978

RESUMEN

Long-term peritoneal dialysis therapy causes inflammation and histological changes in the peritoneal membrane. Inflammation generally activates fibroblasts and results in fibroblast-myofibroblast differentiation. Heat-shock protein 47 (HSP 47), a collagen-specific molecular chaperone, is localized in myofibroblasts and is involved in the progression of peritoneal fibrosis. Daikenchuto (DKT), a Kampo medicine, is used to prevent postoperative colon adhesion. It inhibits inflammation and HSP 47 expression in the gastrointestinal tract. We examined the effect of DKT on chlorhexidine gluconate (CG)-induced peritoneal fibrosis in mice injected with 0.1% CG dissolved in 15% ethanol. DKT was dissolved in the drinking water. Histological changes were assessed using Masson trichrome staining. Cells expressing α-smooth muscle actin (α-SMA), HSP 47, phospho-Smad 2/3, F4/80, and monocyte chemotactic protein-1 were examined immunohistochemically. Compared with the control group, the peritoneal tissues of the CG group were markedly thickened, and the number of cells expressing α-SMA, HSP 47, phospho-Smad 2/3, F4/80, and monocyte chemotactic protein-1 was significantly increased. However, these changes were inhibited in the DKT-treated group. These results indicate that DKT can prevent peritoneal fibrosis by inhibiting inflammation and HSP 47 expression.


Asunto(s)
Fibrosis Peritoneal/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Actinas/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Quimiocina CCL2/metabolismo , Clorhexidina/análogos & derivados , Proteínas del Choque Térmico HSP47/metabolismo , Masculino , Medicina Kampo , Ratones Endogámicos ICR , Panax , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Peritoneo/patología , Fitoterapia , Extractos Vegetales/farmacología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Zanthoxylum , Zingiberaceae
18.
Eur Arch Otorhinolaryngol ; 272(10): 2689-96, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25138153

RESUMEN

We reported previously that keratinocyte growth factor (KGF), a mesenchymal cell-derived paracrine growth factor, plays an important role in middle ear cholesteatoma formation, which is characterized by marked proliferation of epithelial cells. Here, we investigated whether KGF, the main factor that induces cholesteatoma, overexpression in vivo results in the formation of cholesteatoma. Flag-hKGF cDNA driven by CMV14 promoter was transfected through electroporation into the external auditory canal (EAC) of rats once (short-term model) or five times on every fourth day (long-term model). Ears transfected with empty vector were used as controls. Successful transfection of plasmids into epithelial and stromal cells was confirmed by Flag immunohistochemistry. In the short-term model, the intensity of KGF protein was the strongest in hKGF transfected ear at day 4. KGF expression induced epithelial cell proliferation, reaching a peak level at day 4 and then decreased later, while in the long-term model, KGF expression in the EAC led to middle ear cholesteatoma formation. In conclusion, we described here a new experimental model of human middle ear cholesteatoma, and demonstrated that KGF and KGF receptor paracrine action play an essential role in middle ear cholesteatoma formation in an in vivo model.


Asunto(s)
Colesteatoma del Oído Medio/genética , ADN/genética , Factor 7 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Animales , Colesteatoma del Oído Medio/metabolismo , Modelos Animales de Enfermedad , Factor 7 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley
20.
Reprod Med Biol ; 13(2): 59-69, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29699150

RESUMEN

Mammalian spermatogenesis is a successive process consisting of spermatogonial proliferation, spermatocytic meiosis, and spermiogenesis, representing the maturation of haploid spermatids. During the process, 25-75 % of the expected sperm yield is thought to be lost through apoptosis. In addition, spermatogenesis is considered to be a process undergoing successive heterochromatinization, finally reaching a complete condensed form in the sperm head. Thus, cell proliferation, differentiation and death may be strictly regulated by epigenetic factors in this process. This review describes the current understanding of the role of epigenome in spermatogenesis, especially focusing on the following aspects; DNA methylation, modification of histones, and small RNA function. These epigenetic factors affect each other and play a central role in events essential for spermatogenesis, fertilization and embryogenesis, through the regulation of gene expression, transposon activities, meiotic sex chromosome inactivation, histone remodeling and genome imprinting. Finally, a brief discussion of future avenues of study is highlighted.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA