Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
2.
Nat Immunol ; 18(11): 1261-1269, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28945244

RESUMEN

The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Virus del Dengue/inmunología , Epítopos/inmunología , Proteínas del Envoltorio Viral/inmunología , Infección por el Virus Zika/inmunología , Animales , Encéfalo/inmunología , Encéfalo/virología , Chlorocebus aethiops , Reacciones Cruzadas/inmunología , Virus del Dengue/clasificación , Virus del Dengue/metabolismo , Femenino , Feto/inmunología , Feto/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Ratones , Pruebas de Neutralización , Embarazo , Multimerización de Proteína/inmunología , Testículo/inmunología , Testículo/virología , Células Vero , Proteínas del Envoltorio Viral/química , Carga Viral/inmunología , Virus Zika/inmunología , Virus Zika/fisiología , Infección por el Virus Zika/virología
3.
Nature ; 540(7633): 438-442, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798603

RESUMEN

Infection of pregnant women with Zika virus (ZIKV) can cause congenital malformations including microcephaly, which has focused global attention on this emerging pathogen. In addition to transmission by mosquitoes, ZIKV can be detected in the seminal fluid of affected males for extended periods of time and transmitted sexually. Here, using a mouse-adapted African ZIKV strain (Dakar 41519), we evaluated the consequences of infection in the male reproductive tract of mice. We observed persistence of ZIKV, but not the closely related dengue virus (DENV), in the testis and epididymis of male mice, and this was associated with tissue injury that caused diminished testosterone and inhibin B levels and oligospermia. ZIKV preferentially infected spermatogonia, primary spermatocytes and Sertoli cells in the testis, resulting in cell death and destruction of the seminiferous tubules. Less damage was caused by a contemporary Asian ZIKV strain (H/PF/2013), in part because this virus replicates less efficiently in mice. The extent to which these observations in mice translate to humans remains unclear, but longitudinal studies of sperm function and viability in ZIKV-infected humans seem warranted.


Asunto(s)
Testículo/patología , Testículo/virología , Infección por el Virus Zika/patología , Virus Zika/patogenicidad , Animales , Muerte Celular , Virus del Dengue/fisiología , Epidídimo/patología , Epidídimo/virología , Humanos , Inhibinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oligospermia/patología , Oligospermia/virología , Túbulos Seminíferos/patología , Túbulos Seminíferos/virología , Células de Sertoli/virología , Espermatocitos/virología , Espermatogonias/virología , Testosterona/metabolismo , Factores de Tiempo
4.
Am J Physiol Heart Circ Physiol ; 316(5): H1202-H1210, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30901280

RESUMEN

Maternal obesity is correlated with cardiovascular disease in offspring, with a 1.3-fold increase in events observed in offspring of obese women. We have observed that obesity-exposed oocytes demonstrate impaired mitophagy and transmit damaged mitochondria to the offspring. Accordingly, we hypothesized that maternal obesity induces cardiac mitochondrial dysfunction in the offspring via transgenerational inheritance of abnormal oocyte mitochondria. We mated female mice fed a high-fat/high-sucrose (HFS) diet (or chow) with chow-fed males and assessed cardiac structure and function in their descendants that were chow fed in each generation. All F1 to F3 descendants bred via the female in each generation were nonobese and demonstrated cardiac mitochondrial abnormalities with crystal rarefaction and reduced oxygen consumption pointing to a transgenerational effect, while obese F0 dams' hearts were unaffected. Furthermore, male offspring from F1 to F3 generations and female F1 and F2 offspring developed increased left ventricular (LV) mass (vs. chow-fed controls). Increased LV mass was also observed in offspring generated by in vitro fertilization of obesity-exposed oocytes and gestation in nonobese surrogates, ruling out a gestational environment effect. Contrary to our hypothesis, male F1 also transmitted these effects to their offspring, ruling out maternal mitochondria as the primary mode of transmission. We conclude that transmission of obesity-induced effects in the oocyte nucleus rather than abnormal mitochondria underlie transgenerational inheritance of cardiac mitochondrial defects in descendants of obese females. These findings will spur exploration of epigenetic alterations in the oocyte genome as potential mechanisms whereby a family history of maternal obesity predisposes to cardiovascular disease in humans.


Asunto(s)
Núcleo Celular/genética , Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/efectos adversos , Genes Mitocondriales , Cardiopatías/genética , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Obesidad Materna/genética , Efectos Tardíos de la Exposición Prenatal , Alimentación Animal , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Núcleo Celular/metabolismo , Núcleo Celular/patología , Modelos Animales de Enfermedad , Femenino , Ganancia de Peso Gestacional , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/fisiopatología , Herencia , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/patología , Obesidad Materna/metabolismo , Obesidad Materna/fisiopatología , Oocitos/metabolismo , Oocitos/patología , Embarazo , Factores de Riesgo
5.
FASEB J ; 32(1): 63-72, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28842431

RESUMEN

Male exposure to cigarette smoke is associated with seminal defects and with congenital anomalies and childhood cancers in offspring. In mice, paternal exposure to cigarette smoke condensate (CSC) causes molecular defects in germ cells and phenotypic effects in their offspring. Here we used an ex vivo testicular explant model and in vivo exposure to determine the concentration at which CSC impairs spermatogenesis and offspring development. We explanted testis tissue at postnatal day (P)5.5 and cultured it until P11.5. Assessment of growth parameters by analyzing expression of cell-specific markers revealed that the explant system maintained structural and functional integrity. We exposed the P5.5 to -11.5 explants to various concentrations (40-160 µg/ml) of CSC and confirmed that nicotine in the CSC was metabolized to cotinine. We assessed various growth and differentiation parameters, as well as testosterone production, and observed that many spermatogenesis features were impaired at 160 µg/ml CSC. The same parameters were impaired by a similar CSC concentration in vivo Finally, females mated to males that were exposed to 160 µg/ml CSC neonatally had increased rates of pup resorption. We conclude that male exposure to CSC impairs offspring development and that the concentration at which CSC impairs spermatogenesis is similar in vivo and ex vivo. Given that the concentrations of CSC we used contained similar doses of nicotine as human smokers are exposed to, we argue that our model mimics human male reproductive effects of smoking.-Esakky, P., Hansen, D. A., Drury, A. M., Felder, P., Cusumano, A., Moley, K. H. Testicular cells exhibit similar molecular responses to cigarette smoke condensate ex vivo and in vivo.


Asunto(s)
Fumar/efectos adversos , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Animales Recién Nacidos , Células Cultivadas , Cotinina/metabolismo , Pérdida del Embrión/inducido químicamente , Desarrollo Embrionario/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Nicotina/metabolismo , Embarazo , Humo/efectos adversos , Testículo/metabolismo , Testículo/patología , Testosterona/metabolismo , Técnicas de Cultivo de Tejidos
6.
Dev Biol ; 426(1): 126-138, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28438607

RESUMEN

Mitochondria are the most prominent organelle in the oocyte. Somatic cells maintain a healthy population of mitochondria by degrading damaged mitochondria via mitophagy, a specialized autophagy pathway. However, evidence from previous work investigating the more general macroautophagy pathway in oocytes suggests that mitophagy may not be active in the oocyte. This would leave the vast numbers of mitochondria - poised to be inherited by the offspring - vulnerable to damage. Here we test the hypothesis that inactive mitophagy in the oocyte underlies maternal transmission of dysfunctional mitochondria. To determine whether oocytes can complete mitophagy, we used either CCCP or AntimycinA to depolarize mitochondria and trigger mitophagy. After depolarization, we did not detect co-localization of mitochondria with autophagosomes and mitochondrial DNA copy number remained unchanged, indicating the non-functional mitochondrial population was not removed. To investigate the impact of an absence of mitophagy in oocytes with damaged mitochondria on offspring mitochondrial function, we utilized in vitro fertilization of high fat high sugar (HF/HS)-exposed oocytes, which have lower mitochondrial membrane potential and damaged mitochondria. Here, we demonstrate that blastocysts generated from HF/HS oocytes have decreased mitochondrial membrane potential, lower metabolites involved in ATP generation, and accumulation of PINK1, a mitophagy marker protein. This mitochondrial phenotype in the blastocyst mirrors the phenotype we show in HF/HS exposed oocytes. Taken together, these data suggest that the mechanisms governing oocyte mitophagy are fundamentally distinct from those governing somatic cell mitophagy and that the absence of mitophagy in the setting of HF/HS exposure contributes to the oocyte-to-blastocyst transmission of dysfunctional mitochondria.


Asunto(s)
Autofagia/fisiología , ADN Mitocondrial/genética , Dosificación de Gen/genética , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/patología , Mitofagia/fisiología , Oocitos/metabolismo , Animales , Antimicina A/toxicidad , Células Cultivadas , Femenino , Dosificación de Gen/efectos de los fármacos , Hidrazonas/toxicidad , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Obesidad/patología
7.
Biol Reprod ; 90(1): 9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24258214

RESUMEN

Prior studies in our laboratory have demonstrated that cigarette smoke condensate (CSC) activates arylhydrocarbon receptor (Ahr) leading to upregulation of several antioxidant enzymes in murine spermatocytes. In this study, we show that exposure of the spermatocyte cell line GC-2spd(ts) to CSC induces an increase in Cyp1a1, demonstrating AHR activation, and simultaneous expression and nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2), where it is believed to modulate Ahr expression by a feedback mechanism. Pharmacological inhibition by the AHR-antagonist CH223191 and interference by Ahr- and Nrf2-small interfering RNA followed by quantitative real-time PCR implicate the Ahr-Nrf2 pathway in the modulation of DNA damage and growth suppression genes such as Gadd45a and P21 and oxidative stress-related genes Cyp1a1, Nrf2, and Ahrr. Flow cytometry accompanied with cell proliferation assay indicate the CSC induces accumulation of spermatocytes at the S-G2/M phase of the cell cycle. Thus, the data obtained suggest that CSC contains several AHR-agonists that are capable of altering the growth pattern of spermatocytes in vitro through the Ahr-Nrf2 signaling mechanism.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Ciclo Celular/efectos de los fármacos , Factor 2 Relacionado con NF-E2/fisiología , Nicotiana/toxicidad , Receptores de Hidrocarburo de Aril/fisiología , Humo/efectos adversos , Espermatocitos/citología , Espermatocitos/efectos de los fármacos , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Ratones , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Espermatocitos/fisiología
8.
Biol Reprod ; 90(1): 8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24174576

RESUMEN

The aryl hydrocarbon receptor (AHR) is known for its roles in xenobiotic metabolism and essential physiologic processes such as cell growth, death, and differentiation. AHR is also an important regulator of male reproductive processes. However, no studies have characterized the consequences of loss of AHR in spermatogenesis. We used Ahr knockout (Ahr(-/-)) mice to assess the effects of loss of AHR on the architecture and gene expression of the seminiferous epithelium and functional sperm outcomes. The histopathological defects of the Ahr(-/-)seminiferous epithelium included vacuoles, multinucleated giant cells, hypocellularity with widened intercellular spaces, apical sloughing, and an excess number of retained elongated spermatids. Quantitative real-time PCR revealed significant down-regulation of Testin and Magea4, indicating Sertoli cell and spermatogenic dysregulation. Moreover, the reduced expression of Hspa2, Prm1, and Prm2 as well as decreased expression of Nrf2, Sod2, and Ucp2 suggested poorly remodeled germ cells with increased vulnerability to oxidative stress. In wild-type sperm, AHR protein was localized to the acrosome and the principal piece of the mature sperm flagellum. The in vitro fertilization rate was significantly lower with Ahr(-/-) sperm as compared to wild-type sperm, and there were morphologic abnormalities of the Ahr(-/-) sperm head and tail. Taken together, our data indicate that AHR plays an important role in normal sperm development.


Asunto(s)
Receptores de Hidrocarburo de Aril/fisiología , Túbulos Seminíferos/citología , Túbulos Seminíferos/crecimiento & desarrollo , Espermatogénesis/genética , Animales , Embrión de Mamíferos , Femenino , Fertilidad/genética , Fertilización/genética , Fertilización In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Túbulos Seminíferos/metabolismo , Espermatozoides/fisiología
9.
Endocrinology ; 157(2): 956-68, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26677880

RESUMEN

The most significant increase in metabolic syndrome over the previous decade occurred in women of reproductive age, which is alarming given that metabolic syndrome is associated with reproductive problems including subfertility and early pregnancy loss. Individuals with metabolic syndrome often consume excess fructose, and several studies have concluded that excess fructose intake contributes to metabolic syndrome development. Here, we examined the effects of increased fructose consumption on pregnancy outcomes in mice. Female mice fed a high-fructose diet (HFrD) for 6 weeks developed glucose intolerance and mild fatty liver but did not develop other prominent features of metabolic syndrome such as weight gain, hyperglycemia, and hyperinsulinemia. Upon mating, HFrD-exposed mice had lower pregnancy rates and smaller litters at midgestation than chow-fed controls. To explain this phenomenon, we performed artificial decidualization experiments and found that HFrD consumption impaired decidualization. This appeared to be due to decreased circulating progesterone as exogenous progesterone administration rescued decidualization. Furthermore, HFrD intake was associated with decreased bone morphogenetic protein 2 expression and signaling, both of which were restored by exogenous progesterone. Finally, expression of forkhead box O1 and superoxide dismutase 2 [Mn] proteins were decreased in the uteri of HFrD-fed mice, suggesting that HFrD consumption promotes a prooxidative environment in the endometrium. In summary, these data suggest that excess fructose consumption impairs murine fertility by decreasing steroid hormone synthesis and promoting an adverse uterine environment.


Asunto(s)
Deciduoma/efectos de los fármacos , Endometrio/efectos de los fármacos , Fructosa/toxicidad , Tamaño de la Camada/efectos de los fármacos , Índice de Embarazo , Progesterona/metabolismo , Edulcorantes/toxicidad , Aborto Espontáneo , Animales , Proteína Morfogenética Ósea 2/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Decidua/efectos de los fármacos , Decidua/metabolismo , Decidua/patología , Deciduoma/metabolismo , Deciduoma/patología , Técnicas de Cultivo de Embriones , Transferencia de Embrión , Endometrio/metabolismo , Endometrio/patología , Hígado Graso , Femenino , Fertilización In Vitro , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Intolerancia a la Glucosa , Inmunohistoquímica , Síndrome Metabólico , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Embarazo , Superovulación , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo
10.
Reprod Toxicol ; 65: 283-294, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27589885

RESUMEN

Paternal smoking is associated with infertility, birth defects and childhood cancers. Our earlier studies using cigarette smoke condensate (CSC) demonstrated several deleterious changes in male germ cells. Here, we hypothesize that chronic paternal exposure to CSC causes molecular and phenotypic changes in the sire and the offspring, respectively. In this mouse study, CSC caused DNA damage and cytotoxicity in testes via accumulation of benzo(a)pyrene (B[a]P) and cotinine. Decreased expression of growth arrest and DNA damage inducible alpha (Gadd45a), aryl hydrocarbon receptor (Ahr), and cyclin-dependent kinase inhibitor 1A (P21) was seen in CSC exposed testes. Apoptotic germ cell death was detected by induction of Fas, FasL, and activated caspase-3. The CSC-exposed males displayed reduction in sperm motility and fertilizing ability and sired pups with reduced body weight and crown-rump length, and smaller litter size with higher numbers of resorption. This model of CSC exposure demonstrates testicular toxicity and developmental defects in the offspring.


Asunto(s)
Exposición Paterna/efectos adversos , Humo/efectos adversos , Productos de Tabaco , Animales , Apoptosis/efectos de los fármacos , Benzo(a)pireno/metabolismo , Peso Corporal/efectos de los fármacos , Cotinina/metabolismo , Largo Cráneo-Cadera , Daño del ADN , Femenino , Reabsorción del Feto/inducido químicamente , Expresión Génica/efectos de los fármacos , Tamaño de la Camada/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo
11.
Cell Rep ; 16(1): 1-8, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27320925

RESUMEN

Maternal obesity impairs offspring health, but the responsible mechanisms are not fully established. To address this question, we fed female mice a high-fat/high-sugar diet from before conception until weaning and then followed the outcomes in the next three generations of offspring, all fed a control diet. We observed that female offspring born to obese mothers had impaired peripheral insulin signaling that was associated with mitochondrial dysfunction and altered mitochondrial dynamic and complex proteins in skeletal muscle. This mitochondrial phenotype persisted through the female germline and was passed down to the second and third generations. Our results indicate that maternal programming of metabolic disease can be passed through the female germline and that the transfer of aberrant oocyte mitochondria to subsequent generations may contribute to the increased risk for developing insulin resistance.


Asunto(s)
Células Germinativas/metabolismo , Síndrome Metabólico/metabolismo , Mitocondrias/patología , Animales , Dieta Alta en Grasa , Carbohidratos de la Dieta , Femenino , Intolerancia a la Glucosa/complicaciones , Patrón de Herencia , Resistencia a la Insulina , Masculino , Síndrome Metabólico/complicaciones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Oocitos/metabolismo , Oocitos/ultraestructura , Embarazo , Transducción de Señal
12.
J Mol Cell Biol ; 7(1): 73-87, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25548370

RESUMEN

Our earlier studies have demonstrated that the cigarette smoke in the form of cigarette smoke condensate (CSC) causes growth arrest of a mouse spermatocyte cell line [GC-2spd(ts)] through activation of the AHR-NRF2 pathway. The present study demonstrates the CSC-activated p38 and ERK MAPK signaling in GC-2spd(ts) via arylhydrocarbon receptor (AHR). Pharmacological inhibition by using AHR-antagonist, or p38 MAPK and ERK (MEK1) inhibitors significantly abrogates CSC-induced growth arrest by AHR and MAPK inactivation. QRT-PCR, western blot, and immunofluorescence of Ahr-target of Nrf2, and stress-inducible growth suppressive Atf3 and E2f4 following treatments indicate a crosstalk among these pathways. Regulation of Atf3 by Nrf2 and Ahr through RNA interference suggests the existence of a cross-regulatory loop between the targets. CSC induction of E2f4 via Atf3 and its regulation by pharmacological inhibitors reveal a possible regulatory mechanism of growth inhibitory CSC. SiRNA silencing of Ahr, Nrf2, Atf3, and E2f4 genes and downregulation of cyclins by CSC corroborate the growth inhibitory effect of cigarette smoke. Thus, the data obtained suggest that the CSC-mediated MAPKs and AHR-NRF2 crosstalks lay the molecular basis for the growth arrest and cell death of spermatocytes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Puntos de Control del Ciclo Celular , Factor 2 Relacionado con NF-E2/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Fumar , Espermatocitos/metabolismo , Factor de Transcripción Activador 3/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Replicación del ADN , Factor de Transcripción E2F4/metabolismo , Silenciador del Gen , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Receptores de Hidrocarburo de Aril/genética
13.
Endocrinology ; 155(8): 2781-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24840347

RESUMEN

Type 1 diabetes is associated with subfertility in humans. The current treatment for type 1 diabetes, insulin monotherapy, is suboptimal to fully stabilize glycemia, potentially leading to this subfertility. Recent work has demonstrated that treatment with the energy-regulating hormone leptin, alone or in combination with insulin, can more effectively control glycemia in mouse models of type 1 diabetes. Here, we sought to determine whether the fertility defects in a type 1 diabetic mouse model, the Akita mouse, can be rescued with leptin monotherapy in the absence of any exogenous insulin. Akita homozygous mice treated with leptin alone had a larger total body size, testes, and seminal vesicles than their untreated siblings. Leptin treatment prevented testicular degeneration and rescued sperm motility to wild-type levels. Furthermore, sperm obtained from leptin-treated mice could successfully fertilize ooctyes in vitro. Despite completely rescuing spermatogenesis, the critical reproductive hormones LH and testosterone were only modestly higher than in untreated mice, indicating that a minimum threshold of these hormones must be met to maintain spermatogenesis. Cumulatively, these findings implicate the importance of leptin in maintaining fertility and support the use of leptin therapy in the treatment of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/terapia , Infertilidad Masculina/terapia , Leptina/fisiología , Espermatogénesis/genética , Adiposidad/genética , Animales , Atrofia/genética , Atrofia/prevención & control , Atrofia/terapia , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Tamización de Portadores Genéticos , Homocigoto , Infertilidad Masculina/etiología , Infertilidad Masculina/patología , Leptina/genética , Leptina/uso terapéutico , Hormona Luteinizante/genética , Masculino , Ratones , Testosterona/genética
14.
Reprod Sci ; 20(3): 238-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22941942

RESUMEN

Laser microdissection (LMD) is a selective cell isolation technique that enables the separation of desired homogenous cell subpopulations from complex tissues such as the testes under direct microscopic visualization. The LMD accompanied by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) represents an indispensable tool in quantifying messenger RNA (mRNA) expression among defined cell populations. Gene expression is temporally and spatially regulated at 3 sequential phases of mitotic, meiotic, and postmeiotic stages of spermatogenesis. The present study demonstrates a short modified LMD protocol based upon hematoxylin and eosin (H&E) staining. Stage-specific LMD success was validated by the use of mRNA profiling of "marker genes" which are conserved across species and are known to be differentially expressed during spermatogenesis. Magea4, Hspa2, Cox6b2, Tnp1, Prm1, and Prm2 are used to differentiate among the microdissected cell populations, namely spermatogonia (group I), spermatocytes (group II), round and condensing spermatids (group III), and elongated and condensed spermatids (group IV), respectively. The LMD combined with qRT-PCR is further extended to assess the cell stage-specific distribution of selected stress response genes such as Hsp90aa1, Gpx4, Ucp2, Sod1, and Sod2. The germ cell-specific mRNA profiles are suitably complemented by Western blot of the LMD samples, immunohistochemistry, and confocal localization of the corresponding proteins. The current study suggests that LMD can successfully isolate cell subpopulations from the complex tissues of the testes; and establish cell stage-specific basal expression patterns of selected stress response genes and proteins. It is our hypothesis that the baseline expression of stress response genes will differ by cell stage to create discrete stage-specific vulnerabilities to reproductive toxicants.


Asunto(s)
Microdisección , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatogénesis/fisiología , Testículo/citología , Testículo/fisiología , Transcriptoma/fisiología , Animales , Células Germinativas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microdisección/métodos , Microscopía Confocal/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Espermátides/fisiología , Espermatocitos/fisiología
15.
Reprod Toxicol ; 34(4): 665-76, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23069111

RESUMEN

Cigarette smoke contains numerous compounds that cause oxidative stress and alter gene expression in many tissues, and cigarette smoking is correlated with male infertility. To identify mechanisms by which this occurs, we evaluated expression of antioxidant genes in mouse spermatocytes in response to cigarette smoke condensate (CSC). CSC exposure led to oxidative stress and dose-dependent up-regulation of Hsp90aa1, Ahr, Arnt, Sod1, Sod2, and Cyp1a1 expression in a mouse spermatocyte cell line. An antagonist of the aryl hydrocarbon receptor (AHR) abrogated several CSC-mediated changes in mRNA and protein levels. Consistent with these results, spermatocytes isolated by laser-capture microdissection from CSC-treated mice showed increased expression of several antioxidant genes. In vivo exposure to CSC was genotoxic to spermatocytes, resulting in apoptosis and disruptions to the seminiferous tubules. Our in vivo and in vitro data indicate that CSC-mediated damage to murine spermatocytes is AHR-dependent and is mediated by oxidative stress.


Asunto(s)
Receptores de Hidrocarburo de Aril/genética , Humo , Espermatocitos/fisiología , Productos de Tabaco , Animales , Apoptosis , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Línea Celular , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Daño del ADN , Regulación de la Expresión Génica/fisiología , Glutatión Peroxidasa/genética , Proteínas HSP90 de Choque Térmico/genética , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Estrés Oxidativo/fisiología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Espermatocitos/patología , Superóxido Dismutasa/genética , Testículo/fisiología , Proteína Desacopladora 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA