Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Reprod Domest Anim ; 52 Suppl 2: 354-358, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27859771

RESUMEN

The intent of this contribution is to provide an update of the progress we have made towards developing a method/treatment to permanently sterilize cats. Our approach employs two complementary methodologies: RNA interference (RNAi) to silence genes involved in the central control of reproduction and a virus-based gene therapy system intended to deliver RNAi selectively to the hypothalamus (where these genes are expressed) via the systemic administration of modified viruses. We selected the hypothalamus because it contains neurons expressing Kiss1 and Tac3, two genes essential for reproduction and fertility. We chose the non-pathogenic adeno-associated virus (AAV) as a vector whose tropism could be modified to target the hypothalamus. The issues that must be overcome to utilize this vector as a delivery vehicle to induce sterility include modification of the wild-type AAV to target the hypothalamic region of the brain with a simultaneous reduction in targeting of peripheral tissues and non-hypothalamic brain regions, identification of RNAi targets that will effectively reduce the expression of Kiss1 and Tac3 without off-target effects, and determination if neutralizing antibodies to the AAV serotype of choice are present in cats. Successful resolution of these issues will pave the way for the development of a powerful tool to induce the permanent sterility in cats.


Asunto(s)
Gatos , Anticoncepción/veterinaria , Dependovirus , Silenciador del Gen , Vectores Genéticos , Hipotálamo , Animales , Anticoncepción/métodos , Expresión Génica/efectos de los fármacos , Ingeniería Genética/métodos , Ingeniería Genética/veterinaria , Infertilidad/etiología , Infertilidad/veterinaria , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/genética , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/genética , Interferencia de ARN
2.
Endocrinology ; 154(2): 942-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291449

RESUMEN

Lin28 and Lin28b are related RNA-binding proteins that inhibit the maturation of miRNAs of the let-7 family and participate in the control of cellular stemness and early embryonic development. Considerable interest has arisen recently concerning other physiological roles of the Lin28/let-7 axis, including its potential involvement in the control of puberty, as suggested by genome-wide association studies and functional genomics. We report herein the expression profiles of Lin28 and let-7 members in the rat hypothalamus during postnatal maturation and in selected models of altered puberty. The expression patterns of c-Myc (upstream positive regulator of Lin28), mir-145 (negative regulator of c-Myc), and mir-132 and mir-9 (putative miRNA repressors of Lin28, predicted by bioinformatic algorithms) were also explored. In male and female rats, Lin28, Lin28b, and c-Myc mRNAs displayed very high hypothalamic expression during the neonatal period, markedly decreased during the infantile-to-juvenile transition and reached minimal levels before/around puberty. A similar puberty-related decline was observed for Lin28b in monkey hypothalamus but not in the rat cortex, suggesting species conservation and tissue specificity. Conversely, let-7a, let-7b, mir-132, and mir-145, but not mir-9, showed opposite expression profiles. Perturbation of brain sex differentiation and puberty, by neonatal treatment with estrogen or androgen, altered the expression ratios of Lin28/let-7 at the time of puberty. Changes in the c-Myc/Lin28b/let-7 pathway were also detected in models of delayed puberty linked to early photoperiod manipulation and, to a lesser extent, postnatal underfeeding or chronic subnutrition. Altogether, our data are the first to document dramatic changes in the expression of the Lin28/let-7 axis in the rat hypothalamus during the postnatal maturation and after different manipulations that disturb puberty, thus suggesting the potential involvement of developmental changes in hypothalamic Lin28/let-7 expression in the mechanisms permitting/leading to puberty onset.


Asunto(s)
Envejecimiento/genética , Encéfalo/crecimiento & desarrollo , MicroARNs/metabolismo , Proteínas de Unión al ARN/biosíntesis , Animales , Células Madre Embrionarias/citología , Femenino , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Masculino , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Pubertad/efectos de los fármacos , Ratas , Ratas Wistar , Distribución Tisular
3.
Reprod Domest Anim ; 47 Suppl 4: 228-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22827375

RESUMEN

A non-surgical method to induce sterility would be a useful tool to control feral populations of animals. Our laboratories have experience with approaches aimed at targeting brain cells in vivo with vehicles that deliver a payload of either inhibitory RNAs or genes intended to correct cellular dysfunction. A combination/modification of these methods may provide a useful framework for the design of approaches that can be used to sterilize cats and dogs. For this approach to succeed, it has to meet several conditions: it needs to target a gene essential for fertility. It must involve a method that can selectively silence the gene of interest. It also needs to deliver the silencing agent via a minimally invasive method. Finally, the silencing effect needs to be sustained for many years, so that expansion of the targeted population can be effectively prevented. In this article, we discuss this subject and provide a succinct account of our previous experience with: (i) molecular reagents able to disrupt reproductive cyclicity when delivered to regions of the brain involved in the control of reproduction and (ii) molecular reagents able to ameliorate neuronal disease when delivered systemically using a novel approach of gene therapy.


Asunto(s)
Interferencia de ARN/fisiología , Esterilización Reproductiva/veterinaria , Adenoviridae , Animales , Gatos , Perros , Femenino , Fertilidad/fisiología , Vectores Genéticos , Hipotálamo/fisiología , Infertilidad Femenina , Masculino , MicroARNs , Regulación de la Población , Primates , Ratas , Esterilización Reproductiva/métodos
4.
Reprod Domest Anim ; 47 Suppl 6: 381-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23279544

RESUMEN

Population control of feral animals is often difficult, as it can be dangerous for the animals, labour intensive and expensive. Therefore, a useful tool for control of animal populations would be a non-surgical method to induce sterility. Our laboratories utilize methods aimed at targeting brain cells in vivo with vehicles that deliver a payload of either inhibitory RNAs or genes intended to correct cellular dysfunction. A useful framework for design of a new approach will be the combination of these methods with the intended goal to produce a technique that can be used to non-invasively sterilize cats and dogs. For this approach to succeed, it has to meet several conditions: the target gene must be essential for fertility; the method must include a mechanism to effectively and specifically silence the gene of interest; the method of delivering the silencing agent must be minimally invasive, and finally, the silencing effect must be sustained for the lifespan of the target species, so that expansion of the population can be effectively prevented. In this article, we discuss our work to develop gene silencing technology to induce sterility; we will use examples of our previous studies demonstrating that this approach is viable. These studies include (i) the use of viral vectors able to disrupt reproductive cyclicity when delivered to the regions of the brain involved in the control of reproduction and (ii) experiments with viral vectors that are able to ameliorate neuronal disease when delivered systemically using a novel approach of gene therapy.


Asunto(s)
Gatos , Anticoncepción/veterinaria , Perros , Silenciador del Gen/fisiología , Esterilización Reproductiva/veterinaria , Animales , Anticoncepción/métodos , Femenino , Fertilidad/fisiología , Hipotálamo/fisiología , Masculino , MicroARNs , Regulación de la Población , Esterilización Reproductiva/métodos
5.
J Clin Endocrinol Metab ; 86(1): 349-54, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11232023

RESUMEN

The presence of muscarinic receptors (MR) in the ovary of different species has been recognized, but the identity of these receptors as well as ovarian sources of their natural ligand, acetylcholine (ACh), have not been determined. Because luteinized human granulosa cells (GC) in culture express functional MR, we have determined whether the group of the related MR subtypes, M1R, M3R, and M5R, are present in vivo in human and rhesus monkey ovaries. To this end, ribonucleic acids (RNAs) of different human and monkey ovaries as well as RNAs from human GC and monkey oocytes were reverse transcribed and subjected to PCR amplification, followed by sequencing of the amplified complementary DNAs. Results obtained showed that M1R, M3R, and M5R messenger RNAs are present in adult human and monkey ovaries; oocytes express exclusively the M3R subtype, whereas GC express M1R and M5R. To determine the ovarian source(s) of the natural ligand of these ACh receptors, we attempted to localize the enzyme responsible for its synthesis with the help of a monoclonal antibody recognizing choline acetyltransferase for immunohistochemistry. In neither human nor monkey sections did we detect immunoreactive choline acetyltransferase-positive fibers or nerve cells, but, surprisingly, GC of antral follicles showed prominent staining. To determine whether GC can produce ACh, human cultured GC derived from preovulatory follicles were analyzed using a high pressure liquid chromatography technique. The results showed that these cells contained ACh in concentrations ranging from 4.2-11.5 pmol/10(6) cells. Samples of a rat granulosa cell line likewise contained ACh. Thus, the ovary contains multiple MR, and GC of antral follicles are able to synthesize ACh, the ligand of MR. We propose that ACh may serve as an as yet unrecognized factor involved in the complex regulation of ovarian function in the primate, e.g. regulation of cell proliferation or progesterone production.


Asunto(s)
Acetilcolina/biosíntesis , Ovario/metabolismo , Receptores Muscarínicos/metabolismo , Acetilcolina/metabolismo , Adulto , Animales , Encéfalo/metabolismo , Carnitina O-Acetiltransferasa/metabolismo , Células Cultivadas , Colina O-Acetiltransferasa/metabolismo , Femenino , Humanos , Inmunohistoquímica , Macaca mulatta , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores Muscarínicos/genética
6.
Endocrinology ; 141(12): 4736-50, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11108289

RESUMEN

TrkA, the nerve growth factor (NGF) tyrosine kinase receptor, is expressed not only in the nervous system, but also in nonneural cells, including discrete cellular subsets of the endocrine and immune system. In the rat ovary, trkA receptor abundance increases strikingly in thecal-interstitial cells during the hours preceding the first ovulation. Blockade of either trkA transducing capacity or NGF biological activity inhibited ovulation, suggesting a role for NGF in the ovulatory process of this species. To identify some of the processes that may be affected by trkA activation in the thecal compartment, we used purified thecal cells/thecal fibroblasts from bovine ovaries (heretofore referred to as thecal cells). Ribonuclease protection assays employing bovine-specific cRNA probes demonstrated the presence of the messenger RNAs (mRNAs) encoding NGF and its receptors, p75 NTR and trkA, in the thecal compartment of small, medium, and large antral follicles and showed that trkA mRNA is also expressed in granulosa cells. In situ hybridization and immunohistochemical examination of intact ovaries confirmed these cellular sites of NGF and trkA synthesis. TrkA mRNA, but not NGF mRNA, was lost within 48 h of placing thecal cells in culture. Thus, to study trkA-mediated actions of NGF on these cells we transiently expressed the receptor by transfection with a vector containing a full-length rat trkA complementary DNA under transcriptional control of the cytomegalovirus promoter. Because ovulation is preceded by an LH-dependent increase in androgen and progesterone production, the ability of NGF to modify the release of these steroids was determined in freshly plated cells still containing endogenous trkA receptors and in cells undergoing luteinization in culture that were transiently transfected with the trkA-encoding plasmid. NGF stimulated both androgen and progesterone release in freshly plated thecal cells, but not in luteinizing cells provided with trkA receptors. As ovulation in rodents requires an increased formation of PGE2 and has been shown to be antedated by proliferation of thecal fibroblasts, we determined the ability of NGF to affect these parameters in trkA-transfected thecal cells. The neurotrophin rapidly stimulated PGE2 release and amplified the early steroidal response to hCG in trkA-expressing cells, but not in cells lacking the receptor. Likewise, NGF stimulated [3H]thymidine incorporation into trkA-containing cells, but not into cells that had lost the receptor in culture. Induction of ovulation in immature rats by gonadotropin treatment verified that an increased cell proliferation in the thecal compartment, determined by the incorporation of bromodeoxyuridine into cell nuclei, occurs 4-5 h before ovulation in this species. These results suggest that the contribution of NGF to the ovulatory process includes a stimulatory effect of the neurotrophin on steroidogenesis, PGE2 formation, and proliferative activity of thecal compartment cells.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Folículo Ovárico/citología , Células Tecales/efectos de los fármacos , Androstenodiona/metabolismo , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Gonadotropina Coriónica/farmacología , Ciclooxigenasa 2 , Dinoprostona/metabolismo , Femenino , Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Isoenzimas/genética , Factor de Crecimiento Nervioso/genética , Ovulación/efectos de los fármacos , Progesterona/metabolismo , Prostaglandina-Endoperóxido Sintasas/genética , Sondas ARN , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkA/genética , Receptor trkA/fisiología , Receptores de Factor de Crecimiento Nervioso/genética , Ovinos , Células Tecales/citología , Células Tecales/fisiología
7.
J Neurosci ; 15(6): 4223-37, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7790907

RESUMEN

Hypothalamic neurons control a variety of important hormonal and behavioral functions. Little is known, however, about the neurotrophic factors that these neurons may require for survival and/or maintenance of their differentiated functions. We conducted experiments to examine this issue, utilizing a combination of immunohistochemical, in situ hybridization and cell culture approaches. We found that the low affinity receptor for nerve growth factor (p75 NGFR) is present in small subsets of hypothalamic peptidergic neurons identified as such by their content of galanin, luteinizing hormone-releasing hormone (LHRH) and vasointestinal peptide (VIP). More prominently, however, examination of hypothalamic dopaminergic (DA) neurons for the presence of p75 NGFR-like immunoreactivity revealed that the receptor was present on tyrosine hydroxylase (TH)-positive neurons of the zona incerta and periventricular region, but not on neuroendocrine DA neurons of the tuberoinfundibular region. In situ hybridization experiments using a p75 NGFR cRNA confirmed this distribution. Regardless of the presence or absence of p75 NGFR, neither DA group expresses trkA mRNA, indicating that these two major hypothalamic subsets of DNA neurons are NGF-insensitive. A substantial fraction of TH mRNA-positive cells in the zona incerta expresses trkB mRNA, which encodes the receptor for brain derived neurotrophic factor (BDNF); in turn BDNF supports the in vitro survival of hypothalamic TH neurons bearing p75-NGFR, suggesting that BDNF is trophic for DNA neurons of the zona incerta. In contrast, tuberoinfundibular DA neurons do not express trkB mRNA, but some have trkC mRNA, which encodes the receptor for neurotrophin-3 (NT-3). The in vitro survival of TH neurons devoid of p75-NGFR is supported by NT-3, implying that NT-3 may be trophic for a subset of tuberoinfundibular DA neurons. These results suggest that, in spite of expressing an identical neurotransmitter phenotype, anatomically and functionally segregated DA neurons of the neurodendocrine brain are sustained by different neurotrophic factors.


Asunto(s)
Hipotálamo/anatomía & histología , Hipotálamo/fisiología , Neuronas/citología , Neuronas/fisiología , Sistemas Neurosecretores/anatomía & histología , Sistemas Neurosecretores/fisiología , Prosencéfalo/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Animales , Anticuerpos Monoclonales , Femenino , Expresión Génica , Inmunohistoquímica , Especificidad de Órganos , Prosencéfalo/anatomía & histología , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor trkA , Receptores de Factor de Crecimiento Nervioso/análisis , Receptores de Factor de Crecimiento Nervioso/metabolismo
8.
Endocrinology ; 135(4): 1392-400, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7925101

RESUMEN

The importance of transforming growth factor-alpha (TGF alpha) in female reproductive development was assessed using transgenic mice bearing a human TGF alpha complementary DNA under the control of a mouse metallothionein-1 promoter (MT1-hTGF alpha). Examination of the brain and ovaries 5 h after a single sc injection of zinc chloride, administered to activate the MT1-hTGF alpha transgene, revealed that prominent sites of human TGF alpha messenger RNA expression within these tissues were the hypothalamus and ovarian follicles, respectively. In vitro experiments showed that acute transgene activation increased hypothalamic release of LH-releasing hormone. In contrast, the ovarian steroidal response to gonadotropins, examined in vitro, was markedly attenuated. Chronic activation of transgene expression by daily administration of zinc chloride delayed the time of first estrus (an index of peripubertal estrogen secretion), but shortened the interval between first estrus and the onset of estrous cyclicity (an index of reproductive competence). Accumulation of small antral follicles, accompanied by thecal hypertrophy and enhanced androgen production, preceded the acquisition of ovulatory capacity. These changes were accompanied by reduced serum LH levels, suggesting that the relative inability of small antral follicles to develop further in TGF alpha-overexpressing mice is at least in part due to inappropriate gonadotropin support. Serum LH levels in these animals may be reduced by an augmented androgen negative feedback signal. Nontransgenic mouse ovaries, placed under the control of a transgenic hypothalamus by heterologous grafting, rapidly ovulated and initiated estrous cyclicity. In contrast, acquisition of reproductive capacity was severely delayed in nontransgenic mice bearing transgenic ovarian grafts. The results indicate that TGF alpha regulates female reproductive development through two opposing mechanisms: within the brain, it facilitates the neuroendocrine activation of the process; at the ovarian level, modulates the stimulatory effect of gonadotropin hormones on follicular growth and steroidogenesis.


Asunto(s)
Maduración Sexual/fisiología , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/fisiología , Animales , ADN/análisis , ADN/genética , Femenino , Regulación de la Expresión Génica , Hipotálamo/química , Hipotálamo/fisiología , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Transgénicos , Ovario/química , Ovario/fisiología , Caracteres Sexuales , Factor de Crecimiento Transformador alfa/análisis
9.
Front Neuroendocrinol ; 13(2): 120-62, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1468600

RESUMEN

The concept is proposed that polypeptide neurotrophic factors contribute to the developmental regulation of ovarian and hypothalamic function in mammals. Nerve growth factor (NGF) and neurotrophin-3, two members of the neurotrophin family, have been identified in the rat ovary and one of its receptors has been localized to the innervation and thecal cells of developing follicles. Although NGF supports the sympathetic innervation of the gland, the extent to which follicles are innervated appears to be defined by the differential expression of NGF receptors in the theca of developing follicles. The presence of NGF receptors in steroid-producing cells suggests a direct involvement of neurotrophins in the regulation of gonadal endocrine function. Evidence is beginning to emerge suggesting that development of the reproductive hypothalamus is affected by insulin-like growth factor 1 secreted by peripheral tissues, and transforming growth factor alpha (TGF alpha) produced locally. In the rat hypothalamus, TGF alpha appears to be synthesized in both neurons and glial cells. In glial cells it may interact with epidermal growth factor (EGF) receptors to further enhance TGF alpha synthesis and to, perhaps, stimulate eicosanoid formation. In turn, one of these eicosanoids, prostaglandin E2, may act on luteinizing hormone-releasing hormone (LHRH) neurons to stimulate the release of LHRH in a genomic-independent manner. This provides the basis for the notion that during development LHRH secretion is regulated by a dual mechanism, one that involves transsynaptic effects exerted by neurotransmitters, the other that requires a glial-neuronal interaction and that may predominantly regulate release of the neuropeptide. An increased expression of the TGF alpha and EGF receptor genes in reactive astrocytes is postulated to contribute to the process by which hypothalamic injury causes sexual precocity. Morphological maturation of the reproductive hypothalamus is thought to occur during sexual development. The process is accelerated by estradiol, which exerts its neurotrophic effects by enhancing the expression of genes encoding cytoskeletal proteins involved in neuronal development and regeneration. It is suggested that acquisition of functional competence by both the ovaries and the reproductive hypothalamus requires the participation of specific, but not similar, neurotrophic factors. The relevance of these concepts to the process of sexual development in other species, particularly primates, remains to be defined.


Asunto(s)
Hipotálamo/crecimiento & desarrollo , Familia de Multigenes/genética , Factores de Crecimiento Nervioso/fisiología , Ovario/crecimiento & desarrollo , Maduración Sexual/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Neuroglía/fisiología , Neuronas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA