Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 26(7): 1038-45, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16964292

RESUMO

The transition from basic to clinical cancer research for a number of experimental therapeutics is hampered by the lack of a genetically malleable, large animal model. To this end, we genetically engineered primary porcine cells to be tumorigenic by expression of proteins known to perturb pathways commonly corrupted in human cancer. Akin to human cells, these porcine cells were quite resistant to transformation, requiring multiple genetic changes. Moreover, the transformed porcine cells produced tumors when returned to the isogenic host animal. The ability to now rapidly and reproducibly genetically induce tumors of sizes similar to those treated clinically in a large mammal similar to humans in many respects will provide a robust cancer model for preclinical studies dependent on generating large tumors.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Experimentais/genética , Suínos/genética , Animais , Linhagem Celular , Linhagem Celular Transformada , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Engenharia Genética/métodos , Camundongos , Camundongos SCID , Neoplasias Experimentais/etiologia , Neoplasias Experimentais/patologia
2.
J Exp Med ; 194(8): 1043-52, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11602635

RESUMO

T cells are activated by binding of the T cell receptor (TCR) to a peptide-major histocompatibility complex (MHC) complex (pMHC) expressed on the surface of antigen presenting cells. Various models have predicted that activation is limited to a narrow window of affinities (or dissociation rates) for the TCR-pMHC interaction and that above or below this window, T cells will fail to undergo activation. However, to date there have not been TCRs with sufficiently high affinities in order to test this hypothesis. In this report we examined the activity of a CD8-negative T cell line transfected with a high affinity mutant TCR (K(D) = 10 nM) derived from cytotoxic T lymphocyte clone 2C by in vitro engineering. The results show that despite a 300-fold higher affinity and a 45-fold longer off-rate compared with the wild-type TCR, T cells that expressed the mutant TCRs were activated by peptide. In fact, activation could be detected at significantly lower peptide concentrations than with T cells that expressed the wild-type TCR. Furthermore, binding and functional analyses of a panel of peptide variants suggested that pMHC stability could account for apparent discrepancies between TCR affinity and T cell activity observed in several prior studies.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD8/imunologia , Complexo Cetoglutarato Desidrogenase/imunologia , Ativação Linfocitária/imunologia , Oligopeptídeos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/imunologia , Interleucina-2/biossíntese , Complexo Principal de Histocompatibilidade/imunologia , Mutagênese , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Transfecção
3.
J Immunol ; 165(12): 7293-9, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11120864

RESUMO

A number of recent studies have indicated that T cells can be stimulated to attack transplanted brain tumors in rodent models. As IL-12 has been shown to activate cytotoxic T cell responses, we tested the idea that it might stimulate a T cell response against endogenous brain tumors that arise in SV40 large T Ag transgenic mice (SV11). SV11 mice develop tumors of the choroid plexus, a specialization of the ependymal lining of the brain ventricles. They are a particularly relevant model of human disease, because they are immunocompetent but immunologically tolerant of the tumors. SV11 mice were treated with recombinant murine IL-12 for 10 days. Tumors grew more slowly than in control treated mice, and in some cases were reduced in size, as assessed by magnetic resonance imaging before and after treatment. At the end of treatment, tumors, but not brain parenchyma, exhibited extensive infiltration of activated CD8(+) and CD4(+) T cells. Tumors also showed a reduction in vascular density. Mice treated with IL-12 lived significantly longer than control mice. Tumors that progressed were nearly devoid of T cells, indicating that the T cell response was not sustained. In addition, some mice that had a substantial tumor burden at the beginning of treatment displayed evidence of immunosuppression, which might be related to TGF-ss2 detected in tumors. We conclude that IL-12 treatment can initiate an anti-tumor response even against endogenously arising brain tumors, but factors that will allow a sustained and more effective anti-tumor response need to be determined.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Neoplasias do Plexo Corióideo/imunologia , Neoplasias do Plexo Corióideo/terapia , Interleucina-12/administração & dosagem , Animais , Barreira Hematoencefálica/imunologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/fisiopatologia , Neoplasias do Plexo Corióideo/irrigação sanguínea , Neoplasias do Plexo Corióideo/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Hipertrofia , Imuno-Histoquímica , Injeções Intraperitoneais , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Baço/patologia , Análise de Sobrevida , Fator de Crescimento Transformador beta/biossíntese
4.
Int J Cancer ; 83(1): 141-9, 1999 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-10449621

RESUMO

A variety of immunological approaches to cancer treatment are currently being explored. These include strategies designed to enhance or redirect the activity of T cells against tumors. Bispecific antibodies comprise a class of agents capable of redirecting T cells by binding to a tumor antigen and the T-cell receptor (TCR). In vivo pre-clinical testing of bispecific antibodies against human tumors has to date been limited to the use of immunodeficient mice that receive the bispecific agent, activated human effector T cells, and human tumor cells. In this report, we show that TCR transgenic/RAG-1 knockout mice (TCR/RAG) serve as a unique model allowing endogenous T cells to be redirected against transplanted human tumors. The findings show that TCR/RAG mice (i) accepted transplants of human tumors, including the folate-receptor-positive tumor line KB; (ii) contained endogenous cytotoxic T lymphocytes that could be activated in vivo with an antigenic peptide recognized by the transgenic TCR; (iii) rejected human tumors after treatment with the activating peptide and bispecific agents that contained folic acid co-valently linked to an anti-TCR antibody. Successful rejection was achieved with folate conjugates of Fab or scFv fragments. Treatment with activating agents and bispecific conjugates resulted in the complete eradication of freshly transplanted tumors as well as significantly prolonging the survival of mice bearing established solid tumors. Our results highlight the importance of including T-cell-activating modalities in combination with bispecific antibodies. Additionally, we introduce a system that allows endogenous T cells to be redirected against human tumor xenografts and in which the T cells may be followed in vivo by use of a clonotypic marker.


Assuntos
Anticorpos Biespecíficos/imunologia , Proteínas de Transporte/imunologia , Receptores de Superfície Celular , Linfócitos T/metabolismo , Animais , Vacinas Anticâncer , Carcinoma de Células Escamosas/imunologia , Relação Dose-Resposta Imunológica , Citometria de Fluxo , Receptores de Folato com Âncoras de GPI , Genes RAG-1/imunologia , Genes Codificadores dos Receptores de Linfócitos T/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
5.
J Control Release ; 53(1-3): 77-84, 1998 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-9741915

RESUMO

It has been known for some time that mammalian immune systems are capable of eliminating large tumor burdens. Redirecting the immune response of a patient to an established tumor has now become the focus of various therapeutic strategies. In this report, two projects toward this goal are described. The first project involves the development of a transgenic mouse model for T cell directed therapeutics. These mice express specific T cell receptor alpha and beta transgenes on a background in which the recombinational-activating-gene-1 (RAG) has been knocked out. The mice express cytotoxic T cells but not either T helper cells or B cells. Despite these deficiencies, the animals are capable of eliminating tumors that express the appropriate peptide/major histocompatibility complex ligand that is recognized by the alphabeta transgenic T cell receptor. Human tumors grow as transplants in these mice, thereby allowing various agents that redirect the endogenous T cells against human tumors to be tested. The second project involves a description of such agents: bispecific antibodies that simultaneously bind to an immune effector cell and a tumor cell. The bispecific antibody described here consists of folate attached to anti-T cell receptor antibodies, or their fragments. A single-chain Fv coupled with folate can redirect the lysis of human tumor cells that bear the high affinity folate receptor. Preliminary in vivo data showed that the folate/antibody conjugates were also capable of mediating rejection of the human tumor. This transgenic mouse model should now allow the evaluation and optimization of bispecific agents that can redirect a patient's own T cell response.


Assuntos
Anticorpos Biespecíficos/imunologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Biespecíficos/administração & dosagem , Modelos Animais de Doenças , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Humanos , Imunoterapia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neoplasias/terapia , Células Tumorais Cultivadas
6.
Int J Cancer ; 76(5): 761-6, 1998 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-9610737

RESUMO

High-affinity receptors expressed on the surface of some tumors can be exploited by chemically conjugating the ligand for the receptor and an antibody against immune effector cells, thus redirecting their cytolytic potential against the tumor. Ovarian carcinomas and some brain tumors express the high-affinity folate receptor (FR). In this report, a transgenic mouse model that generates endogenously arising choroid plexus tumors was used to show that folate/anti-T-cell receptor antibody conjugates can direct infiltration of T cells into solid brain tumor masses. An engineered single-chain Fv form of the anti-T-cell receptor antibody KJ16 was conjugated with folate, to produce a bispecific agent that was substantially smaller than most previously characterized bispecific antibodies. Folate conjugation to the antibody increased T-cell infiltration into the tumors by 10- to 20-fold, and significantly prolonged survival of the mice.


Assuntos
Neoplasias do Plexo Corióideo/metabolismo , Neoplasias do Plexo Corióideo/terapia , Imunoconjugados/farmacologia , Linfócitos do Interstício Tumoral/imunologia , Receptores de Superfície Celular , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Antineoplásicos/farmacologia , Antígenos Transformantes de Poliomavirus/imunologia , Proteínas de Transporte/metabolismo , Neoplasias do Plexo Corióideo/ultraestrutura , Feminino , Citometria de Fluxo , Receptores de Folato com Âncoras de GPI , Ácido Fólico/metabolismo , Ácido Fólico/farmacologia , Imunoconjugados/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia
7.
J Immunol ; 159(10): 4665-75, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9366389

RESUMO

Three sources of help for the development of a CD8+ CTL response have been described: the CD4+ direct and indirect pathways and the CD8+ direct pathway. In an effort to understand the minimal requirements for the development of a CTL response in vivo, we have bred mice transgenic for the 2C TCR onto a RAG(-/-) background. The 2C T cells in this animal are exclusively CD8+ CTLs of a single specificity, and they exhibit altered thymic maturation compared with that of T cells from 2C TCR/RAG(+/+) mice. T cells from 2C TCR/RAG(-/-) mice can be activated to a high level in vivo by administration of a self-MHC-restricted antigenic peptide. The 2C TCR/RAG(-/-) mice are able to reject B7-negative allogeneic tumors bearing the appropriate peptide/MHC ligand p2C/Ld. These mice fail to reject syngeneic tumors, and their RAG(-/-) littermates lacking 2C T cells uniformly succumb to both allogeneic and syngeneic tumors. Moreover, blockade of B7 costimulatory molecules fails to prevent tumor rejection in the 2C TCR/RAG(-/-) mice, suggesting that allorejection is occurring independently of B7-mediated costimulation as well as in the absence of CD4+ T cells. CTLs isolated from the site of the tumor during the period of rejection express the activation marker CD25 and are able to mediate ex vivo cytolysis of tumor cells bearing the appropriate Ag. These results suggest that in this TCR transgenic model with a very high precursor frequency, CTL development can occur in the absence of B7:CD28 costimulation and without CD4+ help.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica/genética , Genes RAG-1 , Rejeição de Enxerto/genética , Isoantígenos/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Sarcoma Experimental/imunologia , Animais , Antígeno B7-1/fisiologia , Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/imunologia , Isoantígenos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Recidiva Local de Neoplasia , Transplante de Neoplasias , Receptores de Antígenos de Linfócitos T/imunologia , Sarcoma Experimental/genética , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Evasão Tumoral/imunologia
8.
Nature ; 386(6627): 838-42, 1997 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-9126743

RESUMO

Migration of neurons from proliferative zones to their functional sites is fundamental to the normal development of the central nervous system. Mice homozygous for the spontaneous rostral cerebellar malformation mutation (rcm(s)) or a newly identified transgenic insertion allele (rcm(tg)) exhibit cerebellar and midbrain defects, apparently as a result of abnormal neuronal migration. Laminar structure abnormalities in lateral regions of the rostral cerebellar cortex have been described in homozygous rcm(s) mice. We now demonstrate that the cerebellum of both rcm(s) and rcm(tg) homozygotes is smaller and has fewer folia than in the wild-type, ectopic cerebellar cells are present in midbrain regions by three days after birth, and there are abnormalities in postnatal cerebellar neuronal migration. We have cloned the rcm complementary DNA, which encodes a transmembrane receptor of the immunoglobulin superfamily. The sequence of the rcm protein (Rcm) is highly similar to that of UNC-5, a Caenorhabditis elegans protein that is essential for dorsal guidance of pioneer axons and for the movement of cells away from the netrin ligand, which is encoded by the unc-6 gene. As Rcm is a member of a newly described family of vertebrate homologues of UNC-5 which are netrin-binding proteins, our results indicate that UNC-5-like proteins may have a conserved function in mediating netrin-guided migration.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Helminto/química , Proteínas de Membrana/química , Receptores de Superfície Celular , Receptores de Fatores de Crescimento/química , Receptores de Fator de Crescimento Neural/genética , Sequência de Aminoácidos , Animais , Northern Blotting , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Cerebelo/anormalidades , Cerebelo/embriologia , Cerebelo/metabolismo , Clonagem Molecular , Expressão Gênica , Homozigoto , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Fatores de Crescimento Neural/metabolismo , Receptores de Netrina , Netrina-1 , Neurônios/fisiologia , Reação em Cadeia da Polimerase , Receptores de Fator de Crescimento Neural/química , Receptores de Fator de Crescimento Neural/fisiologia , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Proteínas Supressoras de Tumor
9.
Neuroendocrinology ; 56(3): 436-44, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1436381

RESUMO

Deficiency in the secretion of luteinizing hormone-releasing hormone (LHRH) from the median eminence (ME) is one of the factors limiting reinitiation of estrous cycles following parturition in cows. In previous studies, administration of naloxone, an opioid receptor antagonist, to postpartum cows increased LH secretion, suggesting that endogenous opioids inhibit the secretion of LHRH. This study employs quantitative light microscopy to describe morphological changes in the distribution of immunoreactive beta-endorphin (ir-beta-END) neurons in the hypothalamus of anestrous early postpartum (EPP, days 10-16, n = 5), midpostpartum (MPP, days 33-43, n = 4) and multiparous cycling cows (CYC, months 12-14, n = 4). Cryostat sections (60 microns) of perfusion-fixed ventral diencephalon and forebrain were immunostained with anti-beta-END serum via the biotin-avidin-peroxidase method or double stained sequentially with anti-LHRH serum, then anti-beta-END serum. In all cows, beta-END immunoreactive perikarya, mostly bipolar neurons, were located in the arcuate and periarcuate nucleus (ARC), with some perikarya in the ME. Within the ARC, the percentage area immunostained for ir-beta-END was greater (p < 0.01) for the CYC than EPP cows, with MPP intermediate but not significantly different from the other groups. Consistent for all cows, the percentage area of ir-beta-END in ventral ARC regions was greater (p < 0.05) than dorsal ARC regions. Fibers from these neurons coursed into the anterior hypothalamus, preoptic area and bed nucleus of stria terminalis. Ventrally projecting fibers entered the ME forming a densely staining band within the external layer.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Anestro/metabolismo , Núcleo Arqueado do Hipotálamo/química , Fase Luteal/metabolismo , Eminência Mediana/química , Período Pós-Parto/metabolismo , beta-Endorfina/análise , Animais , Núcleo Arqueado do Hipotálamo/citologia , Bovinos , Estrogênios/sangue , Feminino , Hormônio Liberador de Gonadotropina/análise , Técnicas Imunoenzimáticas , Eminência Mediana/citologia , Neurônios/química , Ovário/anatomia & histologia , Progesterona/sangue
10.
Theriogenology ; 38(1): 73-83, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16727119

RESUMO

The response of serum luteinizing hormone (LH) to morphine, naloxone and gonadotropin-releasing hormone (GnRH) in ovariectomized, suckled (n=4) and nonsuckled (n=3) cows was investigated. Six months after ovariectomy and calf removal, the cows were challenged with 1mg, i.v. naloxone/kg body weight and 1 mg i.v. morphine/kg body weight in a crossover design; blood was collected at 15-minute intervals for 7 hours over a 3-day period. To evaluate LH secretion and pituitary responsiveness, 5 microg of GnRH were administered at Hour 6 on Day 1. On Days 2 and 3, naloxone or morphine was administered at Hour 3, followed by GnRH (5 microg/animal) at Hour 6. Mean preinjection LH concentrations (3.6 +/- 0.2 and 4.7 +/- 0.2 ng/ml), LH pulse frequency (0.6 +/- 0.1 and 0.8 +/- 0.1 pulses/hour) and LH pulse amplitude (2.9 +/- 0.5 and 2.9 +/- 0.6 ng/ml) were similar for suckled and nonsuckled cows, respectively. Morphine decreased (P < 0.01) mean serum LH concentrations (pretreatment 4.2 +/- 0.2 vs post-treatment 2.2 +/- 0.2 ng/ml) in both suckled and nonsuckled cows; however, mean serum LH concentrations remained unchanged after naloxone. Nonsuckled cows had a greater (P < 0.001) LH response to GnRH than did suckled cows (area of response curve: 1004 +/- 92 vs 434 +/- 75 arbitrary units). We suggest that opioid receptors are functionally linked to the GnRH secretory system in suckled and nonsuckled cows that had been ovariectomized for a long period of time. However, gonadotropin secretion appears not to be regulated by opioid mechanisms, and suckling inhibits pituitary responsiveness to GnRH in this model.

11.
Neuroendocrinology ; 55(4): 380-9, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1565205

RESUMO

Deficiency in secretion of luteinizing hormone releasing hormone (LHRH) from the median eminence (ME) is one of the factors limiting reinitiation of estrous cycles following parturition in cows. This study employs quantitative light microscopy to describe morphological changes in LHRH neurons obtained from cows at three postpartum times. Tissues were obtained from anestrous early postpartum (EPP; days 10-16, n = 5), midpostpartum (MPP; days 33-43, n = 4), and multiparous cycling (CYC; months 12-14, n = 4) cows. Following perfusion fixation, cryostat sections (60 microns) of the ventral forebrain were immunostained with anti-LHRH serum via the biotin-avidin-peroxidase method. In all cows, LHRH perikarya formed a loosely arranged continuum, extending from anterior to posterior within the diagonal band of Broca, the lateral and medical preoptic areas, and the anterior hypothalamus. Width and length of perikarya were similar between postpartum groups. The number of dendrite-like processes per neuron (p less than 0.01) and average or total length of process per neuron (p less than 0.001) were greater in the CYC than in the EPP and MPP cows. Although all groups of cows contained a spectrum of lengths of dendrite-like processes, both EPP and MPP cows obtained a greater (p less than 0.05) percentage of neurons with shorter processes and a smaller percentage of neurons with longer processes. Within the ME, the percentage area occupied by immunostained fibers was less (p less than 0.05) in EPP cows than in MPP or CYC cows.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Estro/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Neurônios/ultraestrutura , Período Pós-Parto/fisiologia , Área Pré-Óptica/citologia , Animais , Bovinos , Dendritos/ultraestrutura , Feminino , Hormônio Liberador de Gonadotropina/análise , Imuno-Histoquímica , Neurônios/fisiologia
12.
J Anim Sci ; 69(9): 3733-46, 1991 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1657852

RESUMO

Autoradiography was used to quantify opioid receptors in the median eminence (ME) and preoptic area (POA) of the brain of eight heifers, and in vitro perifusion of ME and POA tissue from seven cows and heifers was used to examine the release of LHRH after administration of naloxone (NAL). For quantitative receptor autoradiography, [3H]NAL was used as the radioligand and NAL or morphine as competitors. Specific binding of [3H]NAL in POA and ME resulted in linear Scatchard plots with similar equilibrium dissociation constants (Kd = 4.2 +/- 1.1 nM) and mean binding site densities in the POA and ME (POA: 80.3 +/- 5.8; ME 67.5 +/- 8.0 fmol/mm2). There were no differences between mean binding site densities of zonas externa and interna of the ME; however, between various regions of the POA within individual animals, binding site densities varied threefold (47.6 to 165.1 fmol/mm2). During in vitro perifusions of isolated POA and ME, basal LHRH secretion from ME decreased (P less than .001) from 15.9 +/- 1.8 to 7.3 +/- .8 pg/10 min fraction (500 microliters) but remained constant for POA (3.1 +/- .4 pg/fraction). Injections of medium alone did not affect LHRH secretion. Although there was no significant dose (10(-9) to 10(-7) M) effect, NAL increased (P less than .05) LHRH efflux from the ME and POA when administered at 110 min from the initiation of perfusion and again at 200 min for ME but not for POA. All tissues responded to KCl (30 mM) administered at 290 min of perifusion with increased (P less than .001) LHRH efflux. Both immunoreactive-LHRH and immunoreactive-beta-endorphin were immunocytochemically localized in neurons from some of these perifused tissues. We suggest that endogenous opioids suppress LHRH secretion by actions on specific opioid receptors located within the POA and ME of the brain.


Assuntos
Bovinos/fisiologia , Endorfinas/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/fisiologia , Área Pré-Óptica/fisiologia , Animais , Autorradiografia , Sítios de Ligação , Ligação Competitiva , Técnicas de Cultura , Feminino , Imuno-Histoquímica , Morfina/farmacologia , Naloxona/farmacologia , Receptores Opioides/análise
13.
J Anim Sci ; 68(6): 1656-65, 1990 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2384365

RESUMO

These studies examined responses of serum prolactin (PRL) and growth hormone (GH) to opioid agonist and antagonist administration in heifers. To minimize nonspecific and behavioral effects and to facilitate future studies with specific opioid receptor agonists, a cannula was placed within the third cerebral ventricle of the brain of 4- to 10-mo-old heifers to directly access hypothalamic regions involved in the regulation of PRL and GH secretion. Increasing doses of morphine (M) from 2 to 1,500 micrograms injected into the third cerebral ventricle increased (P less than .001) serum PRL concentrations in a dose-related manner. Growth hormone responses were variable, resulting in elevated (P less than .05) serum concentrations following morphine, but no dose-related effects were apparent. Both PRL and GH responses to 700 micrograms M were absent when an intracerebral ventricle injection of an equimolar dose of naloxone, an opioid receptor antagonist, was administered prior to M. In a replicated 4 x 4 latin square, the effects of intravenous naloxone on PRL and GH responses was tested in young (86 +/- 11 d) and older (234 +/- 6 d) heifers. Naloxone at doses of 1, 2 and 4 mg/kg reduced (P less than .05) serum concentrations of PRL for 45 to 60 min. Mean concentrations of GH tended to be higher (P less than .07) in older heifers All doses of naloxone decreased (P less than .05) serum GH concentrations in older heifers but proved ineffective in younger heifers. There were no differences between doses of naloxone on either PRL or GH. These data suggest that endogenous opioids are involved in the regulation of PRL and GH secretion in heifers.


Assuntos
Bovinos/metabolismo , Hormônio do Crescimento/sangue , Morfina/farmacologia , Naloxona/farmacologia , Prolactina/sangue , Animais , Bovinos/sangue , Relação Dose-Resposta a Droga , Feminino , Hormônio do Crescimento/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraventriculares/veterinária , Morfina/administração & dosagem , Prolactina/metabolismo
14.
Biol Reprod ; 42(4): 619-24, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2346772

RESUMO

The response of serum luteinizing hormone (LH) to naloxone, an opiate antagonist, and gonadotropin-releasing hormone (GnRH) was measured in cows in late pregnancy to assess opioid inhibition of LH. Blood samples were collected at 15-min intervals for 7 h. In a Latin Square arrangement, each cow (n = 6) received naloxone (0, 0.5, and 1.0 mg/kg BW, i.v.; 2 cows each) at Hour 2 on 3 consecutive days (9 +/- 2 days prepartum). GnRH (7 ng/kg body weight, i.v.) was administered at Hour 5 to all cows on each day. Mean serum LH concentrations (x +/- SE) before naloxone injection were similar (0.4 +/- 0.1 ng/ml), with no serum LH pulses observed during the experiment. Mean serum LH concentrations post-naloxone were similar (0.4 +/- 0.1 ng/ml) to concentrations pre-naloxone. Mean serum LH concentrations increased (p less than 0.05) following GnRH administration (7 ng/kg) and did not differ among cows receiving different dosages of naloxone (0 mg/kg, 1.44 +/- 0.20; 0.5 mg/kg, 1.0 +/- 0.1; 1.0 mg/kg, 0.9 +/- 0.1 ng/ml). In Experiment 2, LH response to naloxone and GnRH was measured in 12 ovariectomized cows on Day 19 of estrogen and progesterone treatment (5 micrograms/kg BW estrogen: 0.2 mg/kg BW progesterone) and on Days 7 and 14 after steroid treatment. On Day 19, naloxone failed to increase serum LH concentrations (Pre: 0.4 +/- 0.1; Post: 0.4 +/- 0.1 ng/ml) after 0, 0.5, or 1.0 mg/kg BW.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Hormônio Luteinizante/metabolismo , Naloxona/farmacologia , Ovariectomia , Prenhez , Análise de Variância , Animais , Bovinos , Estradiol/sangue , Feminino , Ovário/fisiologia , Gravidez , Progesterona/sangue
15.
Endocrinology ; 126(1): 622-30, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2152875

RESUMO

Insulin-like growth factor-I (IGF-I) is a mitogenic polypeptide postulated to mediate the effect of GH on adipose tissue development. To determine if the effect of GH could be mediated by the local production of IGF-I, we have characterized IGF-I RNA expression, polypeptide secretion, and binding protein activity in primary preadipocyte cultures derived from porcine adipose tissues. GH acutely regulated the abundance of multiple IGF-I RNA transcripts and resulted in a 2-fold increase in secreted immunoreactive IGF-I (iIGF-I) polypeptide in medium conditioned for 48 h by preadipocyte cultures relative to those not receiving GH. Immunocytochemical data indicated that IGF-I is synthesized by presumptive and mature adipocytes. The effect of GH on iIGF-I secretion was observed in cultures derived from both fetal and postnatal animals, while secreted IGF-binding protein activity was increased due to GH only in cultures from fetal animals. The increase in local IGF-I production in response to GH was associated with a decrease in adipocyte development, suggesting that local IGF-I may contribute to suppression of differentiated phenotype.


Assuntos
Tecido Adiposo/metabolismo , Fator de Crescimento Insulin-Like I/genética , Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Somatomedinas/genética , Tecido Adiposo/citologia , Animais , Ligação Competitiva , Northern Blotting , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura , Hormônio do Crescimento/metabolismo , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/metabolismo , RNA Mensageiro/genética , Radioimunoensaio , Receptores de Somatomedina , Suínos , Transcrição Gênica
16.
Biol Reprod ; 41(3): 431-7, 1989 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2590714

RESUMO

In three experiments, we examined endogenous opioid inhibition of luteinizing hormone (LH) secretion during the bovine estrous cycle. An increase in serum LH in response to the opioid antagonist naloxone (Na; 1 mg/kg i.v.) was the criterion for opioid inhibition. Estrous cycles were synchronized via prostaglandin administration. In Experiment 1, mean serum LH was not different during the luteal phase in yearling heifers (n = 6/group) at Hour 1 after Nal (2.1 ng/ml) compared to controls (1.8 ng/ml). However, LH peak amplitude was increased (p less than 0.05) in the Nal compared to the control group. Serum LH was increased (p less than 0.01) during the follicular phase in heifers at Hour 1 post-Nal compared to controls (4.7 and 3.5 ng/ml, respectively). Again, Nal administration was followed by increased (p less than 0.05) LH pulse amplitude compared to control. In Experiment 2, no effect of Nal upon serum LH was detected in cows (n = 9) during proestrus, metestrus, midluteal and late luteal portions of the estrous cycle. In Experiment 3, the LH response to Nal was examined simultaneously in yearling heifers and cows (n = 5/group) during the luteal and follicular phases. Serum LH increased (p less than 0.001) during Hour 1 post-Nal in heifers compared to cows during the follicular (3.4 vs. 1.7 ng/ml) but not during the luteal phase. LH pulse amplitude also increased (p less than 0.05) during Hour 1 post-Nal in heifers compared to cows during the luteal (2.5 vs. 1.1 ng/nl and follicular (2.5 vs. 1.3 ng/ml) phases.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Bovinos/fisiologia , Estro/fisiologia , Hormônio Luteinizante/metabolismo , Naloxona/farmacologia , Animais , Feminino , Fase Folicular/fisiologia , Fase Luteal/fisiologia , Prostaglandinas/farmacologia , Fatores de Tempo
17.
J Anim Sci ; 67(6): 1527-31, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2670869

RESUMO

The influence of the suckling stimulus and ovarian secretions on LH response to naloxone was studied in 16 postpartum anestrous beef cows that were assigned randomly to one of four groups (n = 4/group): intact suckled (IS), intact nonsuckled (IN), ovariectomized suckled (OS) or ovariectomized nonsuckled (ON). Ovariectomy (OS + ON) and calf removal (IN + ON) were performed on d 2, 3 or 4 after parturition. Jugular venous blood was collected at 15-min intervals for 4 h before and 4 h after administration of naloxone (1 mg/kg BW, i.v.) on d 14 and d 28 after parturition. Gonadotropin-releasing hormone (5 micrograms, i.v.) was given 3 h after naloxone. Both IN and OS increased (P less than .05) mean pretreatment LH above IS values (mean +/- SE, ng/ml; IS 1.6 +/- .1 vs IN 2.5 +/- .3 and OS 2.7 +/- .4; P less than .01), whereas ON increased (P less than .01) LH (3.7 +/- .3 ng/ml) even further. Mean LH increased (P less than .05) after naloxone administration in all treatment groups. However, magnitude of this response was variable and dependent on ovarian status. Amplitude of the naloxone-induced LH response was greater (P less than .05) for ovariectomized (5.9 +/- 1.1 ng/ml) than for intact groups (2.7 +/- .5 ng/ml). Gonadotropin-releasing hormone increased mean LH concentrations in all groups. We suggest that ovarian secretions and the suckling stimulus contribute to endogenous opioid inhibition of LH during the postpartum interval.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Bovinos/metabolismo , Lactação , Hormônio Luteinizante/metabolismo , Naloxona/farmacologia , Ovário/fisiologia , Animais , Animais Lactentes , Feminino , Hormônio Luteinizante/sangue , Hormônios Liberadores de Hormônios Hipofisários/farmacologia , Período Pós-Parto/efeitos dos fármacos , Gravidez , Distribuição Aleatória
18.
Biol Reprod ; 39(4): 963-75, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2974732

RESUMO

The anatomical locations of proopiomelanocortin (POMC) and luteinizing hormone-releasing hormone (LHRH) neurons were examined in brain tissue from peripubertal female calves and from mature, luteal-phase cows. Biotin-avidin-peroxidase procedures were used for single- and double-labeled immunostaining. LHRH perikarya formed a loosely arranged continuum, extending posteriorly from the diagonal band of Broca, passing through the medial and lateral preoptic areas, and ending within the anterior hypothalamic area. LHRH fibers, apparently directed toward the median eminence, passed (1) posteroventrally in the periventricular area and through the arcuate nucleus, and (2) ventromedially lateral to the arcuate nucleus and medial to the supraoptic nucleus. POMC perikarya were located within and about the arcuate nucleus, some penetrating into the median eminence. Fibers from these POMC perikarya passed ventrally to terminate in the median eminence and to regions ventral to the mammillary nuclei. POMC fibers also projected dorsally and laterally from the arcuate nucleus to other hypothalamic regions as well as anteriorly along the third ventricular wall to the preoptic area, bed nucleus of stria terminalis, and stria terminalis. Less than 6% of the POMC or LHRH processes were in close anatomical apposition to LHRH perikarya and dendrites. Extensive intermingling of LHRH and POMC fibers occurred within zona externa of the median eminence.


Assuntos
Hormônio Liberador de Gonadotropina/análise , Hipotálamo/análise , Neurônios/análise , Área Pré-Óptica/análise , Pró-Opiomelanocortina/análise , Animais , Bovinos , Feminino , Imuno-Histoquímica , Neurônios/ultraestrutura , beta-Endorfina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...