Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
Reprod Fertil Dev ; 29(7): 1426-1434, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27328025

ABSTRACT

Androgens synergise with FSH in female reproduction but the nature of their interaction in ovarian function and fertility is not clear. In the present study, we investigated this interaction, notably whether higher endogenous FSH can overcome defective androgen actions in androgen receptor (AR)-knockout (ARKO) mice. We generated and investigated the reproductive function of mutant mice exhibiting AR resistance with or without expression of human transgenic FSH (Tg-FSH). On the background of inactivated AR signalling, which alone resulted in irregular oestrous cycles and reduced pups per litter, ovulation rates and antral follicle health, Tg-FSH expression restored follicle health, ovulation rates and litter size to wild-type levels. However, Tg-FSH was only able to partially rectify the abnormal oestrous cycles observed in ARKO females. Hence, elevated endogenous FSH rescued the intraovarian defects, and partially rescued the extraovarian defects due to androgen insensitivity. In addition, the observed increase in litter size in Tg-FSH females was not observed in the presence of AR signalling inactivation. In summary, the findings of the present study reveal that FSH can rescue impaired female fertility and ovarian function due to androgen insensitivity in female ARKO mice by maintaining follicle health and ovulation rates, and thereby optimal female fertility.


Subject(s)
Follicle Stimulating Hormone, Human/genetics , Follicle Stimulating Hormone, Human/physiology , Infertility, Female/therapy , Receptors, Androgen/deficiency , Animals , Disease Models, Animal , Estrus , Female , Fertility/genetics , Fertility/physiology , Humans , Infertility, Female/genetics , Infertility, Female/physiopathology , Litter Size , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovary/pathology , Ovary/physiopathology , Pregnancy , Receptors, Androgen/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
2.
Endocrinology ; 156(4): 1441-52, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25643156

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with reproductive, endocrine, and metabolic abnormalities. Because hyperandrogenism is the most consistent PCOS feature, we used wild-type (WT) and androgen receptor (AR) knockout (ARKO) mice, together with a mouse model of PCOS, to investigate the contribution of genomic AR-mediated actions in the development of PCOS traits. PCOS features were induced by prenatal exposure to dihydrotestosterone (250 µg) or oil vehicle (control) on days 16-18 of gestation in WT, heterozygote, and homozygote ARKO mice. DHT treatment of WT mice induced ovarian cysts (100% vs 0%), disrupted estrous cycles (42% vs 100% cycling), and led to fewer corpora lutea (5.0±0.4 vs 9.8±1.8). However, diestrus serum LH and FSH, and estradiol-induced-negative feedback as well as hypothalamic expression of kisspeptin, neurokinin B, and dynorphin, were unaffected by DHT treatment in WT mice. DHT-treated WT mice exhibited a more than 48% increase in adipocyte area but without changes in body fat. In contrast, heterozygous and homozygous ARKO mice exposed to DHT maintained comparable ovarian (histo)morphology, estrous cycling, and corpora lutea numbers, without any increase in adipocyte size. These findings provide strong evidence that genomic AR signaling is an important mediator in the development of these PCOS traits with a dose dependency that allows even AR haplosufficiency to prevent induction by prenatal androgenization of PCOS features in adult life.


Subject(s)
Hyperandrogenism/prevention & control , Polycystic Ovary Syndrome/prevention & control , Prenatal Exposure Delayed Effects/prevention & control , Receptors, Androgen/metabolism , Adipose Tissue/metabolism , Androgens , Animals , Disease Models, Animal , Estrous Cycle , Female , Hyperandrogenism/chemically induced , Hyperandrogenism/metabolism , Mice , Mice, Knockout , Ovary/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Receptors, Androgen/genetics
3.
Endocrinology ; 155(8): 3146-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24877633

ABSTRACT

Polycystic ovary syndrome (PCOS) affects 5-10% of women of reproductive age, causing a range of reproductive, metabolic and endocrine defects including anovulation, infertility, hyperandrogenism, obesity, hyperinsulinism, and an increased risk of type 2 diabetes and cardiovascular disease. Hyperandrogenism is the most consistent feature of PCOS, but its etiology remains unknown, and ethical and logistic constraints limit definitive experimentation in humans to determine mechanisms involved. In this study, we provide the first comprehensive characterization of reproductive, endocrine, and metabolic PCOS traits in 4 distinct murine models of hyperandrogenism, comprising prenatal dihydrotestosterone (DHT, potent nonaromatizable androgen) treatment during days 16-18 of gestation, or long-term treatment (90 days from 21 days of age) with DHT, dehydroepiandrosterone (DHEA), or letrozole (aromatase inhibitor). Prenatal DHT-treated mature mice exhibited irregular estrous cycles, oligo-ovulation, reduced preantral follicle health, hepatic steatosis, and adipocyte hypertrophy, but lacked overall changes in body-fat composition. Long-term DHT treatment induced polycystic ovaries displaying unhealthy antral follicles (degenerate oocyte and/or > 10% pyknotic granulosa cells), as well as anovulation and acyclicity in mature (16-week-old) females. Long-term DHT also increased body and fat pad weights and induced adipocyte hypertrophy and hypercholesterolemia. Long-term letrozole-treated mice exhibited absent or irregular cycles, oligo-ovulation, polycystic ovaries containing hemorrhagic cysts atypical of PCOS, and displayed no metabolic features of PCOS. Long-term dehydroepiandrosterone treatment produced no PCOS features in mature mice. Our findings reveal that long-term DHT treatment replicated a breadth of ovarian, endocrine, and metabolic features of human PCOS and provides the best mouse model for experimental studies of PCOS pathogenesis.


Subject(s)
Disease Models, Animal , Hyperandrogenism/complications , Ovary/pathology , Polycystic Ovary Syndrome/etiology , Adipose Tissue/pathology , Adiposity , Animals , Body Weight , Cholesterol/blood , Estrous Cycle , Female , Follicle Stimulating Hormone/blood , Gonadal Steroid Hormones/blood , Insulin Resistance , Liver/pathology , Luteinizing Hormone/blood , Male , Mice , Mice, Inbred C57BL , Myocardium/pathology , Organ Size , Ovary/physiopathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , Triglycerides/blood
4.
Endocrinology ; 150(7): 3274-82, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19359383

ABSTRACT

Female androgen receptor (AR) knockout mice (AR(-/-)) generated by an in-frame Ar exon 3 deletion are subfertile, but the mechanism is not clearly defined. To distinguish between extra- and intraovarian defects, reciprocal ovarian transplants were undertaken. Ovariectomized AR(-/-) hosts with wild-type (AR(+/+)) ovary transplants displayed abnormal estrus cycles, with longer cycles (50%, P < 0.05), and 66% were infertile (P < 0.05), whereas AR(+/+) hosts with either AR(-/-) or surgical control AR(+/+) ovary transplants displayed normal estrus cycles and fertility. These data imply a neuroendocrine defect, which is further supported by increased FSH (P <0.05) and estradiol (P <0.05), and greater LH suppressibility by estradiol in AR(-/-) females at estrus (P <0.05). Additional intraovarian defects were observed by the finding that both experimental transplant groups exhibited significantly reduced pups per litter (P < 0.05) and corpora lutea numbers (P < 0.05) compared with surgical controls. All groups exhibited normal uterine and lactation functions. AR(-/-) uteri were morphologically different from AR(+/+) with an increase in horn length (P < 0.01) but a reduction in uterine diameter (P < 0.05), total uterine area (P < 0.05), endometrial area (P < 0.05), and myometrial area (P < 0.01) at diestrus, indicating a role for AR in uterine growth and development. Both experimental transplant groups displayed a significant reduction in uterine diameter (P < 0.01) compared with transplanted wild-type controls, indicating a role for both AR-mediated intraovarian and intrauterine influences on uterine physiology. In conclusion, these data provide direct evidence that extraovarian neuroendocrine, but not uterine effects, as well as local intraovarian AR-mediated actions are important in maintaining female fertility, and a disruption of AR signaling leads to altered uterine development.


Subject(s)
Infertility, Female/genetics , Ovary/physiology , Receptors, Androgen/genetics , Uterus/physiology , Animals , Estradiol/blood , Estradiol/pharmacology , Estrous Cycle , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/genetics , Male , Mice , Mice, Knockout , Ovariectomy , Ovary/transplantation , Pituitary Gland/metabolism , Receptors, LHRH/genetics , Uterus/physiopathology
5.
Biol Reprod ; 78(3): 380-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18003945

ABSTRACT

Although androgens and the androgen receptor (AR) have defining roles in male reproductive development and function, previously no role in female reproductive physiology beyond testosterone (T) as the precursor in estradiol (E(2)) biosynthesis was firmly established. Understanding the role and specific mechanisms of androgen action via the AR in the ovary has been limited by confusion on how to interpret results from pharmacological studies, because many androgens can be metabolized in vivo and in vitro to steroids that can also exert actions via the estrogen receptor (ESR). Recent genetic studies using mouse models with specific disruption of the Ar gene have highlighted the role that AR-mediated actions play in maintaining female fertility through key roles in the regulation of follicle health, development, and ovulation. Furthermore, these genetic studies have revealed that AR-mediated effects influence age-related female fertility, possibly via mechanisms acting predominantly at the hypothalamic-pituitary axis in a dose-dependent manner. This review focuses on combining the findings from pharmacological studies and novel genetic mouse models to unravel the roles of ovarian androgen actions in relation to female fertility and ovarian aging, as well as creating new insights into the role of androgens in androgen-associated reproductive disorders such as polycystic ovarian syndrome.


Subject(s)
Androgens/pharmacology , Ovary/drug effects , Aging/physiology , Androgens/physiology , Animals , Female , Follicular Atresia/drug effects , Humans , Mice , Mice, Transgenic , Models, Biological , Oogenesis/physiology , Ovarian Follicle/physiology , Ovary/metabolism , Ovary/physiology , Polycystic Ovary Syndrome/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
6.
Endocrinology ; 148(8): 3674-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17463055

ABSTRACT

The role of classical genomic androgen receptor (AR) mediated actions in female reproductive physiology remains unclear. Female mice homozygous for an in-frame deletion of exon 3 of the Ar (AR(-/-)) were subfertile, exhibiting delayed production of their first litter (AR(+/+) = 22 d vs. AR(-/-) = 61 d, P < 0.05) and producing 60% fewer pups/litter (AR(+/+): 8.1 +/- 0.4 vs. AR(-/-): 3.2 +/- 0.9, P < 0.01). Heterozygous females (AR(+/-)) exhibited an age-dependent 55% reduction (P < 0.01) in pups per litter, evident from 6 months of age (P < 0.05), compared with AR(+/+), indicating a significant gene dosage effect on female fertility. Ovulation was defective with a significant reduction in corpora lutea numbers (48-79%, P < 0.01) in 10- to 12- and 26-wk-old AR(+/-) and AR(-/-) females and a 57% reduction in oocytes recovered from naturally mated AR(-/-) females (AR(+/+): 9.8 +/- 1.0 vs. AR(-/-): 4.2 +/- 1.2, P < 0.01); however, early embryo development to the two-cell stage was unaltered. The delay in first litter, reduction in natural ovulation rate, and aromatase expression in AR(+/-) and AR(-/-) ovaries, coupled with the restored ovulation rate by gonadotropin hyperstimulation in AR(-/-) females, suggest aberrant gonadotropin regulation. A 2.7-fold increase (AR(+/+): 35.4 +/- 13.4 vs. AR(-/-): 93.9 +/- 6.1, P < 0.01) in morphologically unhealthy antral follicles demonstrated deficiencies in late follicular development, although growing follicle populations and growth rates were unaltered. This novel model reveals that classical genomic AR action is critical for normal ovarian function, although not for follicle depletion and that haploinsufficiency for an inactivated AR may contribute to a premature reduction in female fecundity.


Subject(s)
Aging/physiology , Infertility, Female/physiopathology , Ovarian Follicle/physiology , Ovulation/physiology , Receptors, Androgen/genetics , Aging/pathology , Animals , Cell Count , Disease Models, Animal , Embryonic Development/physiology , Estradiol/blood , Female , Fertility/physiology , Follicle Stimulating Hormone/blood , Genotype , Infertility, Female/pathology , Luteinizing Hormone/blood , Mice , Mice, Knockout , Ovarian Follicle/growth & development , Ovarian Follicle/pathology , Ovulation Induction , Phenotype , Pregnancy , Receptors, Androgen/metabolism , Testosterone/blood
7.
J Endocrinol ; 188(3): 549-57, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16522734

ABSTRACT

Ovarian primordial follicle reserve is considered hormonally independent or subject to depletion by FSH-driven follicle recruitment. To explore specific in vivo effects of FSH on early follicle populations in the absence of luteinizing hormone (LH) activity, we examined mature hypogonadal (hpg), gonadotrophin-deficient mice expressing transgenic (tg) human FSH. Sustained expression of tg-FSH (5.3 +/- 0.3 IU/l) increased ovary weights fourfold and significantly elevated total primordial follicle numbers twofold in tg-FSH hpg (4209 +/- 457) relative to non-tg hpg (2079 +/- 391) and wild-type (2043 +/- 195) age-matched ovaries. Absolute primary follicle numbers in tg-FSH hpg ovaries were similar to non-tg hpg and wild-type ovaries. Furthermore, tg-FSH quantitatively increased secondary and antral follicles in hpg ovaries to numbers equivalent to wild-type, but did not induce ovulation, indicating a selective FSH response without LH. Circulating inhibin B and inhibin A levels were significantly increased in tg-FSH hpg females compared with hpg controls, and inhibin B correlated with antral number, consistent with FSH-driven antral follicle formation. These findings revealed that sustained pituitary-independent FSH activity, in the absence of endogenous gonadotrophins, promotes an increase in primordial follicle reserve despite also stimulating follicular growth in mature females. Therefore, the tg-FSH hpg ovary presents a novel paradigm to evaluate specific gonadotrophin effects on follicle reserve and recruitment.


Subject(s)
Follicle Stimulating Hormone/genetics , Hypogonadism/metabolism , Ovarian Follicle/physiology , Animals , Body Weight , Female , Gonadotropin-Releasing Hormone/genetics , Humans , Hypogonadism/genetics , Inhibins/blood , Mice , Mice, Transgenic , Organ Size , Ovary/anatomy & histology
8.
Proc Natl Acad Sci U S A ; 102(8): 2992-7, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15710891

ABSTRACT

Toll-like receptors (TLRs) bind pathogen-specific ligands early in infection, initiating signaling pathways that lead to expression of multiple protective cellular genes. Many viruses have evolved strategies that block the effector mechanisms induced through these signaling pathways, but viral interference with critical proximal receptor interactions has not been described. We show here that the NS3/4A serine protease of hepatitis C virus (HCV), a virus notorious for its ability to establish persistent intrahepatic infection, causes specific proteolysis of Toll-IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF or TICAM-1), an adaptor protein linking TLR3 to kinases responsible for activating IFN regulatory factor 3 (IRF-3) and NF-kappaB, transcription factors controlling a multiplicity of antiviral defenses. NS3/4A-mediated cleavage of TRIF reduces its abundance and inhibits polyI:C-activated signaling through the TLR3 pathway before its bifurcation to IRF-3 and NF-kappaB. This uniquely broad mechanism of immune evasion potentially limits expression of multiple host defense genes, thereby promoting persistent infections with this medically important virus.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Hepacivirus/immunology , Viral Nonstructural Proteins/physiology , Amino Acid Sequence , Cells, Cultured , DNA-Binding Proteins/physiology , Humans , Interferon Regulatory Factor-3 , Membrane Glycoproteins/physiology , Molecular Sequence Data , Receptors, Cell Surface/physiology , Signal Transduction , Toll-Like Receptor 3 , Toll-Like Receptors , Transcription Factors/physiology
9.
Biol Reprod ; 72(1): 78-85, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15342359

ABSTRACT

Sensitive and specific measurement of FSH is critical to research in reproductive biology, and the increasing availability of transgenic mouse models has created a need for a robust, sensitive, and specific mouse (m) FSH assay. The present study evaluated a time-resolved immunofluorometric assay (IFMA) for mFSH using monoclonal antibody to human (h) FSHbeta as a capture antibody and a biotinylated polyclonal antibody to rat alpha subunit as a detection probe, with signaling amplified by europium-labeled streptavidin. The mFSH IFMA lowered the detection limit 34-fold (5 vs. 170 pg/sample) compared with standard mFSH RIA. The mFSH IFMA demonstrated parallelism of response to dilutions of castrated mouse serum and rat FSH but no cross-reactivity with hFSH and mLH or hLH, whereas the RIA demonstrated nonparallel cross-reactivity with hFSH. The IFMA has a wide analytical range, with a good precision profile for within- and between-assay reproducibility. Because the IFMA is a sandwich-type assay with strict dimer-specificity by design, the lower readings and recovery obtained were compared with the RIA when both assays used a pituitary-purified mFSH assay standard that contained isolated or fragmented subunits as well as intact dimeric FSH. When used with mouse serum sample, the mFSH IFMA demonstrated the expected increases following orchidectomy as well as markedly enhanced sensitivity to very low levels of endogenous mFSH in gonadotropin-deficient mice. Furthermore, the IFMA measured mFSH with fidelity in both intact and orchidectomized male mice without any interference from transgenic hFSH. The greatly enhanced sensitivity, specificity, and technical convenience of this mFSH IFMA will allow wider application of FSH measurements to very small blood samples in immature and mature mice as well as transgenic models.


Subject(s)
Fluoroimmunoassay/methods , Follicle Stimulating Hormone/blood , Animals , Antibodies, Monoclonal/immunology , Cross Reactions , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/immunology , Follicle Stimulating Hormone, Human/immunology , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/immunology , Hypogonadism/blood , Male , Mice , Mice, Transgenic , Orchiectomy , Sensitivity and Specificity
10.
Endocrinology ; 142(6): 2213-20, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356665

ABSTRACT

Gonadal function is wholly reliant on the two pituitary-derived gonadotropins, FSH and LH. Identifying the specific effects of FSH has been difficult because of the intimate relationship between LH and FSH action and inherent limitations of classic research paradigms. We describe a novel transgenic model to characterize the definitive actions of FSH alone, distinct from LH effects, created by combining transgenic FSH expression with the gonadotropin-deficient background of the hypogonadal (hpg) mouse. A tandem transgene construct encoding each alpha- and beta-subunit of human FSH, under the rat insulin II promoter, expressed biologically active heterodimers at serum levels, by immunoassay, equivalent to circulating FSH concentrations in fertile humans (0.1-25 IU/liter). Transgenic mice were crossed into the hpg mouse genotype to obtain LH-deficient animals secreting FSH alone. Testis weights of adult FSHxhpg mice were increased up to 5-fold, relative to nontransgenic hpg controls (P < 0.001). However, only transgenic males with serum FSH levels more than 1 IU/liter showed testis weights increased relative to hpg controls, indicating a physiological FSH threshold for the testicular response. Histology of enlarged FSHxhpg testes revealed round spermatids and sparse numbers of elongated spermatids, demonstrating that the testosterone-independent FSH response targeting the Sertoli cell can facilitate completion of meiosis and minimal initiation, but not completion, of spermiogenesis. Transgenic FSH also induced inhibin B secretion in FSHxhpg mice, but showed a distinct sexual dimorphism with only females exhibiting a strong FSH dose-dependent increase in serum inhibin B levels (r(2) = 0.84). In addition, ovaries of FSHxhpg females were enlarged up to 10-fold (P < 0.001), characterized by increased follicular recruitment and development to type 7 antral follicles. Thus, these findings show that the transgenic FSHxhpg mouse provides a unique model for detailed investigations of the definitive in vivo actions of FSH alone.


Subject(s)
Follicle Stimulating Hormone/physiology , Gonads/physiology , Luteinizing Hormone/physiology , Animals , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone, beta Subunit , Gene Expression , Glycoprotein Hormones, alpha Subunit/genetics , Humans , Hypogonadism/genetics , Inhibins/blood , Insulin/genetics , Insulinoma , Male , Mice , Mice, Transgenic , Organ Size , Ovary/anatomy & histology , Ovary/physiology , Pancreatic Neoplasms , Promoter Regions, Genetic , Rats , Spermatogenesis/drug effects , Testis/anatomy & histology , Testis/physiology , Testosterone/blood , Transfection , Tumor Cells, Cultured
11.
Comb Chem High Throughput Screen ; 3(3): 185-96, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10903378

ABSTRACT

We describe a technology for generating recombinant polyclonal antibody libraries (PCALs) that enables the creation and perpetuation of standardized mixtures of polyclonal whole antibodies specific for a multiantigen (or polyantigen). Therefore, this technology combines the advantages of targeting multiple antigenic determinants -- high avidity, low likelihood of antigen 'escape variants', and efficient mediation of effector functions, with the advantages of using monoclonal antibodies -- unlimited supply of standardized reagents and the availability of the genetic material for desired manipulations. The technology for generating recombinant polyclonal antibody libraries begins with the creation of phage display Fab (antibody) libraries. This is followed by selection of sublibraries with desired antigen specificities, and mass transfer of the variable region gene pairs of the selected sublibraries to a mammalian expression vector for generation of libraries of polyclonal whole antibodies. We review here our experiments for selection of phage display antibody libraries against microbes and tumor cells, as well as the recent literature on the selection of phage display antibody libraries to multiantigen targets.


Subject(s)
Antibodies/genetics , Gene Library , Recombinant Proteins/immunology , Animals , Breast Neoplasms/immunology , Cryptosporidium parvum/immunology , DNA Primers , Female , Genetic Vectors , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Variable Region/genetics , Ovarian Neoplasms/immunology , Peptide Library , Recombinant Proteins/genetics
12.
Mol Cell Biol Res Commun ; 3(2): 76-81, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10775503

ABSTRACT

We have identified a new factor, CFX, in human serum and plasma that inhibits the growth of cultured human and mouse cell lines. CFX was determined to be a negatively charged, hydrophobic glycoprotein, with a native molecular weight of 110-120 kDa and a minimal active subunit of 55 kDa. It is precipitated by 60% ammonium sulfate and is resistant to heat treatment at 100 degrees C for 30 min. CFX was purified from human plasma to a single band on a gel which retained the cell growth inhibitory activity. Amino acid sequence analysis of the CFX band revealed sequences from four human glycoproteins, alpha1-antichymotrypsin, C1-esterase inhibitor, alpha1-antitrypsin, and alpha2-antiplasmin, all members of the superfamily of serpins. Of the four, C1-esterase inhibitor was shown to be the most potent cell growth inhibitor. These results suggest that serpins may play a cell growth inhibitory role in vivo, in addition to their role as protease inhibitors.


Subject(s)
Cell Division/drug effects , Serpins/blood , Serpins/pharmacology , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Humans , Peptide Mapping , Serpins/chemistry , Serpins/isolation & purification , Trypsin/chemistry , Tumor Cells, Cultured
13.
Endocrinology ; 140(9): 3938-46, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465262

ABSTRACT

We showed previously that testosterone (T) alone could induce spermatogenesis and produce normally fertile spermatozoa in the absence of circulating gonadotropins. These studies used the hpg mouse, which is characterized by a congenital gonadotrophin deficiency due to a major deletion in the GnRH gene. Administering T by a subdermal implant of a SILASTIC brand tube impregnated with crystalline T showed that the androgenic requirement for full induction of spermatogenesis was a 1-cm length implant. Using this unique model of spermatogenesis without gonadotropins, we have now investigated the quantitative requirement for androgens to maintain spermatogenesis by testing the hypothesis that the androgenic threshold required for induction and maintenance of spermatogenesis are the same. Spermatogenesis was induced in homozygous hpg mice by T administration for 6 weeks. The first experiment determined the time-course of the regression of spermatogenesis after removal of the T-impregnated SILASTIC brand implant. Elongated spermatids were absent by 3 weeks and testicular weight regression was maximal by 4 weeks after androgen withdrawal. The second experiment examined the effects on maintenance of spermatogenesis of reducing the T dose. After full induction of spermatogenesis in homozygous hpg mice, the T implants were replaced with a range of smaller size T-impregnated SILASTIC brand implants for a further 4 weeks. All androgen-sensitive end-points (testis weight, tubular, and luminal diameters, round spermatids) were fully maintained with T implants of 0.06 cm and elongated spermatids with T implants of 0.25 cm. A further experiment showed that at very low T doses (0.06, 0.125 cm) the T effects observed at 4 weeks were maintained at 6 and 11 weeks duration. We conclude that the androgenic threshold to maintain spermatogenesis in the mouse is an order of magnitude lower than the threshold required for inducing spermatogenesis. This distinction suggests that the mechanism of action of testosterone in inducing spermatogenesis may involve regulation of a genetic switch to complete meiosis, whereas the maintenance involves a different locus of action. These findings suggest that further studies of androgen-dependent meiotic genes may be central to understanding the regulation and molecular basis of androgen-driven induction and maintenance of spermatogenesis.


Subject(s)
Gonadotropins/physiology , Spermatogenesis/drug effects , Spermatogenesis/physiology , Testosterone/pharmacology , Androgens/deficiency , Androgens/genetics , Animals , Cell Count , Differential Threshold , Gene Deletion , Male , Mice , Mice, Inbred C3H/genetics , Organ Size , Sertoli Cells/cytology , Sperm Count , Spermatids/cytology , Spermatids/ultrastructure , Testis/anatomy & histology
14.
Comb Chem High Throughput Screen ; 2(6): 319-25, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10644857

ABSTRACT

We had developed a technology for creation of recombinant polyclonal antibody libraries, standardized perpetual mixtures of polyclonal whole antibodies for which the genes are available and can be altered as desired. We report here the first phase of generating a polyclonal antibody library to Cryptosporidium parvum, a protozoan parasite that causes severe disease in AIDS patients, for which there is no effective treatment. BALB/c mice, immunized by neonatal oral infection with oocysts followed by intraperitoneal immunization with a sporozoite/oocyst preparation of C. parvum, were used for construction of a Fab phage display library in a specially-designed bidirectional vector. This library was selected for reactivity to an oocyst/sporozoite preparation, and was shown to be antigen-specific and diverse. Following mass transfer of the selected variable region gene pairs to appropriate mammalian expression vectors, such anti-C. parvum Fab phage display libraries could be used to develop chimeric polyclonal antibody libraries, with mouse variable regions and human constant regions, for passive immunotherapy of C. parvum infection.


Subject(s)
Antibodies, Protozoan/genetics , Cryptosporidium parvum/immunology , Immunoglobulin Fab Fragments/genetics , Peptide Library , Animals , Base Sequence , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data
15.
Lab Invest ; 78(4): 393-400, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9564884

ABSTRACT

Epithelial repair following acute lung injury involves proliferation and differentiation of existing Clara cells and type II cells. Other mechanisms of epithelial repair may be involved in particularly severe cases. We used epithelial cell-specific markers to examine changes in the mouse lung epithelium 28 days after bleomycin treatment. The spatial distribution of surfactant proteins A, B, C (SPA, SPB, SPC), and Clara cell-specific protein (CC10) mRNA was compared by in situ hybridization in serial lung sections. CC10 mRNA-containing airway cells were replaced in many areas by SPB mRNA-expressing, ciliated cells that did not contain CC10 mRNA. In distal airway regions, we observed a subpopulation of epithelial cells that appeared to express SPA, SPB, SPC, and CC10 mRNA, and speculated that they may represent a multipotential stem cell population. These cells were found in focal clusters, which suggests that they expanded from a common cell. CC10 mRNA-containing cells were seen in alveolar-like structures thought to be the result of Clara cell migration or outpocketing. Our data suggest that there are repair mechanisms involved in epithelial repair after severe injury that have not previously been described.


Subject(s)
Bleomycin/toxicity , Lung/drug effects , Uteroglobin , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Proteins/genetics , Pulmonary Surfactants/genetics , RNA, Messenger/analysis
16.
J Biol Chem ; 272(46): 29113-9, 1997 Nov 14.
Article in English | MEDLINE | ID: mdl-9360987

ABSTRACT

We show that the liver-specific expression of all four genes in the human apolipoprotein (apo) E/C-I/C-IV/C-II gene cluster in transgenic mice is determined by the coordinate action of two distinct hepatic control regions (HCR). These enhancers are positioned 15 kilobases (kb) (HCR.1) and 26 kb (HCR.2) downstream of the apoE gene. To investigate the action of each HCR, transgenic mice were generated with a 70-kb human genomic fragment that contained the complete apoE gene cluster or with this fragment modified by the specific deletion of HCR.1, HCR.2, or both HCR domains. Hepatic expression of all four apolipoprotein genes was observed in transgenic mice in which either HCR.1 or HCR.2 was deleted, but no transgene expression was found in the liver in the absence of both HCR domains. The overall patterns of transgene expression suggested that HCR.2 was the dominant element for apoC-IV and apoC-II expression and that HCR.1 was dominant for the apoE/C-I expression. No liver-specific transcriptional activity was identified for the proximal promoter of any gene in the cluster; all liver-specific activity was associated with HCR.1 and HCR.2. Thus, the HCRs of the apoE gene cluster constitute unique regulatory domains for determining the requirements for apolipoprotein gene expression in the liver.


Subject(s)
Apolipoproteins C/genetics , Apolipoproteins E/genetics , Liver/metabolism , Multigene Family , Animals , DNA/genetics , Gene Expression , Humans , Mice , Mice, Transgenic
17.
Immunology ; 91(3): 375-82, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9301526

ABSTRACT

The contribution of T-cell-receptor beta-chain diversity to the T-cell antigen-specific repertoire was investigated using single-chain T-cell-receptor transgenic mice. Animals that express the rearranged beta-chain gene from a T hybridoma with specificity for a hen egg lysozyme peptide, designated HEL (85-96) were analysed for their ability to respond to a panel of diverse antigens. Transgenic mice exhibited a significantly elevated response to HEL (85-96) which was shown to be due to an increased frequency of HEL (85-96)-specific T-cell progenitors. This increased frequency of specific progenitors resulted in the ability of transgenic mice to respond to the peptide in the absence of antigen priming. Conversely, transgenic mice failed to respond to any other antigen tested. Furthermore, this apparent deficiency was associated with a significant decrease in the frequency of antigen-specific T-cell progenitors in transgenic mice. Surprisingly, the ability to launch an alloresponse was unaffected by the exclusive expression of the transgene-derived beta-chain. These results indicate that beta-chain diversity is crucial for the ability of the T-cell population to elicit a rapid and robust response to the profusion of different antigen/major histocompatibility complex (MHC) ligands potentially encountered by an individual. Furthermore, these results suggest a lesser role for beta-chain diversity in contributing to allorecognition, and support a model in which the direct recognition of peptide-mediated conformational MHC forms is the major contributor to the alloreactive response exhibited by the majority of T cells.


Subject(s)
Epitopes/immunology , Isoantigens/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Cell Division/immunology , Cytochrome c Group/immunology , H-2 Antigens/immunology , Immune Tolerance , Immunologic Memory , Mice , Mice, Transgenic , Muramidase/immunology , Peptide Fragments/immunology
18.
Toxicol Appl Pharmacol ; 142(2): 303-10, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9070353

ABSTRACT

Clinical use of the antineoplastic agent bleomycin is restricted due to pulmonary toxicity. Murine models of bleomycin-induced pulmonary fibrosis have been developed in an attempt to understand the mechanisms involved in the fibrotic process. Studies have shown that the alveolar epithelium is damaged early after bleomycin treatment. The purpose of this study was to evaluate the pattern of gene expression in airway and alveolar epithelial cells after bleomycin exposure in mice that vary in susceptibility to bleomycin-induced fibrosis. Surfactant protein C (SPC) and Clara cell-specific protein (CC10) mRNA were used as cell-specific markers of alveolar type II cells and airway Clara cells, respectively. Mice were treated with a single intratracheal dose of bleomycin and the pattern of SPC and CC10 transcripts was examined by in situ hybridization. The pattern of SPC mRNA 28 days after treatment was uniform in controls and resistant mice but exhibited a patchy appearance in sensitive mice. Bleomycin treatment also resulted in a strain-dependent loss of CC10 mRNA-expressing cells. In sensitive mice 28 days after treatment, SPC mRNA was ectopically expressed in the distal bronchiolar epithelium in a morphologically distinct cell type. Serial sections revealed that these cells either coexpressed CC10 mRNA or were located adjacent to CC10 mRNA-containing cells. This unique cell population may represent a progenitor cell type important in epithelial repair. The strain-dependent changes in CC10 and SPC gene expression after bleomycin treatment are suggestive of a role for the epithelium in pulmonary fibrosis versus repair.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Enzyme Inhibitors/metabolism , Lung/drug effects , Protein Biosynthesis , Proteolipids/biosynthesis , Pulmonary Surfactants/biosynthesis , Uteroglobin , Animals , Antibiotics, Antineoplastic/administration & dosage , Biomarkers/analysis , Bleomycin/administration & dosage , Epithelial Cells , Epithelium/drug effects , In Situ Hybridization , Intubation, Intratracheal , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteins/analysis , Proteolipids/analysis , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Pulmonary Surfactants/analysis , Species Specificity
19.
J Lipid Res ; 37(7): 1510-8, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8827523

ABSTRACT

The human apolipoprotein (apo) C-IV gene has been recently identified: it is closely linked to the promoter region of the apoC-II gene (Allan, C.M., D. Walker, J. Segrest, and J. M. Taylor. 1995. Genomics. 28: 291-300). To determine the effect of apoC-IV gene expression on lipoprotein metabolism, transgenic mice were generated using a human apoC-IV cDNA construct. Human apoC-IV was found associated with plasma lipoproteins (d < 1.21 g/ml), mainly in very low density lipoproteins (VLDL), and higher molecular mass isoforms were present, due to N-linked glycosylation and variable sialylation of apoC-IV. Human apoC-IV transgenic mice were hypertriglyceridemic compared to nontransgenic controls; the accumulated plasma triglycerides were present mainly in VLDL. There was little change in plasma cholesterol levels, although apoC-IV expression redistributed cholesterol to VLDL and larger particles in low density lipoprotein/large high density lipoprotein fractions. By immunoblot analysis, apoC-IV was not detected in normal adult human plasma or isolated plasma lipoproteins, a finding consistent with our previous observation of very low levels of human apoC-IV mRNA in human liver. However, our analysis of transgenic mice provides unequivocal evidence that human apoC-IV is a lipid-binding protein belonging to the apolipoprotein family and that it has the potential to alter lipoprotein metabolism.


Subject(s)
Apolipoproteins C/genetics , Hypertriglyceridemia/genetics , Animals , Apolipoproteins C/blood , Apolipoproteins C/physiology , Cholesterol/blood , DNA, Complementary/genetics , Humans , Hypertriglyceridemia/metabolism , Lipoproteins, VLDL/blood , Liver/metabolism , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Transcription, Genetic , Triglycerides/blood
20.
N Z Vet J ; 44(3): 116-8, 1996 Jun.
Article in English | MEDLINE | ID: mdl-16031908

ABSTRACT

Examination of 25 cases of multiple benzimidazole and levamisole resistance, identified in sheep by faecal egg count reduction testing at the Batchelar, Lincoln and Invermay Animal Health Laboratories, showed that benzimidazole-levamisole combinations provided effective control in eight (47%) of 17 cases in which they were tested. Overall, the use of combination drenches resulted in average improvements in faecal egg count reductions of 25.6% (p<0.01) and 23.2% (p<0.05), respectively, over those achieved by the use of benzimidazole or levamisole drenches alone. The results suggest that instances of multiple resistance in which combination drenches might be effective are unlikely to be predictable either by identification of the parasites involved, or from the levels of benzimidazole or levamisole resistance present.

SELECTION OF CITATIONS
SEARCH DETAIL