Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 39(1): 111-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21519790

ABSTRACT

An ETS family member, ETS Related Gene (ERG) is involved in the Ewing family of tumors as well as leukemias. Rearrangement of the ERG gene with the TMPRSS2 gene has been identified in the majority of prostate cancer patients. Additionally, overexpression of ERG is associated with unfavorable prognosis in prostate cancer patients similar to leukemia patients. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) regulate transcription as well as epigenetic status of genes through acetylation of both histones and transcription factors. Deregulation of HATs and HDACs is frequently seen in various cancers, including prostate cancer. Many cellular oncogenes as well as tumor viral proteins are known to target either or both HATs and HDACs. Several studies have demonstrated that there are alterations of HDAC activity in prostate cancer cells. Recently, we found that ERG binds and inhibits HATs, which suggests that ERG is involved in deregulation of protein acetylation. Additionally, it has been shown that ERG is associated with a higher expression of HDACs. In this study, we tested the effect of the HDAC inhibitors valproic acid (VPA) and trichostatin-A (TSA) on ERG-positive prostate cancer cells (VCaP). We found that VPA and TSA induce apoptosis, upregulate p21/Waf1/CIP1, repress TMPRSS2-ERG expression and affect acetylation status of p53 in VCaP cells. These results suggest that HDAC inhibitors might restore HAT activity through two different ways: by inhibiting HDAC activity and by repressing HAT targeting oncoproteins such as ERG.


Subject(s)
Apoptosis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-ets/metabolism , Tumor Suppressor Protein p53/metabolism , Valproic Acid/pharmacology , Acetylation/drug effects , Animals , COS Cells , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Humans , Male , Models, Biological , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
2.
Article in English | MEDLINE | ID: mdl-18602321

ABSTRACT

Caspases belong to a family of highly conserved aspartate-specific cysteine proteases and are members of the interleukin-1beta-converting enzyme family, present in multicellular organisms. The caspase gene family consists of 15 mammalian members that are grouped into two major sub-families, namely inflammatory caspases and apoptotic caspases. The apoptotic caspases are further subdivided into two sub-groups, initiator caspases and executioner caspases. The caspases form a caspase-cascade system that plays the central role in the induction, transduction and amplification of intracellular apoptotic signals for cell fate determination, regulation of immunity, and cellular proliferation and differentiation. The substrates of apoptotic caspases have been associated with cellular dismantling, while inflammatory caspases mediate the proteolytic activation of inflammatory cytokines. The activation of this delicate caspase-cascade system and its functions are regulated by a variety of regulatory molecules, such as the inhibitor of apoptosis protein (IAP), FLICE, calpain, and Ca(2+). Based on the available literature we have reviewed and discussed the members of the caspase family, caspase-cascade system, caspase-regulating molecules and their apoptotic and non-apoptotic functions in cellular life and death. Also recent progress in the molecular structure and physiological role of non-mammalian caspases such as paracaspases, metacaspases and caspase-like-protease family members are included in relation to that of mammalian species.


Subject(s)
Caspases/chemistry , Caspases/metabolism , Animals , Caspases/genetics , Cell Physiological Phenomena , Enzyme Activation , Humans , Phylogeny , Protein Structure, Quaternary , Signal Transduction
3.
Endocrine ; 31(2): 125-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17873322

ABSTRACT

In humans, circulating leptin levels are low in early childhood and rise until puberty, whereas the reverse occurs for the soluble leptin receptor (sOB-R). In women, leptin remains high and sOB-R remains low, but in men leptin declines after adolescence and sOB-R increases. These observations suggest that leptin may regulate the production of sOB-R, and that the increased testosterone in adolescent boys may be responsible for the gender differences in leptin and sOB-R. To test this hypothesis, leptin was administered continuously to agonadal juvenile male monkeys for 16 days. No change in sOB-R was observed. Intact juvenile male monkeys were given pulsatile doses of gonadotropins for a period of 7 weeks to induce precocious puberty and assess the effect on plasma testosterone, leptin, and sOB-R. By 4 weeks testosterone had reached adult levels. No changes were observed in leptin, but by week 4, sOB-R was higher than pretreatment values and remained higher at week 7. These data suggest that leptin may not play a significant role in regulating the production of sOB-R and that gender differences in sOB-R in humans may be driven by the increased production of testosterone at puberty in males.


Subject(s)
Leptin/blood , Leptin/pharmacology , Receptors, Cell Surface/blood , Sexual Maturation/physiology , Animals , Gonadal Disorders/blood , Gonadal Disorders/chemically induced , Infusion Pumps , Leptin/administration & dosage , Macaca mulatta , Male , Receptors, Cell Surface/chemistry , Receptors, Leptin , Sexual Maturation/drug effects , Solubility , Testosterone/blood
4.
Cell Mol Biol Lett ; 11(4): 506-25, 2006.
Article in English | MEDLINE | ID: mdl-16977376

ABSTRACT

Apoptosis, or programmed cell death (PCD), involves a complex network of biochemical pathways that normally ensure a homeostatic balance between cellular proliferation and turnover in nearly all tissues. Apoptosis is essential for the body, as its deregulation can lead to several diseases. It plays a major role in a variety of physiological events, including embryonic development, tissue renewal, hormone-induced tissue atrophy, removal of inflammatory cells, and the evolution of granulation tissue into scar tissue. It also has an essential role in wound repair. The various cellular and biochemical mechanisms involved in apoptosis are not fully understood. However, there are two major pathways, the extrinsic pathway (receptor-mediated apoptotic pathway) and the intrinsic pathway (mitochondria-mediated apoptotic pathway), which are both well established. The key component in both is the activation of the caspase cascade. Caspases belong to the family of proteases that ultimately, by cleaving a set of proteins, cause disassembly of the cell. Although the caspase-mediated proteolytic cascade represents a central point in the apoptotic response, its initiation is tightly regulated by a variety of other factors. Among them, Bcl-2 family proteins, TNF and p53 play pivotal roles in the regulation of caspase activation and in the regulation of apoptosis. This review summarizes the established concepts in apoptosis as a physiological cell suicide program, highlighting the recent and significant advances in its study.


Subject(s)
Apoptosis/physiology , Animals , Caspases/physiology , Cell Death/physiology , Humans , Intracellular Membranes/enzymology , Intracellular Membranes/physiology
5.
J Androl ; 27(2): 302-10, 2006.
Article in English | MEDLINE | ID: mdl-16304204

ABSTRACT

Leptin-deficient (ob/ob) male mice are morbidly obese and exhibit impaired reproductive function. The objective of this study was to assess the effect of a leptin deficiency on testicular morphology, germ cell development, apoptotic activity within germ cells, and expression levels of apoptosis-related genes in the testis. Sixteen week-old ob/ob male mice (n = 8) and controls (n = 8) were killed, and their reproductive organs were weighed. Testes were processed for either histomorphological analysis (hematoxylin and eosin [H&E] staining), germ cell apoptosis assessment (deoxy-UTP-digoxigenin nick end labeling [TUNEL] method), or apoptosis-related gene expression analysis (microarray). Cross sections of the testes of leptin-deficient animals showed reduced seminiferous tubule area, fewer pachytene spermatocytes, and fewer tubules exhibiting elongated spermatids/mature spermatozoa. Condensation of germ cell nuclei and Sertoli cell vacuolization were evident in the testes of some ob/ob animals. Overall there was an elevation of apoptotic activity in the germ cells of ob/ob mice, particularly within the pachytene spermatocytes. With microarray technology, we identified 9 proapoptosis-related genes that were expressed at a significantly higher level in the testes of ob/ob mice than in the testes of the controls. Among these were members of the tumor necrosis factor receptor super family 1A and 5 (TNFR1 and 5) and peptidoglycan recognition proteins (associated with the extrinsic apoptotic pathway), and granzymes A and B, growth arrest and DNA damage inducible 45 gamma, sphingosine phosphate lyase 1, and caspase 9 (associated with the intrinsic apoptotic pathway). The results of the current study show that a leptin deficiency in mice is associated with impaired spermatogenesis, increased germ cell apoptosis, and up-regulated expression of proapoptotic genes within the testes.


Subject(s)
Apoptosis/genetics , Leptin/deficiency , Leptin/genetics , Spermatozoa/cytology , Spermatozoa/physiology , Testis/anatomy & histology , 3' Untranslated Regions/genetics , Animals , Body Weight , Gene Expression Regulation , In Situ Nick-End Labeling , Male , Mice , Oligonucleotide Array Sequence Analysis , Organ Size , Spermatogenesis , Testis/cytology
6.
Mol Cell Endocrinol ; 245(1-2): 121-7, 2005 Dec 21.
Article in English | MEDLINE | ID: mdl-16337737

ABSTRACT

Female leptin deficient (ob/ob) mice exhibit abnormal ovarian folliculogenesis resulting in an impaired ability to reproduce. This effect may be related to the hypogonadotropic state of these animals, or leptin may directly modulate ovarian follicle development. In the present study we assessed whether exogenous gonadotropin administration would normalize folliculogenesis and induce ovulation in immature ob/ob animals. Eight 26-day-old ob/ob and eight control mice were injected sc with pregnant mare serum gonadotropin followed 48 h later with a sc injection of human chorionic gonadotropin. Animals were killed 24 h later. Gonadotropin (GTH) administration increased both ovarian and uterine weights in control mice, but this effect was attenuated in leptin deficient animals. The number of preantral follicles was greater in ob/ob mice than controls, but in GTH-treated animals the number of antral follicles was subnormal in the ovaries of leptin deficient animals. Ob/ob animals also failed to ovulate in response to GTH, and the protective actions of GTH against granulosa cell apoptosis and follicular atresia were attenuated in these animals. Interestingly, however, serum levels of estradiol and progesterone were higher in ob/ob mice than controls, regardless of whether or not the animals received GTH treatment. We conclude that the ovarian responsiveness to GTH is subnormal in leptin deficient animals suggesting that leptin may modulate the process of folliculogenesis by directly altering the sensitivity of the ovary to GTH.


Subject(s)
Cell Differentiation/drug effects , Chorionic Gonadotropin/pharmacology , Gonadotropins, Equine/pharmacology , Ovarian Follicle/physiology , Sexual Maturation/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Body Weight/physiology , Cell Differentiation/physiology , Estradiol/blood , Female , Follicular Atresia/drug effects , Follicular Atresia/physiology , Granulosa Cells/cytology , Granulosa Cells/physiology , Leptin/deficiency , Leptin/physiology , Mice , Mice, Inbred C57BL , Mice, Obese , Organ Size/physiology , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovulation/drug effects , Ovulation/physiology , Progesterone/blood
7.
Eur J Endocrinol ; 153(1): 153-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15994757

ABSTRACT

OBJECTIVES: Subnormal leptin levels in low birth weight infants may allow for catch-up growth during infancy. Scant data are available that relate growth with circulating leptin during normal infancy in primates. The current study objective was to examine the association between serum leptin, its soluble receptor (sOB-R), testosterone and IGF-I concentrations, and body weight during infancy in male rhesus monkeys. DESIGN: Hormone levels were assessed longitudinally in animals (n = 7) from birth until 1 year of age. RESULTS: Body weight increased during the first 6 months of life and was strongly correlated with rising IGF-I levels and, as IGF-I plateaued and then declined during the second half of the year, body weight gain decelerated. In contrast, leptin levels declined gradually with age during the first year of life in conjunction with increasing body weight. There was no association between body weight gain and serum leptin levels or between serum testosterone and leptin values. Since sOB-R levels also declined with leptin values, it does not appear that levels of bioavailable leptin changed during infancy. CONCLUSIONS: The data do not support the contention that leptin regulates growth during infancy, but the close association between IGF-I levels and body weight suggested that this hormone may regulate growth in infant male monkeys. The failure to observe an association between serum testosterone and leptin concentrations suggested that leptin is not involved in the activation of the hypothalamic-pituitary -testicular axis during this developmental period.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Leptin/blood , Neurosecretory Systems/growth & development , Receptors, Cell Surface/blood , Testosterone/blood , Animals , Body Weight , Hypothalamo-Hypophyseal System/growth & development , Hypothalamo-Hypophyseal System/metabolism , Macaca mulatta , Male , Neurosecretory Systems/metabolism , Pituitary-Adrenal System/growth & development , Pituitary-Adrenal System/metabolism , Receptors, Leptin , Solubility , Testis/growth & development , Testis/metabolism
8.
Biol Reprod ; 71(1): 66-72, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14985253

ABSTRACT

Leptin purportedly plays an important role in pubertal development in a number of mammalian species. Adult leptin-deficient (ob/ob) female mice are infertile, but the mechanisms responsible for the reproductive failure have not been fully elucidated. The major objective of the current study was to assess the effects of a leptin deficiency on ovarian folliculogenesis and apoptosis. Beginning at 4 wk of age, control (n = 8) and ob/ob (n = 7) mice were weighed and examined daily for vaginal opening. After 3 wk the mice were killed, and the reproductive organs were weighed. Ovaries were paraffin-embedded for hematoxylin and eosin histology, TUNEL assay, and immunohistochemistry for Fas, Fas ligand (FasL), and proliferating cell nuclear antigen (PCNA). Vaginal opening was delayed, uteri were smaller, and the number of primordial follicles and total number of ovarian follicles were subnormal in ob/ob animals. Leptin-deficient animals also had a higher number of atretic follicles than controls. Granulosa cells (predominantly in preantral and early antral follicles) of ob/ob mice exhibited increased apoptotic activity as documented by TUNEL assay and elevated expression of the apoptotic markers Fas and FasL, compared with that in control animals. Ovarian expression of PCNA, a marker of DNA replication, repair, or both, did not differ between ob/ob and control mice. The data suggest that a leptin deficiency in mice is associated with impaired folliculogenesis, which results in increased follicular atresia. This impairment may be one of the causative components of infertility in leptin-deficient animals.


Subject(s)
Apoptosis , Granulosa Cells , Leptin/deficiency , Ovarian Follicle , Steroid Metabolism, Inborn Errors/physiopathology , Aging , Animals , Fas Ligand Protein , Female , Immunohistochemistry , Leptin/blood , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Size , Ovary/pathology , Proliferating Cell Nuclear Antigen/metabolism , Receptors, Leptin , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/pathology , Uterus/pathology , Vagina/physiopathology , fas Receptor/metabolism
9.
Biol Reprod ; 68(6): 2222-31, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12606359

ABSTRACT

We have demonstrated that continuous administration of a gonadotropin-releasing hormone agonist (GnRH-Ag) in vivo suppressed progesterone production and induced apoptosis in the corpus luteum (CL) of the pregnant rat. To investigate the mechanism(s) by which progesterone secretion is suppressed and apoptosis is induced in the luteal cells, we studied nitric oxide (NO) as a messenger molecule for GnRH action. Rats were treated individually on Day 8 of pregnancy with 5 microg/day of GnRH-Ag for 4, 8, and 24 h. GnRH-Ag decreased the production of progesterone and pregnenolone 8 and 24 h after the administration. Corresponding with the reduction in these steroid hormones, luteal NO concentrations decreased at 8 and 24 h. Western blotting and immunohistochemical studies of endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), and neuronal nitric oxide synthase (nNOS) in the CL demonstrated that administration of GnRH-Ag was associated with a marked decrease in eNOS and iNOS compared with sham controls at 4 and 8 h, but nNOS did not change throughout the experimental period. We demonstrated, for the first time, the presence of nNOS protein in the CL of the pregnant rat. To determine if this suppressive action of GnRH-Ag is directly on the CL, luteal cells were treated with GnRH-Ag for 4, 8, 12, and 24 h in vitro. Progesterone and NO concentrations in the media decreased at 8 and 12 h after the treatment and recovered at 24 h. Western blots revealed that eNOS and iNOS decreased in luteal cells treated with GnRH-Ag compared with controls at 4 and 8 h. These results demonstrate that suppression of luteal NO synthesis by GnRH-Ag is direct and leads to a decrease in the luteal production and release of progesterone and pregnenolone and thus suggest that GnRH could induce luteolysis in pregnant rats via NO.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , Luteal Cells/metabolism , Nitric Oxide/biosynthesis , Steroids/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Female , Immunohistochemistry , Isoenzymes/biosynthesis , Luteal Cells/drug effects , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Pregnancy , Pregnenolone/biosynthesis , Progesterone/biosynthesis , Rats , Rats, Sprague-Dawley
10.
Biol Reprod ; 69(1): 30-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12606383

ABSTRACT

Seasonal fluctuations in immune status have been documented for avian and mammalian populations. During the late summer and early fall, immune function is bolstered to help animals cope with the more physiologically demanding winter. The environmental cue for these seasonal changes is apparently decreasing photoperiod. In the present study, we determined the potential role of leptin in mediating the effect of photoperiod on cell-mediated immune responses in male mice. Leptin-deficient (ob/ob) and littermate control mice were housed for 10 wk in either a short (8L:16D) or a long (16L:8D) photoperiod beginning at 6 wk of age. After the mice were killed, immune and reproductive organs were weighed and splenocytes isolated. The proliferative and cytokine responses (interleukin [IL]-2 and IL-4) of splenocytes to the T-cell mitogen, concanavalin A (Con A; 0-40 microg/ml), were determined. Body weights were elevated and both testes and seminal vesicle weights subnormal in ob/ob mice (by ANOVA, main effect of leptin deficiency), but thymuses and spleens were of normal size. Serum leptin levels were at minimum detection limits in ob/ob mice, but leptin levels in control mice housed at 8L:16D were higher than in control mice housed at 16L:8D. The proliferative response of splenocytes from ob/ob mice to Con A was subnormal (by ANOVA, main effect of leptin deficiency), but photoperiod had no effect on this response. Production of IL-2 in splenocytes of ob/ob mice was subnormal (by ANOVA, main effect of leptin deficiency) irrespective of photoperiod, but cells from mice housed at 8L:16D (by ANOVA, main effect of photoperiod) produced more IL-2 than cells from animals housed at 16L:8D. In contrast, a leptin deficiency did not alter IL-4 production, but cells from animals (ob/ob and controls) housed at 16L:8D produced less IL-4 than cells from animals housed at 8L:16D (by ANOVA, main effect of photoperiod). The present study suggests that both photoperiod and leptin have mutually independent effects on the proliferation of lymphocytes and cytokine production profiles. The data do not provide definitive support for the hypothesis that photoperiod-induced changes in leptin secretion mediate the effects of season on immune status.


Subject(s)
Immunity , Leptin/physiology , Photoperiod , Animals , Body Weight , Cell Division , Cytokines/biosynthesis , In Vitro Techniques , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Immunological , Organ Size , Seasons , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Testis/anatomy & histology , Testis/physiology , Testosterone/blood
11.
Biol Reprod ; 66(3): 770-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11870085

ABSTRACT

Recent studies have shown that microgravity induces changes at the cellular level, including apoptosis. However, it is unknown whether microgravity affects luteal cell function. This study was performed to assess whether microgravity conditions generated by clinostat rotation induce apoptosis and affect steroidogenesis by luteal cells. Luteal cells isolated from the corpora lutea of Day 8 pregnant rats were placed in equal numbers in slide flasks (chamber slides). One slide flask was placed in the clinostat and the other served as a stationary control. At 48 h in the clinostat, whereas the levels of progesterone and total cellular protein decreased, the number of shrunken cells increased. To determine whether apoptosis occurred in shrunken cells, Comet and TUNEL assays were performed. At 48 h, the percentage of apoptotic cells in the clinostat increased compared with that in the control. To investigate how the microgravity conditions induce apoptosis, the active mitochondria in luteal cells were detected with JC-1 dye. Cells in the control consisted of many active mitochondria, which were evenly distributed throughout the cell. In contrast, cells in the clinostat displayed fewer active mitochondria, which were distributed either to the outer edge of the cell or around the nucleus. These results suggest that mitochondrial dysfunction induced by clinostat rotation could lead to apoptosis in luteal cells and suppression of progesterone production.


Subject(s)
Apoptosis , Luteal Cells/ultrastructure , Weightlessness Simulation , Animals , Culture Media, Conditioned , DNA Fragmentation , Electrophoresis, Agar Gel , Female , Fluorescent Dyes , In Situ Nick-End Labeling , Luteal Cells/metabolism , Membrane Potentials , Microscopy, Fluorescence , Mitochondria/physiology , Mitochondria/ultrastructure , Pregnancy , Progesterone/analysis , Progesterone/biosynthesis , Propidium , Proteins/analysis , Rats , Rats, Sprague-Dawley , Rotation , Time Factors , Weightlessness Simulation/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...