Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Environ Pollut ; 349: 123840, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38537797

ABSTRACT

Benzophenone-3 (BP3) is a common ingredient in personal care products (PCPs) due to its well-established effectiveness in absorbing UV radiation. Sunscreen products are among the most widely used PCPs-containing BP3 applied to the skin, resulting in significant human exposure to BP3 primarily through a dermal application. In the present work, we have tested the action of three environmentally relevant concentrations of BP3 (2, 20 and 200 µg/L) on an in vitro model of implantation of murine blastocysts and on migration ability of the human trophoblast cell line Swan 71. We showed that BP3 caused a significant reduction of blastocyst expansion and a delayed hatching in a non-monotonic way. Besides, embryos displayed a delayed attachment in the three BP3 groups, resulting in a smaller implantation area on the 6th day of culture: BP3(2) (0.32 ± 0.07 mm2); BP3(20) (0.30 ± 0.08 mm2) and BP3(200) (0.25 ± 0.06 mm2) in comparison to the control (0.42 ± 0.07 mm2). We also found a reduced migration capacity of the human first-trimester trophoblast cell line Swan 71 in a scratch assay when exposed to BP3: the lowest dose displayed a higher uncovered area (UA) at 6h when compared to the control, whereas a higher UA of the wound was observed for the three BP3 concentrations at 18 and 24 h of exposure. The changes in UA provoked by BP3 restored to normal values in the presence of flutamide, an androgen receptor (AR) inhibitor. These results indicate that a direct impairment on early embryo implantation and a defective migration of extravillous trophoblast cells through the androgen receptor pathway can be postulated as mechanisms of BP3-action on early gestation with potential impact on fetal growth.


Subject(s)
Benzophenones , Cell Movement , Embryo Implantation , Sunscreening Agents , Trophoblasts , Ultraviolet Rays , Benzophenones/toxicity , Sunscreening Agents/toxicity , Sunscreening Agents/pharmacology , Trophoblasts/drug effects , Cell Movement/drug effects , Mice , Animals , Humans , Embryo Implantation/drug effects , Blastocyst/drug effects , Female , Cell Line
2.
Front Cell Infect Microbiol ; 13: 1134471, 2023.
Article in English | MEDLINE | ID: mdl-37313339

ABSTRACT

A variety of intestinal-derived culture systems have been developed to mimic in vivo cell behavior and organization, incorporating different tissue and microenvironmental elements. Great insight into the biology of the causative agent of toxoplasmosis, Toxoplasma gondii, has been attained by using diverse in vitro cellular models. Nonetheless, there are still processes key to its transmission and persistence which remain to be elucidated, such as the mechanisms underlying its systemic dissemination and sexual differentiation both of which occur at the intestinal level. Because this event occurs in a complex and specific cellular environment (the intestine upon ingestion of infective forms, and the feline intestine, respectively), traditional reductionist in vitro cellular models fail to recreate conditions resembling in vivo physiology. The development of new biomaterials and the advances in cell culture knowledge have opened the door to a next generation of more physiologically relevant cellular models. Among them, organoids have become a valuable tool for unmasking the underlying mechanism involved in T. gondii sexual differentiation. Murine-derived intestinal organoids mimicking the biochemistry of the feline intestine have allowed the generation of pre-sexual and sexual stages of T. gondii for the first time in vitro, opening a window of opportunity to tackling these stages by "felinizing" a wide variety of animal cell cultures. Here, we reviewed intestinal in vitro and ex vivo models and discussed their strengths and limitations in the context of a quest for faithful models to in vitro emulate the biology of the enteric stages of T. gondii.


Subject(s)
Toxoplasma , Animals , Cats , Mice , Sex Differentiation , Intestines , Intestinal Mucosa , Biology
3.
Front Cell Infect Microbiol ; 13: 1082524, 2023.
Article in English | MEDLINE | ID: mdl-36968103

ABSTRACT

Chagas disease (CD) is a life-threatening illness caused by the parasite Trypanosoma cruzi (T. cruzi). With around seven million people infected worldwide and over 50,000 deaths per year, CD is a major public health issue in Latin America. The main route of transmission to humans is through a triatomine bug (vector-borne), but congenital and oral transmission have also been reported. The acute phase of CD presents mild symptoms but may develop into a long-lasting chronic illness, characterized by severely impaired cardiac, digestive, and neurological functions. The intestinal tissue appears to have a key role during oral transmission and chronic infection of CD. In this immune-privileged reservoir, dormant/quiescent parasites have been suggested to contribute to disease persistence, infection relapse, and treatment failure. However, the interaction between the intestinal epithelium and T. cruzi has not been examined in depth, in part, due to the lack of in vitro models that approximate to the biological and structural complexity of this tissue. Therefore, to understand the role played by the intestinal tissue during transmission and chronic infection, physiological models resembling the organ complexity are needed. Here we addressed this issue by establishing and characterizing adult stem cell-derived colonoid infection models that are clinically relevant for CD. 3D and 2D systems of murine intestinal organoids infected with T. cruzi Dm28c (a highly virulent strain associated with oral outbreaks) were analyzed at different time points by confocal microscopy. T. cruzi was able to invade and replicate in intestinal epithelial primary cells grown as intact organoids (3D) and monolayers (2D). The permissiveness to pathogen infection differed markedly between organoids and cell lines (primate and intestinal human cell lines). So far, this represents the first evidence of the potential that these cellular systems offer for the study of host-pathogen interactions and the discovery of effective anti-chagasic drugs.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Humans , Animals , Mice , Persistent Infection , Chagas Disease/parasitology , Intestinal Mucosa , Colon , Organoids
4.
Front Cell Infect Microbiol ; 13: 1130901, 2023.
Article in English | MEDLINE | ID: mdl-36968102

ABSTRACT

Toxoplasma gondii is a ubiquitous apicomplexan parasite that can infect virtually any warm-blooded animal. Acquired infection during pregnancy and the placental breach, is at the core of the most devastating consequences of toxoplasmosis. T. gondii can severely impact the pregnancy's outcome causing miscarriages, stillbirths, premature births, babies with hydrocephalus, microcephaly or intellectual disability, and other later onset neurological, ophthalmological or auditory diseases. To tackle T. gondii's vertical transmission, it is important to understand the mechanisms underlying host-parasite interactions at the maternal-fetal interface. Nonetheless, the complexity of the human placenta and the ethical concerns associated with its study, have narrowed the modeling of parasite vertical transmission to animal models, encompassing several unavoidable experimental limitations. Some of these difficulties have been overcome by the development of different human cell lines and a variety of primary cultures obtained from human placentas. These cellular models, though extremely valuable, have limited ability to recreate what happens in vivo. During the last decades, the development of new biomaterials and the increase in stem cell knowledge have led to the generation of more physiologically relevant in vitro models. These cell cultures incorporate new dimensions and cellular diversity, emerging as promising tools for unraveling the poorly understood T. gondii´s infection mechanisms during pregnancy. Herein, we review the state of the art of 2D and 3D cultures to approach the biology of T. gondii pertaining to vertical transmission, highlighting the challenges and experimental opportunities of these up-and-coming experimental platforms.


Subject(s)
Toxoplasma , Toxoplasmosis , Animals , Humans , Pregnancy , Female , Placenta/parasitology , Toxoplasmosis/parasitology , Infectious Disease Transmission, Vertical , Models, Animal
5.
Sci Rep ; 12(1): 14425, 2022 08 24.
Article in English | MEDLINE | ID: mdl-36002565

ABSTRACT

Inflammation is an important process for epithelial barrier protection but when uncontrolled, it can also lead to tissue damage. The nuclear factor-kappa light chain enhancer of activated B cells (NF-κB) signaling pathway is particularly relevant in the intestine, as it seems to play a dual role. Whereas NF-κB protects intestinal epithelium against various noxious stimuli, the same pathway mediates intestinal inflammatory diseases by inducing pro-inflammatory gene expression. The availability of appropriate in vitro models of the intestinal epithelium is crucial for further understanding the contribution of NF-κB in physiological and pathological processes and advancing in the development of drugs and therapies against gut diseases. Here we established, characterized, and validated three-dimensional cultures of intestinal organoids obtained from biopsies of NF-κB-RE-Luc mice. The NF-κB-RE-Luc intestinal organoids derived from different intestine regions recreated the cellular composition of the tissue and showed a reporter responsiveness similar to the in vivo murine model. When stimulated with TNF-α, jejunum-derived NF-κB-RE-Luc-reporter organoids, provided a useful model to evaluate the anti-inflammatory effects of natural and synthetic compounds. These reporter organoids are valuable tools to explore the epithelial TNF-α-induced NF-κB contribution in the small intestine, being a reliable alternative method while helping to reduce the use of laboratory animals for experimentation.


Subject(s)
NF-kappa B , Tumor Necrosis Factor-alpha , Animals , Inflammation/metabolism , Jejunum/metabolism , Mice , NF-kappa B/metabolism , Organoids/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Reprod Toxicol ; 98: 117-124, 2020 12.
Article in English | MEDLINE | ID: mdl-32956838

ABSTRACT

Understanding the effects of Bisphenol A (BPA) on early germ cell differentiation and their consequences in adult life is an area of growing interest in the field of endocrine disruption. Herein, we investigate whether perinatal exposure to BPA affects the differentiation of male germ cells in early life using a transgenic mouse expressing the GFP reporter protein under the Oct4 promoter. In this model, the expression of GFP reflects the expression of the Oct4 gene. This pluripotency gene is required to maintain the spermatogonial stem cells in an undifferentiated stage. Thus, GFP expression was used as a parameter to evaluate the effect of BPA on early germ cell development. Female pregnant transgenic mice were exposed to BPA by oral gavage, from embryonic day 5.5 to postnatal day 7 (PND7). The effects of BPA on male germ cell differentiation were evaluated at PND7, while sperm quality, testicular morphology, and protein expression of androgen receptor and proliferating cell nuclear antigen were studied at PND130. We found that perinatal/lactational exposure to BPA up-regulates the expression of Oct4-driven GFP in testicular cells at PND7. This finding suggests a higher proportion of undifferentiated spermatogonia in BPA-treated animals compared with non-exposed mice. Moreover, in adulthood, the number of spermatozoa per epididymis was reduced in those animals perinatally exposed to BPA. This work shows that developmental exposure to BPA disturbed the normal differentiation of male germ cells early in life, mainly by altering the expression of Oct4 and exerted long-lasting sequelae at the adult stage, affecting sperm count and testis.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Germ Cells/drug effects , Phenols/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Cell Differentiation , Cell Proliferation/drug effects , Female , Germ Cells/cytology , Germ Cells/growth & development , Germ Cells/metabolism , Male , Maternal-Fetal Exchange , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Receptors, Androgen/metabolism , SOXB1 Transcription Factors/genetics , Sperm Count , Sperm Motility/drug effects , Testis/drug effects , Testis/growth & development , Testis/metabolism
7.
J Mol Endocrinol ; 63(3): 187-197, 2019 10.
Article in English | MEDLINE | ID: mdl-31416050

ABSTRACT

Testicular Leydig cells (LC) are modulated by several pathways, one of them being the histaminergic system. Heme oxygenase-1 (HO-1), whose upregulation comprises the primary response to oxidative noxae, has a central homeostatic role and might dysregulate LC functions when induced. In this report, we aimed to determine how hemin, an HO-1 inducer, affects LC proliferative capacity and whether HO-1 effects on LC functions are reversible. It was also evaluated if HO-1 interacts in any way with histamine, affecting its regulatory action over LC. MA-10 and R2C cell lines and immature rat LC were used as models. Firstly, we show that after a 24-h incubation with 25 µmol/L hemin, LC proliferation is reversibly impaired by cell cycle arrest in G2/M phase, with no evidence of apoptosis induction. Even though steroid production is abrogated after a 48-h exposure to 25 µmol/L hemin, steroidogenesis can be restored to control levels in a time-dependent manner if the inducer is removed from the medium. Regarding HO-1 and histamine interaction, it is shown that hemin abrogates histamine biphasic effect on steroidogenesis and proliferation. Working with histamine receptors agonists, we elucidated that HO-1 induction affects the regulation mediated by receptor types 1, 2 and 4. In summary, HO-1 induction arrests LC functions, inhibiting steroid production and cell cycle progression. Despite their reversibility, HO-1 actions might negatively influence critical phases of LC development and differentiation affecting their function as well as other androgen-dependent organs. What's more, we have described a hitherto unknown interaction between HO-1 induction and histamine effects.


Subject(s)
Heme Oxygenase-1/metabolism , Histamine/pharmacology , Leydig Cells/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Enzyme Induction/drug effects , G2 Phase/drug effects , Hemin/pharmacology , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mice , Mitosis/drug effects , Rats, Sprague-Dawley , Steroids/biosynthesis
8.
J Biotechnol ; 301: 79-87, 2019 Aug 10.
Article in English | MEDLINE | ID: mdl-31145935

ABSTRACT

Nuclear Factor kappa B (NF-κB) is a conserved transcription factor involved in the expression of genes that are critical to inflammation and cell survival. Exposure to particular signals results in phosphorylation of NF-κB inhibitor proteins, which in turn allows NF-κB dimers to translocate to the nucleus and induce gene expression. Pathologic consequences of NF-κB activation are vast, mainly because of the pleiotropic roles that NF-κB-induced genes have on inflammation, cell proliferation and apoptosis. Thus, experimental models assessing NF-κB activation have direct screening applications for drug discovery. In this scenario, pathway-specific reporter cell systems become valuable tools to identify and elucidate the mechanism of action of novel compounds. Here, we describe the generation, characterization, and validation of human cancer epithelial reporter cell lines for functional studies of NF-κB activation by different pro- and anti-inflammatory mediators. Human lung (H460) and breast (T-47D) cancer cell lines were transfected with a pNF-κB-hrGFP plasmid which contains the GFP gene under the control of NF-κB binding elements. The pro-inflammatory cytokine TNF-α was able to activate the reporter systems in a concentration-response manner, correlating to the activation of the NF-κB signaling pathway. Finally, the reporter cell lines were validated using dexamethasone, a potent anti-inflammatory drug, a synthetic inhibitor of NF-κB (BAY 11-7082) and a new anti-cancer peptide (CIGB-552). We have established robust H460-NF-κB-hrGFP and T-47D-NF-κB-hrGFP reporter cell lines which represent a useful cancer model for primary screening and identification of compounds with anti-inflammatory activity.


Subject(s)
Breast Neoplasms/metabolism , Cell Line, Tumor/metabolism , Drug Discovery/methods , Lung Neoplasms/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Female , Humans , Models, Biological , Reproducibility of Results
9.
J Steroid Biochem Mol Biol ; 154: 53-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26151743

ABSTRACT

Oct4 is involved in regulation of pluripotency during normal development and is down-regulated during formation of postnatal reservoir of germ cells. We propose thatOct4/GFP transgenic mouse, which mimics the endogenous expression pattern of Oct4, could be used as a mammalian model to study the effects of environmental estrogens on the development of male germ cells. Oct4/GFP maturation profile was assessed during postnatal days -PND- 3, 5, 7, 10, 14 and 80, using flow cytometry. Then, we exposed pregnant mothers to 17α-ethinylestradiol (EE2) from day post coitum (dpc) 5 to PND7. Percentage of Oct4/GFP-expressing cells and levels of expression of Oct4/GPF were increased in PND7 after EE2 exposure. These observations were confirmed by analysis of GFP and endogenous Oct4 protein in the seminiferous tubules and by a reduction in epididymal sperm count in adult mice. We introduced Oct4/GFP mouse together with flow cytometry as a tool to evaluate changes in male germ cells development.


Subject(s)
Environmental Pollutants/pharmacology , Ethinyl Estradiol/pharmacology , Octamer Transcription Factor-3/physiology , Spermatozoa/drug effects , Animals , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Sperm Count , Sperm Motility
10.
J Endocrinol ; 223(3): 241-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253872

ABSTRACT

The histamine H4 receptor (HRH4), discovered only 13 years ago, is considered a promising drug target for allergy, inflammation, autoimmune disorders and cancer, as reflected by a steadily growing number of scientific publications and patent applications. Although the presence of HRH4 has been evidenced in the testis, its specific localization or its role has not been established. Herein, we sought to identify the possible involvement of HRH4 in the regulation of Leydig cell function. We first evaluated its expression in MA-10 Leydig tumor cells and then assessed the effects of two HRH4 agonists on steroidogenesis and proliferation. We found that HRH4 is functionally expressed in MA-10 cells, and that its activation leads to the inhibition of LH/human chorionic gonadotropin-induced cAMP production and StAR protein expression. Furthermore, we observed decreased cell proliferation after a 24-h HRH4 agonist treatment. We then detected for the sites of HRH4 expression in the normal rat testis, and detected HRH4 immunostaining in the Leydig cells of rats aged 7-240 days, while 21-day-old rats also presented HRH4 expression in male gametes. Finally, we evaluated the effect of HRH4 activation on the proliferation of normal progenitor and immature rat Leydig cell culture, and both proved to be susceptible to the anti-proliferative effect of HRH4 agonists. Given the importance of histamine (2-(1H-imidazol-4-yl)ethanamine) in human (patho)physiology, continued efforts are directed at elucidating the emerging properties of HRH4 and its ligands. This study reveals new sites of HRH4 expression, and should be considered in the design of selective HRH4 agonists for therapeutic purposes.


Subject(s)
Cell Proliferation , Leydig Cells/metabolism , Progesterone/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Animals , Blotting, Western , Bucladesine/pharmacology , Cell Line, Tumor , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Guanidines/pharmacology , Histamine Agonists/pharmacology , Immunohistochemistry , Indoles/pharmacology , Leydig Cells/drug effects , Male , Microscopy, Confocal , Oximes/pharmacology , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, Histamine H4 , Testis/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology
11.
Radiat Environ Biophys ; 53(3): 611-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24811726

ABSTRACT

We have previously reported the radioprotective effect of propylthiouracil (PTU) on thyroid cells. The aim of the present study was to analyze whether tumor cells and normal cells demonstrate the same response to PTU. Human colon carcinoma cells were irradiated with γ-irradiation with or without PTU. We evaluated the clonogenic survival, intracellular reactive oxygen species levels, catalase, superoxide dismutase and glutathione peroxidase activities, and apoptosis by nuclear cell morphology and caspase-3 activity assays. Cyclic AMP (cAMP) levels were measured by radioimmunoassay. PTU treatment increased surviving cell fraction at 2 Gy (SF2) from 56.9 ± 3.6 in controls to 75.0 ± 3.5 (p < 0.05) and diminished radiation-induced apoptosis. In addition, we observed that the level of antioxidant enzymes' activity was increased in cells treated with PTU. Moreover, pretreatment with PTU increased intracellular levels of cAMP. Forskolin (p < 0.01) and dibutyryl cAMP (p < 0.05) mimicked the effect of PTU on SF2. Co-treatment with H89, an inhibitor of protein kinase A, abolished the radioprotective effect of PTU. PTU reduces the toxicity of ionizing radiation by increasing cAMP levels and also possibly through a reduction in apoptosis levels and in radiation-induced oxidative stress damage. We therefore conclude that PTU protects both normal and cancer cells during exposure to radiation in conditions mimicking the radiotherapy.


Subject(s)
Antithyroid Agents/pharmacology , Colonic Neoplasms/pathology , Gamma Rays/adverse effects , Propylthiouracil/pharmacology , Radiation-Protective Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cyclic AMP/metabolism , Humans , Oxidative Stress/drug effects , Oxidative Stress/radiation effects
12.
J Endocrinol ; 221(1): 15-28, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424290

ABSTRACT

Histamine (HA) is a neurotransmitter synthesized in most mammalian tissues exclusively by histidine decarboxylase enzyme. Among the plethora of actions mediated by HA, the modulatory effects on steroidogenesis and proliferation in Leydig cells (LCs) have been described recently. To determine whether the effects on LCs reported could be extrapolated to all steroidogenic systems, in this study, we assessed the effect of this amine on adrenal proliferation and steroidogenesis, using two adrenocortical cell lines as experimental models, murine Y1 cells and human NCI-H295R cells. Even when steroidogenesis was not modified by HA in adrenocortical cells, the biogenic amine inhibited the proliferation of H295R cells. This action was mediated by the activation of HRH1 subtype and an increase in the production of inositol phosphates as second messengers, causing cell-cycle arrest in the G2/M phase. These results indicate a new role for HA in the proliferation of human adrenocortical cells that could contribute to a better understanding of tumor pathology as well as to the development of new therapeutic agents.


Subject(s)
Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Cell Proliferation , Histamine/metabolism , Steroids/metabolism , Animals , Cell Line , G2 Phase Cell Cycle Checkpoints , Humans , M Phase Cell Cycle Checkpoints , Mice
13.
J Dermatol Sci ; 72(3): 252-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23999004

ABSTRACT

BACKGROUND: Functional presence of histamine H4 receptor (H4R) was demonstrated in human melanoma cell lines and biopsies. OBJECTIVE: The purposes of this work were to investigate signal transduction pathways and biological responses triggered by the activation of H4R in human primary (WM35) and metastatic (M1/15) melanoma cell lines and to evaluate the in vivo antitumor activity of histamine (HA) and clozapine (CLZ) on human M1/15 melanoma xenografts. METHODS: Clonogenic assay, incorporation of BrdU, cell cycle distribution, phosphorylation levels of ERK1/2 and cAMP production were evaluated in vitro. An experimental human melanoma model was developed into athymic nude mice. Tumor growth, survival and histochemical studies were performed in order to investigate the expression levels of H4R, HA, PCNA, mitotic index (MI), and angiogenesis. RESULTS: The results indicate that H4R agonists inhibited forskolin-induced cAMP levels only in M1/15 cells while increased phosphorylation levels of ERK1/2 and decreased proliferation in both cell types. In vivo studies show that HA and CLZ (1mgkg(-1), sc) significantly increased median survival and decreased tumor volume. These effects were associated to a reduction in MI, in the expression of proliferation marker and in intratumoral neovascularization. CONCLUSIONS: We conclude that HA and CLZ exhibit an antitumoral effect in vitro and in vivo on human melanoma, suggesting the therapeutic potential of these compounds for the treatment of malignant melanoma.


Subject(s)
Clozapine/therapeutic use , Histamine/therapeutic use , Melanoma, Experimental/drug therapy , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clozapine/pharmacology , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine/pharmacology , Humans , Male , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Receptors, Histamine , Receptors, Histamine H4 , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
Radiat Res ; 179(3): 352-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23398355

ABSTRACT

Many epidemiologic studies have shown that the exposure to high external radiation doses increases thyroid neoplastic frequency, especially when given during childhood or adolescence. The use of radioprotective drugs may decrease the damage caused by radiation therapy and therefore could be useful to prevent the development of thyroid tumors. The aim of this study was to investigate the possible application of 6-propyl-2-thiouracil (PTU) as a radioprotector in the thyroid gland. Rat thyroid epithelial cells (FRTL-5) were exposed to different doses of γ irradiation with or without the addition of PTU, methimazole (MMI), reduced glutathione (GSH) and perchlorate (KClO4). Radiation response was analyzed by clonogenic survival assay. Cyclic AMP (cAMP) levels were measured by radioimmunoassay (RIA). Apoptosis was quantified by nuclear cell morphology and caspase 3 activity assays. Intracellular reactive oxygen species (ROS) levels were measured using the fluorescent dye 2',7'-dichlorofluorescein-diacetate. Catalase, superoxide dismutase and glutathione peroxidase activities were also determined. Pretreatment with PTU, MMI and GSH prior to irradiation significantly increased the surviving cell fraction (SF) at 2 Gy (P < 0.05), while no effect was observed with KClO4. An increase in extracellular levels of cAMP was found only in PTU treated cells in a dose and time-dependent manner. Cells incubated with agents that stimulate cAMP (forskolin and dibutyril cAMP) mimicked the effect of PTU on SF. Moreover, pretreatment with the inhibitor of protein kinase A, H-89, abolished the radioprotective effect of PTU. PTU treatment diminished radiation-induced apoptosis and protected cells against radiation-induced ROS elevation and suppression of the antioxidant enzyme's activity. PTU was found to radioprotect normal thyroid cells through cAMP elevation and reduction in both apoptosis and radiation-induced oxidative stress damage.


Subject(s)
Propylthiouracil/pharmacology , Radiation-Protective Agents/pharmacology , Thyroid Gland/drug effects , Thyroid Gland/radiation effects , Animals , Caspase 3/metabolism , Cell Line , Cyclic AMP/metabolism , Oxidative Stress/drug effects , Radioimmunoassay , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Thyroid Gland/metabolism
15.
Biol Reprod ; 87(6): 150, 2012 Jun.
Article in English | MEDLINE | ID: mdl-23077168

ABSTRACT

Mast cells (MC) occur normally in the testis with a species-specific distribution, yet their precise role remains unclear. Testicular MC express histidine decarboxylase (HDC), the unique enzyme responsible for histamine (HA) generation. Evidence to date supports a role for HA as a local regulator of steroidogenesis via functional H1 and H2 receptor subtypes (HRH1 and HRH2, respectively) present in Leydig cells. Given that HA is a well-known modulator of physiological and pathological proliferation in many different cell types, we aimed in the present study to evaluate whether HA might contribute to the regulation of Leydig cell number as well as to the control of androgen production. Herein, we demonstrate, to our knowledge for the first time, that MA-10 Leydig tumor cells, but not normal immature Leydig cells (ILC), exhibit a proliferative response upon stimulation with HA that involves HRH2 activation, transient elevation of cAMP levels, and increased extracellular signal-regulated kinase (ERK) phosphorylation. Our results also reveal that MA-10 cells show significantly heightened HDC expression compared to normal ILC or whole-testicular lysate and that inhibition of HDC activity decreases MA-10 cell proliferation, suggesting a possible correlation between autocrine overproduction of HA and abnormally increased proliferation in Leydig cells. The facts that germ cells are also both source and target of HA and that multiple testicular cells are susceptible to HA action underline the importance of the present study, which we hope will serve as a first step for further research into regulation of non-MC-related HDC expression within the testis and its significance for testicular function.


Subject(s)
Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine/metabolism , Leydig Cell Tumor/metabolism , Leydig Cells/metabolism , Receptors, Histamine H2/metabolism , Second Messenger Systems , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Histamine Agonists/metabolism , Histamine Agonists/pharmacology , Histidine Decarboxylase/antagonists & inhibitors , Histidine Decarboxylase/biosynthesis , Histidine Decarboxylase/metabolism , Leydig Cell Tumor/drug therapy , Leydig Cell Tumor/enzymology , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/enzymology , Male , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Histamine H2/chemistry , Second Messenger Systems/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
16.
Biochim Biophys Acta ; 1810(12): 1212-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21871537

ABSTRACT

BACKGROUND: Amylase is synthesized in submandibular glands (SMG) and released into the oral cavity to degrade carbohydrates in the mouth. Bitter taste receptors (T2R) belong to the G-protein coupled receptor (GPCR) family and are expressed in the taste cells and also in the digestive tract. METHODS: The activity of amylase secreted by murine SMG was measured, detecting maltose by Bernfeld's method. Amylase and T2R6 were detected by imunohistochemistry and Western blot. The expression of Ggustducin, Gi, and phospholipase Cß2 was also studied by Western blot. cAMP levels were measured by radioimmunoassay and inositol monophosphate production was quantified by ELISA. RESULTS: Theophylline, denatonium and cycloheximide exerted a dose-dependent inhibition on amylase secretion. This effect was reverted by preincubating SMG with an anti-Gαi antibody. cAMP production was increased by the same compounds, an effect that was also abrogated by an anti-Gαi antibody. Bitter compounds reduced inositol monophosphate formation in SMG and H-89, a protein kinase A inhibitor, reverted this action, revealing that this protein kinase down regulates phospholipase C activity. GENERAL SIGNIFICANCE: We demonstrated that theophylline, denatonium and cycloheximide inhibit salivary amylase secretion, activating an intracellular signaling pathway that involves cAMP and phospholipase C, that cross talks via protein kinase A.


Subject(s)
Amylases/metabolism , Signal Transduction , Submandibular Gland/enzymology , Animals , Blotting, Western , Cyclic AMP/metabolism , Immunohistochemistry , Mice , Mice, Inbred BALB C , Submandibular Gland/metabolism
17.
Mol Cell Endocrinol ; 332(1-2): 253-60, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-21075169

ABSTRACT

Cumulative evidence demonstrated effective downstream metabolism of pregnenolone in renal tissue. The aim of this study was to evaluate the expression and functional activity of cytochrome P450 side chain cleavage enzyme (CYP11A1), which converts cholesterol into pregnenolone, in adult rat kidney. Immunohistochemical labeling for CYP11A1 was observed in renal cortex and medulla, on structures identified as distal convoluted tubule and thick ascending limb of Henle's loop, respectively. Immunoblotting analysis corroborated the renal expression of the protein in inner mitochondrial membrane fractions. The incubation of isolated mitochondria with the membrane-permeant cholesterol analogue 22R-hydroxycholesterol resulted in efficient formation of pregnenolone, the immediate precursor for the synthesis of all the steroid hormones. The low progesterone production rate observed in these experiments suggested a poor activity of 3ß-hydroxysteroid dehydrogenase enzyme in renal mitochondria. The steroidogenic acute regulatory protein (StAR), involved in the mitochondrial import of cholesterol, was detected in renal tissue at both mRNA and protein level. Immunostaining for StAR showed similar distribution to that observed for CYP11A1. The expression of StAR and CYP11A1 was found to be higher in medulla than in cortex. This enhanced expression of steroidogenesis-related proteins correlated with a greater pregnenolone synthesis rate and higher steroid hormones tissular content measured in medulla. In conclusion, we have established the expression and localization of StAR and CYP11A1 protein, the ability of synthesizing pregnenolone and a region-specific content of sex hormones in the adult rat kidney. These data clearly show that the kidney is a steroid hormones synthesizing organ. It is proposed that the existence in the kidney of complete steroidogenic machinery would respond to a physiological significance.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme/metabolism , Kidney/enzymology , Animals , Carrier Proteins/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Kidney/anatomy & histology , Male , Mitochondrial Membranes/enzymology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnenolone/biosynthesis , Progesterone/biosynthesis , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Testosterone/metabolism
18.
J Endocrinol ; 203(1): 155-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19648213

ABSTRACT

In the present study, we demonstrate the expression of heme oxygenase (HO) isozymes, HO-1 and HO-2 (listed as HMOX1 and HMOX2 in the MGI Database), in MA-10 Leydig tumor cells and its effect on steroidogenesis. The well-known HO inducer, hemin, increased both HO-1 and HO-2 protein levels and HO-specific activity. Induction of HO by hemin inhibited basal, hCG-, and dibutyryl cAMP (db-cAMP)-induced steroidogenesis in a reversible way. When we studied the effect of HO isozymes along the steroid synthesis, we found that steroidogenic acute regulatory protein levels were decreased, and the conversion of cholesterol to pregnenolone was inhibited by hemin treatment, with no changes in the content of cholesterol side-chain cleavage enzyme (P450scc). hCG and db-cAMP also stimulated the expression of HO-1 and HO-2, and HO enzymatic activity in MA-10 cells. Basal and hCG-stimulated testosterone synthesis was also inhibited by hemin in rat normal Leydig cells. Taken together, these results suggest that: i) at least one of HO products (presumably carbon monoxide) inhibits cholesterol transport to the inner mitochondrial membrane and Leydig cell steroidogenesis by binding to the heme group of the cytochrome P450 enzymes, in a similar way as we described for nitric oxide, and ii) hCG stimulation results in the induction of an antioxidant enzymatic system (HO) acting as a cytoprotective mechanism in Leydig cells, as already demonstrated in the adrenal gland.


Subject(s)
Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Leydig Cells/enzymology , Testosterone/biosynthesis , Animals , Carbon Monoxide/metabolism , Cell Line, Tumor , Chorionic Gonadotropin/metabolism , Dibutyryl Cyclic GMP , Isoenzymes/metabolism , Male , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley
19.
Biol Reprod ; 80(1): 144-52, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18768916

ABSTRACT

This study was conducted to shed light on the so far unexplored intracellular mechanisms underlying negative modulation of Leydig cell steroidogenesis by histamine (HA). Using the MA-10 cell line and highly purified rat Leydig cells as experimental models, we examined the effect of the amine on biochemical steps known to be modulated by HA or involved in LH/hCG action. In agreement with previous findings, HA at 10 microM showed a potent inhibitory effect on hCG-stimulated steroid synthesis, regardless of the gonadotropin concentration used. Moreover, HA decreased not only LH/hCG-induced cAMP production but also steroid synthesis stimulated by the permeable cAMP analog dibutyryl cAMP (db-cAMP). Considering the post-cAMP sites of HA action, it is shown herein that HA markedly inhibited db-cAMP-stimulated steroidogenic acute regulatory (STAR) protein expression, as well as steps catalyzed by P450-dependent enzymes, mainly the conversion of cholesterol to pregnenolone by cholesterol side-chain cleavage enzyme (CYP11A). The antisteroidogenic action of HA was blocked by addition of the phospholipase C (PLC) inhibitor U73122, and HA significantly augmented inositol triphosphate (IP3) production, suggesting a major role for the PLC/IP3 pathway in HA-induced inhibition of Leydig cell function. Finally, HA increased nitric oxide synthase (NOS) activity, and the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) markedly attenuated the effect of the amine on steroid synthesis. On the basis of our findings, HA antagonizes the gonadotropin action in Leydig cells at steps before and after cAMP formation. NOS activation is the main intracellular mechanism by which HA exerts its antisteroidogenic effects.


Subject(s)
Histamine/pharmacology , Leydig Cells/metabolism , Nitric Oxide Synthase Type II/metabolism , Progesterone/biosynthesis , Receptors, Histamine/metabolism , Testosterone/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Chorionic Gonadotropin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Inositol Phosphates/metabolism , Leydig Cells/drug effects , Leydig Cells/enzymology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/biosynthesis , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
20.
Biochim Biophys Acta ; 1782(2): 82-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18078830

ABSTRACT

Macrophages (Mps) are essential cellular components of the innate immune system. They are released from the bone marrow as immature monocytes and after circulating in the blood stream, migrate into tissues to undergo final differentiation into resident Mps. In general terms Mps behavior in breast tumors, was described as being either for or against tumor growth. Under certain well defined circumstances Mps are able to kill cells in two ways: direct tumor cytotoxicity or antibody dependent cytotoxicity. We had previously demonstrated that peritoneal Mps from LMM3 mammary tumor bearing mice (TMps) enhanced in vivo the LMM3 induced angiogenesis, promoting tumor growth while Mps from normal BALB/c mice (NMps) did not. In this work, we demonstrate that Mps, expressing functional muscarinic acetylcholine receptors, are able to proliferate in vitro in response to the muscarinic agonist carbachol. These peritoneal cells use two distinct metabolic pathways: TMps are primed by tumor presence and they proliferate mainly by activating arginase pathway and by producing high levels of prostaglandin E(2) via M(1)-M(3) receptors activation. In NMps, carbachol stimulates M(2) receptors function, triggering protein kinase C activity and induces moderate prostaglandin E(2) liberation via M(1) receptor.


Subject(s)
Adenocarcinoma/pathology , Cell Proliferation , Macrophages, Peritoneal/metabolism , Mammary Neoplasms, Experimental/pathology , Receptors, Muscarinic/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Animals , Arginase/physiology , Carbachol/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/pathology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation/pathology , Prostaglandin-Endoperoxide Synthases/physiology , Protein Kinase C/physiology , Receptors, Muscarinic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...