Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Am J Physiol Cell Physiol ; 323(2): C322-C332, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35704696

ABSTRACT

The increasing amount of data points to the circadian timing system as an essential part of processes regulating androgen homeostasis. However, the relationship between stress response, timekeeping-, and steroidogenesis-related systems is unexplored. Here, the stress-response of the testosterone-producing rat Leydig cells depending on the time of stressful events was studied. The study analyzes the effects of 3-h immobilization (IMO) applied at different periods during the day. The IMO performed once [1 time immobilization stress (1×IMO)] or repeated in 10 consecutive days [10 time repeated immobilization stress (10×IMO)]. Both types of IMO increased corticosterone and decreased testosterone blood level. However, the effect of 10×IMO occurring in the active phase on blood testosterone was less pronounced. This is related to different sensitivity to IMO-events depending on the diurnal time. Most steroidogenesis-related genes [gene encoding luteinizing hormone/choriogonadotropin receptor (Lhcgr), gene encoding cytochrome P450, family 11, subfamily a, polypeptide 1 (Cyp11a1), gene encoding hydroxy-δ-5-steroid dehydrogenase, 3 ß- and steroid δ-isomerase 1 (Hsd3b1/2), and gene encoding cytochrome P450, family 17, subfamily a, polypeptide 1 (Cyp17a1)] were downregulated in the inactive phase but unchanged or even upregulated in the active phase of the day. Both types of IMO stimulated the expression of clock elements gene encoding aryl hydrocarbon receptor nuclear translocator-like (Bmal1)/aryl hydrocarbon receptor nuclear translocator-like (BMAL1), gene encoding period circadian regulator 1 (Per1)/period circadian regulator 1 (PER1) regardless of the day's stage and reduced gene encoding nuclear receptor subfamily 1, group D, member 1 (Rev-erba) in the inactive phase. The principal component analysis (PCA) confirmed a major shift, for both IMO-types, in the transcription of the genes across the passive/active stage. Further, 10×IMO changed a diurnal pattern of the glucocorticoid receptor [gene encoding nuclear receptor subfamily 3, group C, member 1 (Nr3c1)/nuclear receptor subfamily 3, group C, member 1 (GR)] expression, whereas the observed time-dependent IMO-response of the Leydig cells correlated with different corticosterone engagements. Altogether, the Leydig cell's stress response depends on the daytime of the stressful event, emphasizing the importance of the circadian system in supporting androgen homeostasis and male fertility.


Subject(s)
Androgens , Leydig Cells , ARNTL Transcription Factors/metabolism , Androgens/metabolism , Androgens/pharmacology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/pharmacology , Corticosterone/pharmacology , Leydig Cells/metabolism , Male , Rats , Rats, Wistar , Testosterone/pharmacology
2.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34071734

ABSTRACT

Here, we study possible mechanisms of (in/sub)fertility related to the acute or repeated psychological stresses (the most common stresses in human society) by following the transcriptional profile of 22 mitochondrial dynamics/function markers and 22 signaling molecules regulating both mitochondrial dynamics and spermatozoa number/functionality. An in vivo study mimicking acute (once for 3 h) and repeated (3 h for 10 consecutive days) psychophysical stress was performed on adult rats. The analysis of hormones, the number/functionality of spermatozoa, and 44 transcriptional markers were performed on individual samples from up to 12 animals per group. Results showed that both types of stress reduced spermatozoa functionality (acute by 4.4-fold, repeated by 3.3-fold) and ATP production (acute by 2.3-fold, repeated by 14.5-fold), while only repeated stress reduces the number of spermatozoa (1.9-fold). Stress significantly disturbed transcription of 34-out-of-44 markers (77%). Mitochondrial dynamics and functionality markers: 18-out-of-22 =>82% (mitochondrial-biogenesis-markers ->6-out-of-8 =>75%; mitochondrial-fusion-markers ->3-out-of-3 =>100%; mitochondrial-fission-markers ->1-out-of-2 =>50%; mitochondrial-autophagy-markers ->3-out-of-3 =>100%; mitochondrial-functionality-markers ->5-out-of-6 =>83%). Markers of signaling pathways regulating both mitochondrial dynamics/functionality and spermatozoa number/functionality important for male (in/sub)fertility ->16-out-of-22 =>73% (cAMP-signaling-markers ->8-out-of-12 =>67%; MAPK-signaling-markers ->8-out-of-10 =>80%). Accordingly, stress-triggered changes of transcriptional profile of mitochondrial dynamics/functionality markers as well as signaling molecules regulating both mitochondrial dynamics and spermatozoa number and functionality represent adaptive mechanisms.


Subject(s)
Biomarkers , Mitochondria/physiology , Mitochondrial Dynamics/physiology , Signal Transduction , Sperm Count , Spermatozoa/physiology , Adenosine Triphosphate/blood , Adenosine Triphosphate/metabolism , Animals , Cyclic AMP/metabolism , Gene Expression Profiling , Hormones/blood , Hormones/metabolism , Male , Models, Biological , Rats , Stress, Psychological
3.
Biol Reprod ; 100(5): 1406-1415, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30722003

ABSTRACT

In mammals, circadian clock regulates concentration of many reproductive hormones including testosterone. Previously, we characterized pattern of circadian transcription of core clock genes in testosterone-producing Leydig cells. Here, the potential role of luteinizing hormone receptor (LHR)-cAMP signaling in synchronization of Leydig cell's circadian clock and rhythmic testosterone production were examined. Results showed that activation of LHR-cAMP signaling in primary rat Leydig cell culture increased Star/STAR and changed expression of many clock genes (upregulated Per1/PER1, Dec1/2, and Rorb, and downregulated Bmal1 and Rev-erba/b). Inhibition of protein kinase A prevented LHR-triggered increase in transcription of Per1 and Dec1. Effect of stimulated LHR-cAMP signaling on Leydig cell's clock transcription was also confirmed in vivo, using rats treated with single hCG injection. To analyze in vivo effect of low LH-cAMP activity on rhythmical Leydig cell function, rats with experimental hypogonadotropic hypogonadism were used. Characteristics of hypogonadal rats were decreased LH and testosterone secretion without circadian fluctuation; in Leydig cells decreased arrhythmic cAMP and transcription of steroidogenic genes (Cyp11a1 and Cyp17a1) were observed, while decreased Star/STAR expression retains circadian pattern. However, expression of clock genes, despite changes in transcription levels (increased Bmal1, Per2, Cry1, Cry2, Rora, Rorb, Rev-erba/b/REV-ERBB, Dec1, Csnk1e, and decreased Npas2 and PER1) kept circadian patterns observed in control groups. Altogether, the results strengthened the hypothesis about role of LH-cAMP signaling as synchronizer of Leydig cell's clock. However, clock in Leydig cells is not sufficient to sustain rhythmicity of testosterone production in absence of rhythmic activity of LH-cAMP signaling.


Subject(s)
Circadian Rhythm/physiology , Leydig Cells/physiology , Luteinizing Hormone/metabolism , Signal Transduction/physiology , Testosterone/metabolism , Animals , Cells, Cultured , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Cyclic AMP/physiology , Gene Expression Regulation/drug effects , Luteinizing Hormone/genetics , Male , Promethazine/administration & dosage , Promethazine/pharmacology , Rats , Rats, Wistar
4.
Biol Reprod ; 100(1): 253-267, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30084987

ABSTRACT

Controlled changes in mitochondrial biogenesis and morphology are required for cell survival and homeostasis, but the molecular mechanisms are largely unknown. Here, male and female prepubertal mice (P21) with insulin and IGF1 receptors deletions in steroidogenic tissues (Insr/Igf1r-DKO) were used to investigate transcription of the key regulators of mitochondrial biogenesis (Ppargc1a, Ppargc1b, Pparg, Nrf1, Tfam) and architecture in Leydig cells, ovaries, and adrenals. Results showed that the expression of PGC1, a master regulator of mitochondrial biogenesis and integrator of environmental signals, and its downstream target Tfam, significantly decreased in androgen-producing Leydig cells. This is followed by reduction of Mtnd1, a mitochondrial DNA encoded transcript whose core subunit belongs to the minimal assembly required for catalysis. The same markers remained unchanged in ovaries. In contrast, in adrenals, the pattern of transcripts for mitochondrial biogenesis markers was the same in both sexes, but opposite from that observed in Leydig cells. The level of transcripts for markers of mitochondrial architecture (Mfn1, Mfn2) significantly increased in Leydig cells from Insr/Igf1r-DKO, but not in ovaries. This was followed by mitochondrial morphology disturbance, suggesting that the mitochondrial phase of steroidogenesis could be affected. Indeed, basal and pregnenolone stimulated progesterone productions in the mitochondria of Leydig cells from Insr/Igf1r-DKO decreased more than androgen production, and were barely detectable. Our results are the first to show that INSR/IGF1R are important for mitochondrial biogenesis in gonadal steroidogenic cells of prepubertal males, but not females and they serve as important regulators of mitochondrial architecture and biogenesis markers in Leydig cells.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Leydig Cells/drug effects , Mitochondria/drug effects , Organelle Biogenesis , Testis/drug effects , Animals , Biomarkers/metabolism , Cells, Cultured , Female , Gonadal Steroid Hormones/metabolism , Leydig Cells/metabolism , Male , Mice , Mice, Transgenic , Mitochondria/physiology , Ovary/drug effects , Ovary/metabolism , Sexual Maturation/drug effects , Sexual Maturation/physiology , Signal Transduction/drug effects , Testis/cytology , Testis/metabolism
5.
FASEB J ; 32(6): 3321-3335, 2018 06.
Article in English | MEDLINE | ID: mdl-29401624

ABSTRACT

The insulin family of growth factors (insulin, IGF1, and IGF2) are critical in sex determination, adrenal differentiation, and testicular function. Notably, the IGF system has been reported to mediate the proliferation of steroidogenic cells. However, the precise role and contribution of the membrane receptors mediating those effects, namely, insulin receptor (INSR) and type-I insulin-like growth factor receptor (IGF1R), have not, to our knowledge, been investigated. We show here that specific deletion of both Insr and Igf1r in steroidogenic cells in mice leads to severe alterations of adrenocortical and testicular development. Double-mutant mice display drastic size reduction of both adrenocortex and testes, with impaired corticosterone, testosterone, and sperm production. Detailed developmental analysis of the testes revealed that fetal Leydig cell (LC) function is normal, but there is a failure of adult LC maturation and steroidogenic function associated with accumulation of progenitor LCs (PLCs). Cell-lineage tracing revealed PLC enrichment is secondary to Insr and Igf1r deletion in differentiated adult LCs, suggesting a feedback mechanism between cells at different steps of differentiation. Taken together, these data reveal the cell-autonomous and nonautonomous roles of the IGF system for proper development and maintenance of steroidogenic lineages.-Neirijnck, Y., Calvel, P., Kilcoyne, K. R., Kühne, F., Stévant, I., Griffeth, R. J., Pitetti, J.-L., Andric, S. A., Hu, M.-C., Pralong, F., Smith, L. B., Nef, S. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells.


Subject(s)
Cell Differentiation , Leydig Cells/metabolism , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Stem Cells/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Animals , Corticosterone/genetics , Corticosterone/metabolism , Leydig Cells/cytology , Male , Mice , Mice, Knockout , Receptor, Insulin/genetics , Receptors, Somatomedin/genetics , Stem Cells/cytology , Testosterone/genetics , Testosterone/metabolism
6.
Adv Physiol Educ ; 41(3): 405-414, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28679579

ABSTRACT

In response to the Bologna Declaration and contemporary trends in Animal Physiology education, the Animal Physiology course at the Faculty of Sciences, University of Novi Sad, Serbia, has evolved over a 12-yr period (2001-2012): from a classical two-semester course toward a one-semester course utilizing computer simulations of animal experiments, continual assessment, lectures, and an optional oral exam. This paper presents an overview of student achievement, the impact of reforms on learning outcomes, and lessons that we as educators learned during this process. The reforms had a positive impact on the percentage of students who completed the course within the same academic year. In addition, the percentage of students who completed the practical exam increased from 54% to >95% following the transition to a Bologna-based approach. However, average final grades declined from 8.0 to 6.8 over the same period. Students also appear reluctant to take the optional oral exam, and 82-91% of students were satisfied with the lower final grade obtained from only assessments and tests administered during the semester. In our endeavor to achieve learning outcomes set during the pre-Bologna period, while adopting contemporary teaching approaches, we sought to increase students' motivation to strive toward better performance, while ensuring that the increased quantity of students who complete the course is coupled with increased quality of education and a more in-depth understanding of animal physiology.


Subject(s)
Physiology/education , Teaching/standards , Animals , Educational Measurement , Learning , Motivation , Students/psychology , Teaching/trends
7.
Biochim Biophys Acta ; 1853(10 Pt A): 2217-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26036344

ABSTRACT

Adaptability to stress is a fundamental prerequisite for survival. Mitochondria are a key component of the stress response in all cells. For steroid-hormones-producing cells, including also Leydig cells of testes, the mitochondria are a key control point for the steroid biosynthesis and regulation. However, the mitochondrial biogenesis in steroidogenic cells has never been explored. Here we show that increased mitochondrial biogenesis is the adaptive response of testosterone-producing Leydig cells from stressed rats. All markers of mitochondrial biogenesis together with transcription factors and related kinases are up-regulated in Leydig cells from rats exposed to repeated psychophysical stress. This is followed with increased mitochondrial mass. The expression of PGC1, master regulator of mitochondrial biogenesis and integrator of environmental signals, is stimulated by cAMP-PRKA, cGMP, and ß-adrenergic receptors. Accordingly, stress-triggered mitochondrial biogenesis represents an adaptive mechanism and does not only correlate with but also is an essential for testosterone production, being both events depend on the same regulators. Here we propose that all events induced by acute stress, the most common stress in human society, provoke adaptive response of testosterone-producing Leydig cells and activate PGC1, a protein required to make new mitochondria but also protector against the oxidative damage. Given the importance of mitochondria for steroid hormones production and stress response, as well as the role of steroid hormones in stress response and metabolic syndrome, we anticipate our result to be a starting point for more investigations since stress is a constant factor in life and has become one of the most significant health problems in modern societies.


Subject(s)
Leydig Cells/metabolism , Mitochondria/metabolism , Stress, Psychological/metabolism , Testosterone/biosynthesis , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Humans , Leydig Cells/ultrastructure , Male , Mitochondria/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Stress, Psychological/pathology , Transcription Factors/metabolism
8.
Mol Hum Reprod ; 20(1): 77-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23894150

ABSTRACT

The molecular mechanism of stress-associated reproductive dysfunction is complex and largely unknown. This study was designed to systematically analyze molecular effects of systemic in vivo blockade of α1-adrenergic receptors (α1-ADRs) on stress-induced disturbance of cAMP/cGMP signaling in testosterone-producing Leydig cells using the following parameters (i) level of circulating stress hormones, LH and testosterone; (ii) level of main molecular markers of Leydig cell functionality (testosterone, Insl3, cAMP); (iii) expression of cAMP signaling (cAMP 'producers'/'effectors'/'removers') and (iv) expression of NO-cGMP signaling (NO-cGMP 'producers'/'effectors'/'removers'). The results showed that oral administration of α1-ADR blocker before stress increased cGMP and diminished stress-reduced cAMP production in Leydig cells. In the same cells, stress-induced effects on cAMP/cGMP signaling pathways elements were changed. Sustained in vivo α1-ADR blockade completely abolished stress-increased transcription of most abundantly expressed phosphodiesterase that remove cAMP (Pde4b) and potentiated stress-increased expression of PRKA, the main stimulator of Leydig cell steroidogenesis. In the same Leydig cells, stress-decreased NOS3 expression was abolished, while stress-increased GUCY1 (cGMP 'producer') and PRKG1 (cGMP 'effector') were potentiated. It is possible that all molecules mentioned could contribute, at least in part, in recovery of Leydig cell testosterone production. Presented data provide new role of α1-ADRs in stress-triggered disturbance of cAMP/cGMP signaling, and new molecular insights into the relationship between stress and mammalian reproduction. Regardless of whether the effects of α1-blocker + stress are direct or indirect, the results are important in terms of human reproductive health and the wide use of α1-ADR antagonists, alone or in combination, to treat post-traumatic stress disorders, hypertension, benign prostatic hyperplasia symptoms and potential drugs for prostate cancer prevention/treatment.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Leydig Cells/metabolism , Stress, Physiological/drug effects , AMP-Activated Protein Kinases/biosynthesis , Animals , Corticosterone/blood , Cyclic AMP/biosynthesis , Cyclic GMP/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type I/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Doxazosin/pharmacology , Epinephrine/blood , Guanylate Cyclase/biosynthesis , Insulin/biosynthesis , Luteinizing Hormone/blood , Male , Nitric Oxide Synthase Type III/biosynthesis , Proteins , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Cytoplasmic and Nuclear/biosynthesis , Signal Transduction , Soluble Guanylyl Cyclase , Testosterone/biosynthesis , Testosterone/blood
9.
Am J Physiol Endocrinol Metab ; 304(1): E51-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23149620

ABSTRACT

The stress-induced initiation of proapoptotic signaling in Leydig cells is relatively well defined, but the duration of this signaling and the mechanism(s) involved in opposing the stress responses have not been addressed. In this study, immobilization stress (IMO) was applied for 2 h daily, and animals were euthanized immediately after the first (IMO1), second (IMO2), and 10th (IMO10) sessions. In IMO1 and IMO2 rats, serum corticosterone and adrenaline were elevated, whereas serum androgens and mRNA transcription of insulin-like factor-3 in Leydig cells were inhibited. Reduced oxygen consumption and the mitochondrial membrane potential coupled with a leak of cytochrome c from mitochondria and increased caspase-9 expression, caspase-3 activity, and number of apoptotic Leydig cells was also observed. Corticosterone and adrenaline were also elevated in IMO10 rats but were accompanied with a partial recovery of androgen secretion and normalization of insulin-like factor-3 transcription coupled with increased cytochrome c expression, abolition of proapoptotic signaling, and normalization of the apoptotic events. Blockade of intratesticular glucocorticoid receptors diminished proapoptotic effects without affecting antiapoptotic effects, whereas blockade of intratesticular α(1)-adrenergic receptors diminished the antiapoptotic effects without affecting proapoptotic effects. These results confirmed a critical role of glucocorticoids in mitochondria-dependent apoptosis and showed for the first time the relevance of stress-induced upregulation of α(1)-adrenergic receptor expression in cell apoptotic resistance to repetitive IMOs. The opposite role of two hormones in control of the apoptotic rate in Leydig cells also provides a rationale for a partial recovery of androgen production in chronically stressed animals.


Subject(s)
Apoptosis , Glucocorticoids/pharmacology , Leydig Cells/physiology , Receptors, Adrenergic, alpha-1/physiology , Stress, Psychological , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Cells, Cultured , Corticosterone/blood , Corticosterone/metabolism , Corticosterone/pharmacology , Corticosterone/physiology , Drug Antagonism , Glucocorticoids/physiology , Immobilization/psychology , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Psychological/blood , Stress, Psychological/genetics , Stress, Psychological/metabolism
10.
Am J Physiol Endocrinol Metab ; 305(2): E194-204, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23695211

ABSTRACT

This study was designed to systematically analyze and evaluate the effects of in vivo blockade of α1-adrenergic receptors (α1-ADRs) on the stress-induced disturbance of steroidogenic machinery in Leydig cells. Parameters followed 1) steroidogenic enzymes/proteins, transcription factors, and cAMP/testosterone production; 2) the main hallmarks of stress (epinephrine, glucocorticoids); and 3) transcription profiles of ADRs and oxidases with high affinity to inactivate glucocorticoids. Results showed that sustained blockade of α1-ADRs prevented stress-induced 1) decrease of the transcripts/proteins for main steroidogenic CYPs (CYP11A1, CYP17A1); 2) decrease of Scarb1 and Hsd3b1 transcripts; 3) decrease of transcript for Nur77, one of the main activator of the steroidogenic expression; and 4) increase of Dax1 and Arr19, the main steroidogenic repressors in Leydig cells. In the same cells, the expression of steroidogenic stimulatory factor Creb1, StAR, and androgen receptor increased. In this signaling scenario, stress-induced stimulation of Adra1a/Adra1b/Adrbk1 and Hsd11b2 (the unidirectional oxidase with high affinity to inactivate glucocorticoids) was not changed. Blockade additionally stimulated stress-increased transcription of the most abundantly expressed ADRs Adra1d/Adrb1/Adrb2 in Leydig cells. In the same cells, stress-decreased testosterone production, the main marker of Leydig cells functionality, was completely prevented, while reduction of cAMP, the main regulator of androgenesis, was partially prevented. Accordingly, the presented data provide a new molecular/transcriptional base for "fight/adaptation" of steroidogenic cells and new molecular insights into the role of α1-ADRs in stress-impaired Leydig cell steroidogenesis. The results are important in term of wide use of α1-ADR selective antagonists, alone/in combination, to treat high blood pressure, nightmares associated with posttraumatic stress disorder, and disrupted sexual health.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Leydig Cells/drug effects , Leydig Cells/metabolism , Steroids/biosynthesis , Stress, Psychological/metabolism , Androgens/biosynthesis , Animals , Cells, Cultured , Cyclic AMP/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Doxazosin/pharmacology , Homeostasis/drug effects , Hormones/metabolism , Luteinizing Hormone/blood , Male , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, alpha-1/metabolism , Restraint, Physical , Testis/cytology , Testis/drug effects , Testis/metabolism , Transcription Factors/genetics
11.
Life (Basel) ; 13(7)2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37511832

ABSTRACT

Mitochondrial dynamics plays a significant role in shaping the mitochondrial network and maintaining mitochondrial function. Imbalanced mitochondrial dynamics can cause mitochondrial dysfunction leading to a wide range of diseases/disorders. The aim of this study was to investigate the expression of mitochondrial dynamics markers and regulatory molecules in whole adrenal glands, cortices, and medullae obtained from adult male rats exposed to acute and repeated psychophysical stress, the most common stress in human society. The transcriptional profiles of most of the mitochondrial dynamics markers investigated here were altered: 81%-(17/21) in the whole adrenal gland, 76.2%-(16/21) in the adrenal cortex, and 85.7%-(18/21) in the adrenal medulla. Changes were evident in representatives of every process of mitochondrial dynamics. Markers of mitobiogenesis were changed up to 62.5%-(5/8) in the whole adrenal gland, 62.5%-(5/8) in the adrenal cortex, and 87.5%-(7/8) in the adrenal medulla. Markers of mitofusion were changed up to 100%-(3/3) in the whole adrenal gland, 66.7%-(5/8) in the adrenal cortex, and 87.5%-(7/8) in the adrenal medulla, while all markers of mitofission and mitophagy were changed in the adrenal glands. Moreover, almost all markers of mitochondrial functionality were changed: 83.3%-(5/6) in the whole adrenal, 83.3%-(5/6) in the cortex, 66.7%-(4/6) in the medulla. Accordingly, the study highlights the significant impact of acute and repeated stress on mitochondrial dynamics in the adrenal gland.

12.
Eur J Cell Biol ; 102(2): 151323, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37201364

ABSTRACT

Decreased male fertility is a growing health problem that requires a better understanding of molecular events regulating reproductive competence. Here the effects of circadian desynchrony on the rat spermatozoa functionality were studied. Circadian desynchrony was induced in rats that lived for 2 months under disturbed light conditions designed to mimic shiftwork in humans (two days of constant light, two days of continual dark, and three days of 14:10 h light:dark schedule). Such a condition abolished circadian oscillations in the rats' voluntary activity, followed by a flattened transcriptional pattern of the pituitary gene encoding follicle stimulating hormone subunit (Fshb), and genes important for germ cell maturation (Tnp1 and Prm2) as well as the clock in seminiferous tubules. However, the number of spermatozoa isolated from the epididymis of the rats suffering from circadian desynchrony did not deviate from the controls. Nevertheless, spermatozoa functionality, estimated by motility and progesterone-induced acrosome reaction, was reduced compared to the control. These changes were associated with the altered level of main markers of mitochondrial biogenesis (Pprgc1a/PGC1A, Nrf1/NRF1, Tfam, Cytc), decreased mitochondrial DNA copy number, ATP content, and clock genes (Bmal1/BMAL1, Clock, Cry1/2, and Reverba). The principal-component-analysis (PCA) points to a positive association of the clock and mitochondrial biogenesis-related genes in spermatozoa from rats suffering circadian desynchrony. Altogether, the results show the harmful effect of circadian desynchrony on spermatozoa functionality, targeting energetic homeostasis.


Subject(s)
ARNTL Transcription Factors , Spermatozoa , Humans , Rats , Male , Animals , ARNTL Transcription Factors/genetics
13.
Biofactors ; 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38147453

ABSTRACT

The involvement of the molecular clock in regulating cell physiological processes on a specific time scale is a recognized concept, yet its specific impact on optimizing androgen production in Leydig cells has been unclear. This study aimed to confirm the role of the REVERBA (NR1D1) gene in controlling the transcription of key genes related to Leydig cell steroid production. We investigated daily variations by collecting Leydig cells from rats at various times within a 24-h period. Chromatin immunoprecipitation study showed a time-dependent pattern for genes linked to steroid production (Nur77, Star, Cyp11a1, and Cyp17a1), which closely matched the 24-h REVERBA levels in Leydig cells, peaking between zeitgeber time (ZT) 7-11. To understand the physiological significance of REVERBA's interaction with promoters of steroidogenesis-related genes, Leydig cells from rats at two different times (ZT7 and ZT16; chosen based on REVERBA expression levels), were treated with either an agonist (GSK4112) or an antagonist (SR8278). The results revealed that the REVERBA agonist stimulated gene transcription, while the antagonist inhibited it, but only when REVERBA was sufficiently present, indicating a reliance on REVERBA's circadian fluctuation. Moreover, this REVERBA-dependent stimulation had a clear impact on testosterone production in the culture medium, underscoring REVERBA's involvement in the circadian regulation of testosterone. This study indicates that REVERBA, in addition to being a core component of the cellular clock, plays a key role in regulating androgen production in Leydig cells by influencing the transcription of critical steroidogenesis-related genes.

14.
Am J Physiol Endocrinol Metab ; 302(10): E1239-51, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22374756

ABSTRACT

This study was designed to evaluate the effect of acute (2 h daily) and repeated (2 h daily for 2 or 10 consecutive days) immobilization stress (IMO) on: 1) the steroidogenic machinery homeostasis; 2) cAMP signaling; and the expression of receptors for main markers of 3) adrenergic and 4) glucocorticoid signaling in Leydig cells of adult rats. The results showed that acute IMO inhibited steroidogenic machinery in Leydig cells by downregulation of Scarb1 (scavenger receptor class B), Cyp11a1 (cholesterol side-chain cleavage enzyme), Cyp17a1 (17α-hydroxylase/17,20 lyase), and Hsd17b3 (17ß-hydroxysteroid dehydrogenase) expression. In addition to acute IMO effects, repeated IMO increased transcription of Star (steroidogenic acute regulatory protein) and Arr19 (androgen receptor corepressor 19 kDa) in Leydig cells. In the same cells, the transcription of adenylyl cyclases (Adcy7, Adcy9, Adcy10) and cAMP-specific phosphodiesterases (Pde4a, Pde4b, Pde4d, Pde7a, Pde8a) was stimulated, whereas the expression of the genes encoding protein kinase A subunits were unaffected. Ten times repeated IMO increased the levels of all adrenergic receptors and ß-adrenergic receptor kinase (Adrbk1) in Leydig cells. The transcription analysis was supported by cAMP/testosterone production. In this signaling scenario, partial recovery of testosterone production in medium/content was detected. The physiological significance of the present results was proven by ex vivo application of epinephrine, which increased cAMP/testosterone production by Leydig cells from control rats in greater fashion than from stressed. IMO did not affect the expression of transcripts for Crhr1/Crhr2 (corticotropin releasing hormone receptors), Acthr (adrenocorticotropin releasing hormone receptor), Gr (glucocorticoid receptor), and Hsd11b1 [hydroxysteroid (11-ß) dehydrogenase 1], while all types of IMO stimulated the expression of Hsd11b2, the unidirectional oxidase with high affinity to inactivate glucocorticoids. Thus, presented data provide new molecular/transcriptional base for "fight/adaptation" of Leydig cells and new insights into the role of cAMP, epinephrine, and glucocorticoid signaling in recovery of stress-impaired Leydig cell steroidogenesis.


Subject(s)
Cyclic AMP/metabolism , Leydig Cells/physiology , Receptors, Adrenergic/metabolism , Signal Transduction/physiology , Steroids/blood , Stress, Physiological/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Androgens/blood , Animals , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Corticosterone/blood , Luteinizing Hormone/blood , Male , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Restraint, Physical , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Transcription, Genetic/physiology
15.
J Sex Med ; 9(10): 2534-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22429315

ABSTRACT

INTRODUCTION: Phosphodiesterase type 5 (PDE5) inhibitors have been established in therapy for a variety of physiological disorders including erectile dysfunction. Despite its popularity and wide usage in erectile dysfunction treatment, the short-term effect of PDE5 inhibition on Leydig cell functionality and testosterone dynamics is missing. AIM: This study was designed to assess the acute in vivo effects of sildenafil citrate (Viagra) treatment on testosterone production. METHODS: Male adult rats were given sildenafil (1.25 mg/kg BW) per os, and testosterone production were analyzed 30, 60, 120, and 180 minutes after treatment. Additionally, in vitro effect of sildenafil extract on Leydig cell steroidogenesis was estimated. MAIN OUTCOME MEASURES: The formation of testicular interstitial fluid (TIF), and testosterone, cyclic guanosine monophosphate (cGMP), cyclic adenosine monophosphate (cAMP) content was followed. Occurrence and phosphorylation of mature steroidogenic acute regulatory protein (StAR) and interaction with protein kinase G 1 (PRKG1) were assessed by immunoprecipitation and Western blot. RESULTS: Serum testosterone was increased 60 and 120 minutes after sildenafil treatment. In 60 minutes, TIF volume was doubled and stayed increased till the end of the experimental period. cGMP and testosterone content in TIF were increased 30 minutes after treatment, and cAMP decreased in 60 minutes. Further, sildenafil-induced stimulation of testosterone production was abolished by ex vivo addition of PRKG1 inhibitor but not by protein kinase A inhibitor. Sildenafil treatment increased the level of phosphorylated and total StAR protein. Moreover, co-immunoprecipitation of StAR and PRKG1 was increased following sildenafil treatment suggesting the active role of this kinase in initiation of testosterone synthesis. Additionally, sildenafil extract applied in vitro on primary Leydig cell culture increased cGMP accumulation and testosterone production in time- and dose-dependent manner without effect on cAMP level. CONCLUSION: Acute sildenafil treatment enlarged TIF volume but also stimulated testosterone production which may be significant considering the positive testosterone effect in regulation of sexual activity.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Leydig Cells/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Phosphoproteins/metabolism , Piperazines/pharmacology , Sulfones/pharmacology , Testis/metabolism , Testosterone/blood , Animals , Blotting, Western , Cell Culture Techniques , Cyclic GMP/pharmacology , Immunoprecipitation , Leydig Cells/metabolism , Male , Purines/pharmacology , Rats , Rats, Wistar , Sildenafil Citrate
16.
Sci Rep ; 12(1): 15520, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109553

ABSTRACT

The increased frequency of different lifestyles that disrupts circadian rhythms, together with a trend in the accretion of male idiopathic infertility, imposes the necessity to understand the contribution of circadian rhythms disruption to fertility regulation. In this study, the effects of circadian desynchrony (CD) on the steroidogenic capacity of adult Leydig cells were studied. Adult rats were housed under a disturbing light regime (2 days of constant light, 2 days of continual dark, and 3 days of 12:12 h light:dark schedule) designed to mimic shiftwork in humans. CD was characterized by changed and decreased rhythmic locomotor activity and reduced blood testosterone. In the Leydig cells changed transcription of the clock genes (Bmal1, Clock, Cry1 and Reverba/b increased while Per1/2 reversed phase) was detected. This was followed by reduced transcription of genes (Star, Cyp11a1, and Hsd3b1/2) primarily involved in mitosteroidogenesis. In parallel, mitochondrial membrane potential (Δψi) and ATP production declined losing their characteristic oscillatory pattern. Also, the main markers of mitochondrial biogenesis (Ppargc1a, Nrf1, Tfam, Cytc), fusion (Mfn2), and mitophagy (Pink1 and Tfeb) were disturbed. Collectively, CD targets mitochondria in Leydig cells by reducing mitosteroidogenesis, mitoenergetics, and disturbing mitochondrial dynamics. These changes contribute to testosterone decline compromising androgen-dependent functions, including reproduction.


Subject(s)
ARNTL Transcription Factors , Leydig Cells , ARNTL Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Androgens/metabolism , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Gene Expression Regulation , Humans , Leydig Cells/metabolism , Male , Multienzyme Complexes/metabolism , Protein Kinases/metabolism , Rats , Testosterone/metabolism
17.
Cells ; 11(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35326444

ABSTRACT

In the search for the possible role of the mitochondrial dynamics markers in spermatozoa adaptation, an in vivo approach was designed to mimic situations in which human populations are exposed to 3 h of repeated psychological stress (the most common stress in human society) at different time points during the day (24 h). The hormones (stress hormone corticosterone and testosterone), the number and the functionality of spermatozoa (response to acrosome-reaction-inducer progesterone), as well as the transcriptional profiles of 22 mitochondrial dynamics and function markers and 22 signaling molecules regulating both mitochondrial dynamics and spermatozoa number and functionality were followed at three time points (ZT3, ZT11, and ZT23). The results show that repeated stress significantly decreased the number and functionality of spermatozoa at all time points. In the same samples, the transcriptional profiles of 91% (20/22) of mitochondrial dynamics and functionality markers and 86% (19/22) of signaling molecules were disturbed after repeated stress. It is important to point out that similar molecular changes in transcriptional profiles were observed at ZT3 and ZT23, but the opposite was observed at ZT11, suggesting the circadian nature of the adaptive response. The results of PCA analysis show the significant separation of repeated stress effects during the inactive/light and active/dark phases of the day, suggesting the circadian timing of molecular adaptations.


Subject(s)
Mitochondrial Dynamics , Signal Transduction , Biomarkers , Corticosterone , Humans , Male , Mitochondrial Dynamics/physiology , Sperm Count , Spermatozoa/physiology
18.
Front Endocrinol (Lausanne) ; 13: 896193, 2022.
Article in English | MEDLINE | ID: mdl-35909555

ABSTRACT

This study was designed to search for the possible mechanism(s) of male (in/sub)fertility by following the molecular response of spermatozoa on acute psychological stress (the most common stress in human society) and on a 20-h time-dependent recovery period. To mimic in vivo acute stress, the rats were exposed to immobilization once every 3 h. The recovery periods were as follows: 0 (immediately after stress and 3 h after the light is on-ZT3), 8 (ZT11), 14 (ZT17), and 20 (ZT23) h after stress. Results showed that acute stress provoked effects evident 20 h after the end of the stress period. Numbers of spermatozoa declined at ZT17 and ZT23, while functionality decreased at ZT3 and ZT11, but recovered at ZT17 and ZT23. Transcriptional profiles of 91% (20/22) of tracked mitochondrial dynamics and functionality markers and 91% (20/22) of signaling molecules regulating both mitochondrial dynamics and spermatozoa number/functionality were disturbed after acute stress and during the recovery period. Most of the changes presented as increased transcription or protein expression at ZT23. The results of the principal component analysis (PCA) showed the clear separation of acute stress recovery effects during active/dark and inactive/light phases. The physiological relevance of these results is the recovered positive-acrosome-reaction, suggesting that molecular events are an adaptive mechanism, regulated by acute stress response signaling. The results of the PCA confirmed the separation of the effects of acute stress recovery on gene expression related to mitochondrial dynamics, cAMP, and MAPK signaling. The transcriptional patterns were different during the active and inactive phases. Most of the transcripts were highly expressed during the active phase, which is expected given that stress occurred at the beginning of the inactive phase. To the best of our knowledge, our results provide a completely new view and the first presentation of the markers of mitochondrial dynamics network in spermatozoa and their correlation with signaling molecules regulating both mitochondrial dynamics and spermatozoa number and functionality during recovery from acute stress. Moreover, the interactions between the proteins important for spermatozoa homeostasis and functionality (MFN2 and PRKA catalytic subunit, MFN2 and p38MAPK) are shown for the first time. Since the existing literature suggests the importance of semen quality and male fertility not only as the fundamental marker of reproductive health but also as the fundamental biomarkers of overall health and harbingers for the development of comorbidity and mortality, we anticipate our result to be a starting point for more investigations considering the mitochondrial dynamics markers or their transcriptional profiles as possible predictors of (in/sub)fertility.


Subject(s)
Semen Analysis , Sperm Motility , Animals , Fertility/physiology , Humans , Male , Rats , Signal Transduction , Spermatozoa
19.
Mol Cell Endocrinol ; 538: 111469, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34601003

ABSTRACT

The study aimed to analyze the time-dependent consequences of stress on gene expression responsible for diurnal endocrine Leydig cell function connecting them to the glucocorticoid-signaling. In the first 24h after the stress event, a daily variation of blood corticosterone increased, and testosterone decreased; the testosterone/corticosterone were lowest at the end of the stress session overlapping with inhibition of Leydig cells' steroidogenesis-related genes (Nr3c1/GR, Hsd3b1/2, Star, Cyp17a1) and changed circadian activity of the clock genes (the increased Bmal1/BMAL1 and Per1/2/PER1 and decreased Cry1 and Rev-erba). The glucocorticoid-treated rats showed a similar response. The principal-component-analysis (PCA) displayed an absence of significant differences between treatments especially on Per1 and Rev-erba, the findings confirmed by the in vivo blockade of the testicular glucocorticoid receptor (GR) during stress and ex vivo treatment of the Leydig cells with hydrocortisone and GR-blocker. In summary, stressful stimuli can entrain the clock in the Leydig cells through glucocorticoid-mediated communication.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/genetics , Corticosterone/blood , Leydig Cells/metabolism , Testosterone/blood , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Leydig Cells/cytology , Male , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Stress, Physiological
20.
J Gerontol A Biol Sci Med Sci ; 76(2): 177-186, 2021 01 18.
Article in English | MEDLINE | ID: mdl-32459846

ABSTRACT

Since mitochondria play an essential role in the testosterone biosynthesis, serve as power centers and are a source of oxidative stress, a possible mitochondrial dysfunction could be connected with decreased activity of Leydig cells and lowered testosterone production during aging. Here we chronologically analyzed age-related alterations of mitochondrial function in Leydig cells correlated by the progressive rise of cGMP signaling and with respect to testosterone synthesis. To target cGMP signaling in Leydig cells, acute or long-term in vivo or ex vivo treatments with sildenafil (phosphodiesterase 5 [PDE5] inhibitor) were performed. Aging-related accumulation of cGMP in the Leydig cells is associated with mitochondrial dysfunction illustrated by reduced ATP and steroid production, lowered O2 consumption, increased mitochondrial abundance and mtDNA copies number, decreased expression of genes that regulate mitochondrial biogenesis (Ppargc1a/PGC1a-Tfam-Nrf1/NRF1), mitophagy (Pink1), fusion (Mfn1, Opa1), and increased Nrf2/NRF2. Acute in vivo PDE5 inhibition overaccumulated cGMP and stimulated testosterone but reduced ATP production in Leydig cells from adult, middle-aged, and old rats. The increased ATP/O ratio observed in cells from old compared to adult rats was diminished after stimulation of cGMP signaling. Opposite, long-term PDE5 inhibition decreased cGMP signaling and improved mitochondrial function/dynamics in Leydig cells from old rats. Mitochondrial abundance in Leydig cells decreased while ATP levels increased. Chronic treatment elevated Tfam, Nrf1, Nrf2, Opa1, Mfn1, Drp1, and normalized Pink1 expression. Altogether, long-term PDE5 inhibition prevented age-related NO and cGMP elevation, improved mitochondrial dynamics/function, and testosterone production. The results pointed on cGMP signaling in Leydig cells as a target for pharmacological manipulation of aging-associated changes in mitochondrial function and testosterone production.


Subject(s)
Aging/metabolism , Cyclic GMP/metabolism , Leydig Cells/metabolism , Mitochondrial Dynamics/physiology , Adenosine Triphosphate/metabolism , Aging/genetics , Animals , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Gene Expression , Homeostasis , Leydig Cells/drug effects , Male , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Mitophagy/drug effects , Models, Biological , Nitric Oxide/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Sildenafil Citrate/pharmacology , Testosterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL