Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Anat Histol Embryol ; 53(4): e13062, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38837469

ABSTRACT

Although the order Rodentia does not present a high risk of extinction compared to mammals as a whole, several families demonstrate high levels of threat and/or data deficiency, therefore highlighting the need for targeted research and the application of ecological and reproductive data to the development of conservation actions. The order Rodentia, the largest among mammals, includes 9 families, and the family Cricetidae is the most diverse of the Brazilian rodents. In Brazil, 12 of the 16 genera of Oecomys are found. Oecomys bicolor is known in Brazil as the 'arboreal rat' and is, found in dry, deciduous and tropical forests. The mean body weight of Oecomys bicolor was 35.8 g and the gonadal, tubular and epithelial somatic indexes were, 0.53%, 0.47% and 0.37%, respectively. Seminiferous tubules volume density was 89.72% and the mitotic and meiotic indexes corresponded to 8.59 and 2.45 cells, respectively, and the yield of spermatogenesis was 23.83 cells. The intertubular compartment represented 10.28% of the testis parenchyma and around 5% of the interstitial space was occupied by Leydig cells, whose number per gram of testis was 11.10 × 107 cells. By evaluating the biometric and histomorphometric characteristics of the testis, there is evidence that this species has a high investment in reproduction. Due to the high contribution of the seminiferous epithelium and the intertubular compartment in this species, compared to the others of the same family, it is possible to infer that the species Oecomys bicolor has a promiscuous reproductive behaviour.


Subject(s)
Arvicolinae , Leydig Cells , Spermatogenesis , Testis , Animals , Spermatogenesis/physiology , Male , Testis/anatomy & histology , Testis/physiology , Leydig Cells/cytology , Leydig Cells/physiology , Arvicolinae/anatomy & histology , Arvicolinae/physiology , Seminiferous Tubules/anatomy & histology , Brazil
2.
Anim. Reprod. (Online) ; 19(3): e20210124, set. 2022. tab, graf, ilus
Article in English | VETINDEX | ID: biblio-1403209

ABSTRACT

Duchenne Muscular Dystrophy (DMD) reproductive alterations and the influence of antioxidant treatments may aid in understanding morphometry testicular quantification. In this context, the aim of the present study was to characterize the intertubular compartment (ITC) morphometry of animal testes in mdx mice supplemented with ascorbic acid (AA). Sixteen mice were used, namely the C57BL/10 (non-dystrophic) and C57BL/10Mdx (dystrophic) lineages, distributed into the following groups: Control (C60), Dystrophic (D60), Control supplemented with AA (CS60), Dystrophic supplemented with AA (DS60). A total of 200 mg/kg of AA were administered to mice for 30 days. Subsequently, the testicles were collected, weighed, and fragmented. The obtained fragments were fixed in Karnovsky's solution (pH 7.2) and embedded in historesin for morphometric and transmission electron microscopy assessments. Leydig cells were hypertrophic in the D60 group, but was reverted by AA supplementation in the DS60 group. The DS60 group also exhibited increased intertubular volume compared to the CS60 group. The ultrastructural images identified multilamellar bodies in dystrophic animals (lipid storage) and telocyte cells (transport substances) in both control and dystrophic animals. Morphometric alterations were, therefore, noted in the intertubular compartment due to Duchenne muscular dystrophy (DMD), with AA administration capable of altering Leydig cells in this condition.(AU)


Subject(s)
Animals , Male , Mice , Seminiferous Tubules/physiopathology , Mice, Inbred mdx/physiology , Leydig Cells/physiology , Muscular Diseases/veterinary
3.
Cell Tissue Res ; 380(3): 615-625, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31950264

ABSTRACT

Mutations in Foxn1 and Prkdc genes lead to nude and severe combined immunodeficiency (scid) phenotypes, respectively. Besides being immunodeficient, previous reports have shown that nude mice have lower gonadotropins and testosterone levels, while scid mice present increased pachytene spermatocyte (PS) apoptosis. Therefore, these specific features make them important experimental models for understanding Foxn1 and Prkdc roles in reproduction. Hence, we conducted an investigation of the testicular function in nude and scid BALB/c adult male mice and significant differences were observed, especially in Leydig cell (LC) parameters. Although the differences were more pronounced in nude mice, both immunodeficient strains presented a larger number of LC, whereas its cellular volume was smaller in comparison to the wild type. Besides these alterations in LC, we also observed differences in androgen receptor and steroidogenic enzyme expression in nude and scid mice, suggesting the importance of Foxn1 and Prkdc genes in androgen synthesis. Specifically in scid mice, we found a smaller meiotic index, which represents the number of round spermatids per PS, indicating a greater cell loss during meiosis, as previously described in the literature. In addition and for the first time, Foxn1 was identified in the testis, being expressed in LC, whereas DNA-PKc (the protein produced by Prkdc) was observed in LC and Sertoli cells. Taken together, our results show that the changes in LC composition added to the higher expression of steroidogenesis-related genes in nude mice and imply that Foxn1 transcription factor may be associated to androgen production regulation, while Prkdc expression is also important for the meiotic process.


Subject(s)
DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , Forkhead Transcription Factors/physiology , Leydig Cells/physiology , Sertoli Cells/physiology , Animals , Leydig Cells/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Receptors, Androgen/metabolism , Sertoli Cells/cytology
4.
Braz J Med Biol Res ; 51(5): e6693, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29561954

ABSTRACT

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 µM and 1 mM), L-arginine (10, 100, 300, and 500 µM), ODQ (300 µM), and 8-Br-cGMP (100 µM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 µM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Subject(s)
Adenosine Triphosphate/physiology , Leydig Cells/physiology , Nitric Oxide/physiology , Receptors, Purinergic/metabolism , Action Potentials , Animals , Arginine/administration & dosage , Arginine/metabolism , Cells, Cultured , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/biosynthesis , Patch-Clamp Techniques , Thionucleotides/administration & dosage , Thionucleotides/metabolism
5.
Reprod Fertil Dev ; 30(7): 1029-1037, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29325604

ABSTRACT

The insectivorous bat Myotis nigricans is widely distributed throughout the Neotropics, including Brazil, and has a reproductive biology that is affected by climate and food availability. To evaluate the reproductive capacity of this species, morphofunctional parameters of the testes were correlated with environmental variables and the body condition of individuals captured. After bats had been killed, their testes were removed, fixed in Karnovsky's fluid for 24h and embedded in resin for evaluation by light microscopy. The mean annual tubulosomatic index (0.58%) and the percentage of seminiferous tubules in the testes (88.96%) were the highest ever recorded for the Order Chiroptera. The percentage of Leydig cells and volume of the cytoplasm of Leydig cells were higher in the rainy than dry season (80.62±3.19% and 573.57±166.95µm, respectively; mean±s.d.). Conversely, the percentage of nuclei of the Leydig cells in the dry season (26.17±3.70%; mean±s.d.) and the total number of Leydig cells (6.38±1.84×109; mean±s.d.) were higher in the dry season. The results of the present study could help in future conservation of these bats because they provide a better understanding of the bats' reproductive strategies and how the species can adapt to changes.


Subject(s)
Reproduction/physiology , Seasons , Spermatogenesis/physiology , Testis/anatomy & histology , Animals , Chiroptera , Leydig Cells/physiology , Male , Seminiferous Tubules/anatomy & histology , Seminiferous Tubules/physiology , Testis/physiology
6.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;51(5): e6693, 2018. graf
Article in English | LILACS | ID: biblio-889091

ABSTRACT

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 μM and 1 mM), L-arginine (10, 100, 300, and 500 μM), ODQ (300 μM), and 8-Br-cGMP (100 μM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 μM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Subject(s)
Animals , Male , Mice , Adenosine Triphosphate/physiology , Receptors, Purinergic/metabolism , Leydig Cells/physiology , Nitric Oxide/physiology , Arginine/administration & dosage , Arginine/metabolism , Thionucleotides/administration & dosage , Thionucleotides/metabolism , Action Potentials , Cells, Cultured , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Patch-Clamp Techniques , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/biosynthesis
7.
Environ Sci Pollut Res Int ; 22(11): 8526-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25561257

ABSTRACT

Ametryn is an herbicide used to control broadleaf and grass weeds and its acute and chronic toxicity is expected to be low. Since toxicological data on ametryn is scarce, the aim of this study was to evaluate rat reproductive toxicity. Thirty-six adult male Wistar rats (90 days) were divided into three groups: Co (control) and T1 and T2 exposed to 15 and 30 mg/kg/day of ametryn, respectively, for 56 days. Testicular analysis demonstrated that ametryn decreased sperm number per testis, daily sperm production, and Leydig cell number in both treated groups, although little perceptible morphological change has been observed in seminiferous tubule structure. Lipid peroxidation was higher in group T2, catalase activity decreased in T1 group, superoxide dismutase activity diminished, and a smaller number of sulphydryl groups of total proteins were verified in both exposed groups, suggesting oxidative stress. These results showed negative ametryn influence on the testes and can compromise animal reproductive performance and survival.


Subject(s)
Herbicides/toxicity , Leydig Cells/physiology , Oxidative Stress/drug effects , Reproduction/drug effects , Spermatozoa/physiology , Triazines/toxicity , Analysis of Variance , Animals , Biometry , Body Weights and Measures , Brazil , Catalase/metabolism , Cell Count , Leydig Cells/drug effects , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar , Spermatogenesis/drug effects , Spermatozoa/drug effects , Superoxide Dismutase/metabolism
8.
Anat Histol Embryol ; 44(5): 361-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25258091

ABSTRACT

Yellowish myotis, Myotis levis, is a seasonal, epididymal sperm-storing Neotropical vespertilionid. In the dry season, males show simultaneous testis regression and sperm storage in cauda epididymis, enabling them to mate during this season. In this study, we investigated seasonal variations in body mass, diameter and height of seminiferous tubules and nuclei of Leydig cells in a population of southeastern Brazil. We also determined the frequencies of the stages of the seminiferous epithelium cycle (SEC) of mature individuals of this population. Body mass and diameter of Leydig cell nuclei showed no significant differences between dry and rainy seasons and stages of annual reproductive cycle; however, all other morphometric parameters varied significantly. The relative cumulative frequency of pre-meiotic stages of the SEC (1-3) was 51%, of meiotic stage (4) was 2% and of post-meiotic stages (5-8) was 47%. We confirmed that the yellowish myotis presents seasonal sperm production as revealed by testis regression and epididymal sperm storage during the dry season.


Subject(s)
Chiroptera/anatomy & histology , Epididymis/anatomy & histology , Seminiferous Epithelium/anatomy & histology , Sexual Behavior, Animal/physiology , Animals , Body Mass Index , Brazil , Cell Nucleus/physiology , Epididymis/cytology , Leydig Cells/physiology , Male , Seasons , Spermatogenesis , Spermatozoa/cytology
9.
Acta Histochem ; 116(6): 1037-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24894511

ABSTRACT

Spermatogenesis is disrupted in Graomys griseoflavus×Graomys centralis male hybrids. This study was aimed to determine whether morphological alterations in Leydig cells from hybrids accompany the arrest of spermatogenesis and cell death of germ cells and whether apoptotic pathways are also involved in the response of these interstitial cells. We used three groups of 1-, 2- and 3-month-old male animals: (1) G. centralis, (2) G. griseoflavus and (3) hybrids obtained by crossing G. griseoflavus females with G. centralis males. Testicular ultrastructure was analyzed by transmission electron microscopy. TUNEL was studied using an in situ cell death detection kit and the expression of apoptotic molecules by immunohistochemistry. The data confirmed arrest of spermatogenesis and intense apoptotic processes of germ cells in hybrids. These animals also showed ultrastructural alterations in the Leydig cells. Fas, FasL and calbindin D28k overexpression without an increase in DNA fragmentation was detected in the Leydig cells from hybrids. In conclusion, the sterility of Graomys hybrids occurs with ultrastructural changes in germ and Leydig cells. The enhancement of Fas and FasL is not associated with cell death in the Leydig cells. Probably the apoptosis in these interstitial cells is inhibited by the high expression of the antiapoptotic molecule calbindin D28k.


Subject(s)
Apoptosis , Leydig Cells/physiology , Rodentia/anatomy & histology , Animals , Calbindins/metabolism , Fas Ligand Protein/metabolism , Female , Hybridization, Genetic , Leydig Cells/ultrastructure , Male , Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Spermatogenesis , fas Receptor/metabolism
10.
Andrology ; 2(2): 175-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24574094

ABSTRACT

Nicotine is largely consumed as a component of cigarettes. It induces apoptosis, interferes with endocrine function by changing the sex hormones secretion and leads to male infertility. Testosterone is produced from cholesterol by Leydig cells (LC), with the participation of testicular macrophages (MO). Thus, to investigate whether nicotine administration to pregnant and lactating rats changes cholesterol and sexual hormone levels and LC and MO populations of offspring, female rats received nicotine (2 mg/kg/day) through osmotic minipumps from the first day of pregnancy up to the end of weaning. At 1, 30, 60 and 90 days post-partum (dpp) the plasma cholesterol and testosterone levels were obtained, as well as the biometric, histopathological and stereological testicular parameters. Nicotine reduced the body weight, cholesterol levels and lipid droplet number in foetal LC at 1 dpp. The number of apoptotic LC did not change in the offspring of nicotine group at any age studied. No alterations in the numerical densities of MO and LC occurred at 60 and 90 dpp. Hypertrophy of mature LC and increase in cholesterol and testosterone levels were noted at 90 dpp. In conclusion, nicotine when administered to rats throughout pregnancy and lactation induces morphofunctional alterations of foetal and mature LC and affects cholesterol and testosterone levels.


Subject(s)
Leydig Cells/physiology , Macrophages/physiology , Nicotine/pharmacology , Prenatal Exposure Delayed Effects , Animals , Apoptosis/drug effects , Body Weight/drug effects , Cholesterol/blood , Female , Ganglionic Stimulants/pharmacology , Lactation , Leydig Cells/drug effects , Macrophages/drug effects , Male , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacology , Pregnancy , Rats , Testosterone/blood
11.
Acta Sci. Biol. Sci. ; 36(4): 483-489, out.-dez. 2014. tab, ilus
Article in English | VETINDEX | ID: vti-694962

ABSTRACT

The goal of present study was to evaluate the effects of diabetes on quantitative parameters of Leydig cells. Twelve adults Wistar rats were divided in: 1) Diabetic Group (DG), which was induced by a single intraperitoneal injection of streptozotocin (60 mg kg-1 of body weight); and 2) Control Group (CG), which received citrate buffer intraperitoneal. After eight weeks of diabetic induction, the animals were weighted, anesthetized and testicles were removed and routinely processed to paraffin embedded. Body weight (40%) and testicular weight (18%) of diabetic rats were significantly lower than control group.  Diabetic rats showed an increase in interstitial compartment but the tubular compartment did not differ. The individual volume of Leydig cells and nuclear diameter were lower in DG. However, the population of these cells was increased. In conclusion, diabetes induced by streptozotocin in adult rats promoted alterations in testicular compartments and changes on volume, nuclear diameter and population of Leydig cells, compromising the testicular function.(AU)


O objetivo do presente estudo foi avaliar os efeitos do diabetes nos parâmetros quantitativos de células de Leydig. Doze ratos machos adultos foram divididos em: 1) Grupo Diabético (GD) induzidos por injeção intraperitoneal única de estreptozotocina (60 mg kg-1 de peso corporal); e 2) Grupo Controle (GC) receberam tampão citrato, via intraperitoneal. Após oito semanas da indução, os animais foram pesados, anestesiados, os testículos foram removidos e processados rotineiramente em parafina. O peso corporal (40%) e testicular (18%) dos ratos diabéticos reduziu significativamente em relação ao grupo controle. Ratos diabéticos mostraram aumento no compartimento intersticial, mas o compartimento tubular não apresentou diferença significativa. O volume individual e o diâmetro nuclear de células de Leydig reduziram em GD. No entanto, a população dessas células aumentou. Em conclusão, o diabetes induzido por estreptozotocina, em ratos adultos, promoveu alterações nos compartimentos testiculares e mudanças no volume, diâmetro nuclear e população das células de Leydig, comprometendo a função testicular.(AU)


Subject(s)
Animals , Male , Adult , Rats , Leydig Cells/physiology , Rats/blood , Diabetes Mellitus, Experimental/diagnosis
12.
Andrologia ; 45(6): 379-85, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23013062

ABSTRACT

We characterised and correlated the histological and hormonal aspects of a cohort of 261 azo/oligozoospermic men, applying a quantitative/qualitative evaluation of testicular tissue and serum and intratesticular hormonal measurements. One hundred and 93 azo/oligozoospermic patients were diagnosed as: complete sertoli cell only syndrome (cSCOS), n = 76; focal SCOS, n = 31; maturation arrest, n = 34; hypospermatogenesis, n = 17; mixed atrophy, n = 25; and severe atrophy, n = 10. Normal spermatogenesis was observed in 68 infertile men (controls). Patients with cSCOS, focal SCOS, mixed and severe atrophy had larger LC/clusters (11.5; 11.0; 10.7; 18.9 LC/cluster) than controls (6 LC/cluster; P < 0.001). cSCOS, focal SCOS, mixed and severe atrophy patients had higher FSH, LH and lower T/LH ratio serum levels than the other groups. Intratesticular testosterone concentrations were higher in tissues with complete or focal SCOS (45.6 ng mg(-1) protein) and mixed atrophy (79.0 ng mg(-1) protein) than normal tissues (20.3 ng mg(-1) protein; P = 0.03 and P = 0.007). Considering all subjects, significant correlations were found between T/LH ratio and Leydig cells/cluster (r = 0.510, P < 0.001), FSH levels (r = -0.692, P < 0.001) and with intratesticular testosterone (r = -0.354, P = 0.001); these correlations follow the pattern of severity of spermatogenic damage. By a thorough histological evaluation, we validate the concept that the severity of spermatogenic impairment is associated with major morphological and functional disturbance of the Leydig cell compartment.


Subject(s)
Infertility, Male/pathology , Spermatogenesis/drug effects , Testis/pathology , Testis/physiopathology , Atrophy , Azoospermia/blood , Follicle Stimulating Hormone/blood , Humans , Leydig Cells/metabolism , Leydig Cells/physiology , Luteinizing Hormone/blood , Male , Oligospermia/pathology , Sertoli Cell-Only Syndrome , Testosterone/blood
13.
Int J Androl ; 34(5 Pt 2): e487-98, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21831236

ABSTRACT

Male patients with an extra sex chromosome or autosome are expected to present primary hypogonadism at puberty owing to meiotic germ-cell failure. Scarce information is available on trisomy 21, a frequent autosomal aneuploidy. Our objective was to assess whether trisomy 21 presents with pubertal-onset, germ-cell specific, primary hypogonadism in males, or whether the hypogonadism is established earlier and affects other testicular cell populations. We assessed the functional status of the pituitary-testicular axis, especially Sertoli cell function, in 117 boys with trisomy 21 (ages: 2months-20year). To compare with an adequate control population, we established reference levels for serum anti-Müllerian hormone (AMH) in 421 normal males, from birth to adulthood, using a recently developed ultrasensitive assay. In trisomy 21, AMH was lower than normal, indicating Sertoli cell dysfunction, from early infancy, independently of the existence of cryptorchidism. The overall prevalence rate of AMH below the 3rd percentile was 64.3% in infants with trisomy 21. Follicle-stimulating hormone was elevated in patients <6months and after pubertal onset. Testosterone was within the normal range, but luteinizing hormone was elevated in most patients <6months and after pubertal onset, indicating a mild Leydig cell dysfunction. We conclude that in trisomy 21, primary hypogonadism involves a combined dysfunction of Sertoli and Leydig cells, which can be observed independently of cryptorchidism soon after birth, thus prompting the search for new hypotheses to explain the pathophysiology of gonadal dysfunction in autosomal trisomy.


Subject(s)
Anti-Mullerian Hormone/blood , Down Syndrome/physiopathology , Hypogonadism/physiopathology , Adolescent , Adult , Child , Child, Preschool , Down Syndrome/complications , Follicle Stimulating Hormone/blood , Humans , Hypogonadism/blood , Hypogonadism/etiology , Infant , Infant, Newborn , Leydig Cells/physiology , Luteinizing Hormone/blood , Male , Organ Size , Sertoli Cells/physiology , Testis/anatomy & histology , Testosterone/blood
14.
Anim Reprod Sci ; 127(1-2): 73-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21802871

ABSTRACT

The present study aimed to compare testicular histology and the testicular cell population as well as spermatogenic efficiency in goats with different scrotal conformations. Eighteen goats were divided into 3 groups: Group I - goats without bipartition of the scrotum, Group II - animals with bipartition of the scrotum up to 50% of the testicular length, Group III - goats with scrotal bipartition more than 50% of the testicular length. In goats in Groups I, II and III, the values for the volume density of seminiferous epithelium were 68.9 ± 0.6%, 71.5 ± 2.8% and 73.4 ± 4.7% (P<0.05), the height of the seminiferous epithelium were 60.2 ± 4.9 µm, 61.0 ± 5.0 µm and 73.1 ± 6.6 µm (P<0.05), total length of seminiferous tubules found for Groups I, II and III were 2091.9 ± 27 m, 2172.5 ± 24.1 m, and 2340.1 ± 14 m (P<0.05), number of Sertoli and Leydig cells were 1.8 ± 0.4×10(9) and 1.4 ± 0.1×10(9), 2.2 ± 0.4 and 2.2 ± 0.7×10(9), and 2.5 ± 0.1 10(9) and 2.3 ± 0.5 10(9) (P<0.05) and daily sperm production observed were 2.1 ± 0.3×10(9), 2.8 ± 0.4×10(9), and 3.1 ± 0.7×10(9) (P<0.05). In conclusion, goats with greater scrotal bipartition have a greater capacity to produce reproductive cells that is reflected in a greater reproductive potential.


Subject(s)
Goats/physiology , Scrotum/anatomy & histology , Spermatogenesis/physiology , Testis/physiology , Animals , Brazil , Cell Count/veterinary , Goats/anatomy & histology , Histocytochemistry/veterinary , Leydig Cells/physiology , Male , Sertoli Cells/physiology , Statistics, Nonparametric , Testis/anatomy & histology , Testis/cytology , Tropical Climate
15.
Acta Paediatr ; 100(6): 830-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21214886

ABSTRACT

UNLABELLED: Male hypogonadism implies decreased function of one or more testicular cell population, i.e. germ, Leydig and/or Sertoli cells. In the normal prepubertal boy, Sertoli cells are very active, as indicated by high anti-Müllerian hormone (AMH) and inhibin B secretion, whereas the functional activity of Leydig cells is minimal, as evidenced by low testosterone production, and germ cells do not undergo the full spermatogenic process. Klinefelter syndrome is the most frequent cause of hypogonadism in the adult male. In this review, we discuss whether the gonadal failure is already established during infancy and childhood. In Klinefelter syndrome, there is increased germ cells degeneration from mid-foetal life - resulting in a decreased number at birth - which persists during infancy and childhood and becomes dramatic during puberty. Controversial results exist in the literature regarding Leydig cell function in Klinefelter boys: while some authors have found normal to low testosterone levels in infancy and childhood, others have reported normal to high values. Sertoli cell products AMH and inhibin B are normal in prepubertal boys and only decline during mid- to late puberty. CONCLUSION: Klinefelter syndrome is a primary hypogonadism affecting all testicular cell populations. Germ cells are affected from foetal life, and a severe depletion occurs at puberty. Leydig cell function may be normal or mildly affected in foetal and early postnatal life. Sertoli cell function is not impaired until mid- to late puberty, as reflected by normal AMH and inhibin B in Klinefelter boys.


Subject(s)
Hypogonadism/diagnosis , Klinefelter Syndrome/physiopathology , Child , Germ Cells/physiology , Humans , Infant , Klinefelter Syndrome/embryology , Leydig Cells/physiology , Male , Puberty/physiology , Sertoli Cells/physiology , Testis/embryology , Testis/metabolism
16.
Best Pract Res Clin Endocrinol Metab ; 24(2): 243-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20541150

ABSTRACT

Disorders of androgen production can occur in all steps of testosterone biosynthesis and secretion carried out by the foetal Leydig cells as well as in the conversion of testosterone into dihydrotestosterone (DHT). The differentiation of Leydig cells from mesenchymal cells is the first walk for testosterone production. In 46,XY disorders of sex development (DSDs) due to Leydig cell hypoplasia, there is a failure in intrauterine and postnatal virilisation due to the paucity of interstitial Leydig cells to secrete testosterone. Enzymatic defects which impair the normal synthesis of testosterone from cholesterol and the conversion of testosterone to its active metabolite DHT are other causes of DSD due to impaired androgen production. Mutations in the genes that codify the enzymes acting in the steps from cholesterol to DHT have been identified in affected patients. Patients with 46,XY DSD secondary to defects in androgen production show a variable phenotype, strongly depending of the specific mutated gene. Often, these conditions are detected at birth due to the ambiguity of external genitalia but, in several patients, the extremely undervirilised genitalia postpone the diagnosis until late childhood or even adulthood. These patients should receive long-term care provided by multidisciplinary teams with experience in this clinical management.


Subject(s)
Androgens/biosynthesis , Disorders of Sex Development/genetics , Gonadal Dysgenesis, 46,XY/diagnosis , Gonadal Dysgenesis, 46,XY/therapy , Adrenal Hyperplasia, Congenital/physiopathology , Adult , Cell Differentiation , Child , Female , Glucocorticoids/therapeutic use , Gonadal Dysgenesis, 46,XY/pathology , Humans , Infant , Infant, Newborn , Leydig Cells/physiology , Male , Mineralocorticoids/therapeutic use , Receptors, LH/genetics , Smith-Lemli-Opitz Syndrome/genetics , Testosterone/biosynthesis
17.
J Reprod Dev ; 56(4): 389-95, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20431248

ABSTRACT

Transforming growth factor beta 1 (TGF-beta1) modulates male reproductive function. Genetically modified mice overexpressing alpha/beta subunits of hCG (hCG+) show Leydig cell hyperplasia/hypertrophy at prepuberty that disappears as the mice approach adulthood. In this study we analyzed the gene expression of TGF-beta1, its specific receptors, type II (TGF-betaRII) and type I (activin receptor-like kinase 1 and 5: ALK1 and ALK5), and co-receptor endoglin (CD105) in purified Leydig cells from hCG+ and wild-type mice at 3 and 8 weeks of age and the occurrence of TGF-beta1, ALK1 and ALK5 by immunohistochemistry. The expression of TGF-beta1 was higher in hCG+ mice at both ages studied, and no changes were observed in TGF-betaRII. ALK5 diminished with age in wild-type mice, whereas ALK1 decreased in hCG+ mice at 8 weeks of age. Endoglin expression showed a marked increase in 3-week-old hCG+ animals. In vitro incubation of Leydig cells from wild-type animals with hCG (10 IU/ml) increased TGF-beta1 and ALK5 expression. Progesterone (10(-6) M) induced endoglin expression. These studies provide novel evidence for differential gene and protein expression of ALK1 and ALK5 at different ages and endoglin expression and hormonal, in purified Leydig cells.


Subject(s)
Chorionic Gonadotropin/pharmacology , Leydig Cells/drug effects , Leydig Cells/metabolism , Progesterone/pharmacology , Transforming Growth Factor beta1/physiology , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II , Age Factors , Animals , Cells, Cultured , Chorionic Gonadotropin/genetics , Endoglin , Gene Expression Regulation, Developmental/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leydig Cells/physiology , Male , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Sexual Maturation/genetics , Sexual Maturation/physiology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
18.
J Reprod Dev ; 56(4): 400-4, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20431251

ABSTRACT

Several reports indicate that transforming growth factor beta1 (TGF-beta1) participates in the regulation of cell cycle progression. In this work, we analyzed the in vitro effect of TGF-beta1 on Leydig cell proliferation markers and the in vivo effect of this cytokine in Leydig cell hyperplasia/hypertrophy. The in vitro effect of TGF-beta1 (1 ng/ml) plus progesterone (10(-6) M) on purified Leydig cells from 3 week-old mice increased the immunocytochemically detected PCNA and stimulated the phosphorylation of Smad 1/5. Progesterone (10(-6) M) in the presence or absence of TGF-beta1 diminished the ratio Bax/Bcl-2. Morphometric testicular studies of mice treated with progesterone (s.c.) plus TGF-beta1 (intratesticular), showed an increase in interstitial volume and a decrease in tubular volume. Furthermore, the cytoplasmic volume of Leydig cells showed an increment in this experimental group with a diminution in nuclear volume. Thus, it turned out that the administration of progesterone and TGF-beta1 augmented the volume of Leydig cells. These results indicate a clear effect of TGF-beta1 in the hypertrophy/hyperplasia of Leydig cells.


Subject(s)
Leydig Cells/metabolism , Leydig Cells/pathology , Progesterone/pharmacology , Smad1 Protein/physiology , Smad5 Protein/physiology , Transforming Growth Factor beta1/metabolism , Animals , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , Hyperplasia/etiology , Hyperplasia/genetics , Hyperplasia/metabolism , Hypertrophy/etiology , Hypertrophy/genetics , Hypertrophy/metabolism , Leydig Cells/drug effects , Leydig Cells/physiology , Male , Mice , Organ Size/drug effects , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Testis/metabolism , Testis/pathology , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/physiology
19.
Anim Reprod Sci ; 120(1-4): 129-35, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20434281

ABSTRACT

The present study was aimed at evaluating the sequence of the histological and quantitative changes that occur in diverse components of the interstitial tissue, as well as the variations in the number and volume of Leydig cells of the testes of Gallus domesticus from 8-day-old embryo to 28-week-old chickens. Results indicate an increase in total volume of all components of the interstitial tissue: Leydig cells, blood vessels, and interstitium of chicken testes with advancing development. The proportion of each component differs markedly according to the age of the animal. Data indicate that Leydig cells are present in the chicken testes from pre-hatching to sexual maturity, and show an increase in the total volume, number per testis, and individual cell volume along the age of the animal. Values of P<0.05 were considered statistically significant. The results indicate that the total volume of Leydig cells is the dominant component of the interstitial tissue in the testes of 6-week-old chickens, but in testes of chicken embryos correspond to the lowest percentage of interstitial tissue. Results also reveal that hyperplasia is more important than cell hypertrophy in the increase of the total volume of Leydig cells during advancing development.


Subject(s)
Chickens , Leydig Cells/cytology , Sexual Maturation/physiology , Testis/embryology , Testis/growth & development , Aging/physiology , Animals , Cell Count , Cell Differentiation , Cell Enlargement , Cell Proliferation , Cell Size , Chick Embryo , Chickens/anatomy & histology , Chickens/growth & development , Chickens/physiology , Leydig Cells/physiology , Male , Organ Size , Testis/anatomy & histology , Testis/cytology
20.
Theriogenology ; 74(1): 11-23, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20189235

ABSTRACT

Although seminiferous tubule maturation in horses begins in the central area of the testis, this process is thought to occur randomly throughout the testis in most mammals. Studies in our laboratory revealed that the establishment of spermatogenesis may not be a synchronous event in the testicular parenchyma of pigs. The objectives of the present study were to evaluate the pattern of seminiferous cord/tubule maturation and the morphological and functional characteristics of testicular somatic cells during postnatal development in three regions of the pig testis: a) near the tunica albuginea (TA); b) in the transitional area between the seminiferous tubules and mediastinum (TR); and c) in the intermediate area (ID) between the TA and TR. Based on the diameter of seminiferous cords/tubules, nucleus size of Sertoli cells and fluid secretion, mainly at 90 and 120 d of age, seminiferous tubule maturation was more advanced in the ID and TR. The mitotic activity of Sertoli cells was higher (P<0.05) in the TR than the ID and TA at 7 and 120 d. Except for the mitotic index of the Leydig cells, which was lower (P<0.05) in the ID at 7, 30, and 180 d than in the TA and TR, other Leydig cell ebd points, e.g., individual cell size, nuclear volume, and cytoplasmic volume, were consistently higher (P<0.05) in the ID, suggesting that steroidogenesis was more active in this region during the period investigated. Overall, we inferred that Leydig cells in the ID may play a pivotal role in postnatal testis development in pigs and this type of cell is likely related to asynchronous testicular parenchyma development, with the transitional area providing the primary zone for growth of seminiferous tubules.


Subject(s)
Cell Division , Seminiferous Tubules/growth & development , Swine/growth & development , Testis/cytology , Testis/growth & development , Animals , Cell Nucleus/ultrastructure , Cytoplasm/ultrastructure , Leydig Cells/physiology , Leydig Cells/ultrastructure , Male , Mitosis , Sertoli Cells/physiology , Sertoli Cells/ultrastructure , Sexual Maturation , Spermatogenesis
SELECTION OF CITATIONS
SEARCH DETAIL