Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
1.
Life Sci Alliance ; 5(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35086936

RESUMO

Unlike constitutively secreted proteins, peptide hormones are stored in densely packed secretory granules, before regulated release upon stimulation. Secretory granules are formed at the TGN by self-aggregation of prohormones as functional amyloids. The nonapeptide hormone vasopressin, which forms a small disulfide loop, was shown to be responsible for granule formation of its precursor in the TGN as well as for toxic fibrillar aggregation of unfolded mutants in the ER. Several other hormone precursors also contain similar small disulfide loops suggesting their function as a general device to mediate aggregation for granule sorting. To test this hypothesis, we studied the capacity of small disulfide loops of different hormone precursors to mediate aggregation in the ER and the TGN. They indeed induced ER aggregation in Neuro-2a and COS-1 cells. Fused to a constitutively secreted reporter protein, they also promoted sorting into secretory granules, enhanced stimulated secretion, and increased Lubrol insolubility in AtT20 cells. These results support the hypothesis that small disulfide loops act as novel signals for sorting into secretory granules by self-aggregation.


Assuntos
Hormônios Peptídicos/metabolismo , Vesículas Secretórias/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Grânulos Citoplasmáticos/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Complexo de Golgi/metabolismo , Hormônios/genética , Hormônios/metabolismo , Humanos , Hormônios Peptídicos/genética , Transporte Proteico , Vesículas Secretórias/fisiologia , Vasopressinas/metabolismo , Rede trans-Golgi/metabolismo
2.
Front Immunol ; 12: 717808, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34394125

RESUMO

Its semi-allogeneic nature renders the conceptus vulnerable to attack by the maternal immune system. Several protective mechanisms operate during gestation to correct the harmful effects of anti-fetal immunity and to support a healthy pregnancy outcome. Pregnancy is characterized by gross alterations in endocrine functions. Progesterone is indispensable for pregnancy and humans, and it affects immune functions both directly and via mediators. The progesterone-induced mediator - PIBF - acts in favor of Th2-type immunity, by increasing Th2 type cytokines production. Except for implantation and parturition, pregnancy is characterized by a Th2-dominant cytokine pattern. Progesterone and the orally-administered progestogen dydrogesterone upregulate the production of Th2-type cytokines and suppress the production of Th1 and Th17 cytokine production in vitro. This is particularly relevant to the fact that the Th1-type cytokines TNF-α and IFN-γ and the Th17 cytokine IL-17 have embryotoxic and anti-trophoblast activities. These cytokine-modulating effects and the PIBF-inducing capabilities of dydrogesterone may contribute to the demonstrated beneficial effects of dydrogesterone in recurrent spontaneous miscarriage and threatened miscarriage. IL-17 and IL-22 produced by T helper cells are involved in allograft rejection, and therefore could account for the rejection of paternal HLA-C-expressing trophoblast. Th17 cells (producing IL-17 and IL-22) and Th22 cells (producing IL-22) exhibit plasticity and could produce IL-22 and IL-17 in association with Th2-type cytokines or with Th1-type cytokines. IL-17 and IL-22 producing Th cells are not harmful for the conceptus, if they also produce IL-4. Another important protective mechanism is connected with the expansion and action of regulatory T cells, which play a major role in the induction of tolerance both in pregnant women and in tumour-bearing patients. Clonally-expanded Treg cells increase at the feto-maternal interface and in tumour-infiltrating regions. While in cancer patients, clonally-expanded Treg cells are present in peripheral blood, they are scarce in pregnancy blood, suggesting that fetal antigen-specific tolerance is restricted to the foeto-maternal interface. The significance of Treg cells in maintaining a normal materno-foetal interaction is underlined by the fact that miscarriage is characterized by a decreased number of total effector Treg cells, and the number of clonally-expanded effector Treg cells is markedly reduced in preeclampsia. In this review we present an overview of the above mechanisms, attempt to show how they are connected, how they operate during normal gestation and how their failure might lead to pregnancy pathologies.


Assuntos
Citocinas/metabolismo , Hormônios/metabolismo , Reprodução/fisiologia , Animais , Citocinas/genética , Suplementos Nutricionais , Didrogesterona/administração & dosagem , Feminino , Regulação da Expressão Gênica , Hormônios/genética , Humanos , Imunomodulação , Troca Materno-Fetal/imunologia , Gravidez , Progesterona/genética , Progesterona/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
3.
J Assist Reprod Genet ; 38(8): 2049-2059, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33791895

RESUMO

Non-obstructive azoospermia (NOA) is one of the leading causes of male factor infertility, which results from impaired spermatogenesis. Currently, the sole feasible therapeutic option for men with NOA to father their biologic children is sperm retrieval by testicular sperm extraction (TESE) approaches followed by an intracytoplasmic sperm injection program. Nevertheless, the rate of sperm retrieval from NOA men following TESE has remained as low as 50%, leading to a significant number of unsuccessful TESE operations. Given that TESE is associated with multiple side effects, the prediction of TESE outcome preoperatively can abolish unnecessary operations and thereby prevent NOA patients from sustaining adverse side effects. As the process of spermatogenesis is under the regulation of hormones, the hormonal profile of serum and/or seminal plasma may contain useful information about spermatogenesis status and can potentially predict the chance of sperm retrieval from NOA patients. A large body of literature is available on the predictive capability of different serum and seminal plasma hormones such as FSH, LH, testosterone, inhibin B, AMH, estradiol, prolactin, and leptin in a stand-alone basis or combinational fashion with respect to the TESE outcome. The present review aimed to evaluate the potential of these hormonal markers as noninvasive predictors of sperm retrieval in men with NOA.


Assuntos
Azoospermia/genética , Hormônios/sangue , Sêmen/metabolismo , Espermatogênese/genética , Azoospermia/sangue , Azoospermia/patologia , Estradiol/sangue , Hormônio Foliculoestimulante Humano/sangue , Hormônios/genética , Hormônios/metabolismo , Humanos , Inibinas/sangue , Leptina/sangue , Hormônio Luteinizante/sangue , Masculino , Prolactina/sangue , Injeções de Esperma Intracitoplásmicas , Recuperação Espermática , Testosterona/sangue
4.
J Mol Biol ; 433(7): 166843, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33539880

RESUMO

Kisspeptin receptor (Kiss1R) is an important receptor that plays central regulatory roles in reproduction by regulating hormone release in the hypothalamus. We hypothesize that the formation of heterocomplexes between Kiss1R and other hypothalamus G protein-coupled receptors (GPCRs) affects their cellular signaling. Through screening of potential interactions between Kiss1R and hypothalamus GPCRs, we identified G protein-coupled estrogen receptor (GPER) as one interaction partner of Kiss1R. Based on the recognised function of kisspeptin and estrogen in regulating the reproductive system, we investigated the Kiss1R/GPER heterocomplex in more detail and revealed that complex formation significantly reduced Kiss1R-mediated signaling. GPER did not directly antagonize Kiss1R conformational changes upon ligand binding, but it rather reduced the cell surface expression of Kiss1R. These results therefore demonstrate a regulatory mechanism of hypothalamic hormone receptors via receptor cooperation in the reproductive system and modulation of receptor sensitivity.


Assuntos
Hipotálamo/metabolismo , Complexos Multiproteicos/genética , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1/genética , Animais , Hormônios/biossíntese , Hormônios/genética , Humanos , Complexos Multiproteicos/ultraestrutura , Ligação Proteica/genética , Receptores de Superfície Celular/genética , Receptores de Estrogênio/ultraestrutura , Receptores Acoplados a Proteínas G/ultraestrutura , Receptores de Kisspeptina-1/ultraestrutura , Transdução de Sinais/genética
5.
BMC Cancer ; 20(1): 884, 2020 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-32928177

RESUMO

BACKGROUND: In recent years, there is increasing evidence showing a beneficial outcome (e.g. progression free survival; PFS) after metastases-directed therapy (MDT) with external beam radiotherapy (EBRT) or targeted surgery for oligometastatic hormone sensitive prostate cancer (oHSPC). However, many patients do not qualify for these treatments due to prior interventions or tumor location. Such oligometastatic patients could benefit from radioligand therapy (RLT) with 177Lu-PSMA; a novel tumor targeting therapy for end-stage metastatic castration-resistant prostate cancer (mCRPC). Especially because RLT could be more effective in low volume disease, such as the oligometastatic status, due to high uptake of radioligands in smaller lesions. To test the hypothesis that 177Lu-PSMA is an effective treatment in oHSPC to prolong PFS and postpone the need for androgen deprivation therapy (ADT), we initiated a multicenter randomized clinical trial. This is globally, the first prospective study using 177Lu-PSMA-I&T in a randomized multicenter setting. METHODS & DESIGN: This study compares 177Lu-PSMA-I&T MDT to the current standard of care (SOC); deferred ADT. Fifty-eight patients with oHSPC (≤5 metastases on PSMA PET) and high PSMA uptake (SUVmax > 15, partial volume corrected) on 18F-PSMA PET after prior surgery and/or EBRT and a PSA doubling time of < 6 months, will be randomized in a 1:1 ratio. The patients randomized to the interventional arm will be eligible for two cycles of 7.4GBq 177Lu-PSMA-I&T at a 6-week interval. After both cycles, patients are monitored every 3 weeks (including adverse events, QoL- and xerostomia questionnaires and laboratory testing) at the outpatient clinic. Twenty-four weeks after cycle two an end of study evaluation is planned together with another 18F-PSMA PET and (whole body) MRI. Patients in the SOC arm are eligible to receive 177Lu-PSMA-I&T after meeting the primary study objective, which is the fraction of patients who show disease progression during the study follow up. A second primary objective is the time to disease progression. Disease progression is defined as a 100% increase in PSA from baseline or clinical progression. DISCUSSION: This is the first prospective randomized clinical study assessing the therapeutic efficacy and toxicity of 177Lu-PSMA-I&T for patients with oHSPC. TRIAL REGISTRATION: Clinicaltrials.gov identifier: NCT04443062 .


Assuntos
Lutécio/administração & dosagem , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Radioisótopos/administração & dosagem , Antagonistas de Androgênios/administração & dosagem , Antagonistas de Androgênios/efeitos adversos , Progressão da Doença , Hormônios/genética , Hormônios/metabolismo , Humanos , Lutécio/efeitos adversos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/radioterapia , Intervalo Livre de Progressão , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Neoplasias de Próstata Resistentes à Castração/patologia , Neoplasias de Próstata Resistentes à Castração/radioterapia , Qualidade de Vida , Radioisótopos/efeitos adversos , Compostos Radiofarmacêuticos/administração & dosagem , Resultado do Tratamento
6.
Genes Genomics ; 42(11): 1319-1326, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32980992

RESUMO

BACKGROUND: The acute hypoxic injury caused by the plain population entering the plateau in a short period of time has become the main cause of endangering the health of the people who rush into the plateau. OBJECTIVE: The study aimed to identify the key genes which participate in resisting the acute hypoxic injury in SD Rats by transcriptomic profile analysis. METHODS: 48 Sprague Dawley (SD) male rats were enrolled and randomly divided into four groups (0h, 24h, 48h, 72h) and housed in hypobaric hypoxia chamber with altitude 6000m for different periods of time to make them acute hypoxic injury. The transcriptomic profile of the lung tissue of the rats was analysed by RNA second-generation sequencing combined with bioinformatics analysis. RESULTS: The results of GO and KEGG function classification analysis revealed that the differential expression genes enriched in steroid hormone synthesis pathway especially in 48h group compared to F0 group. Further analysis revealed that Farnesyl Diphosphate Farnesyl Transferase 1 (fdft1) gene encoding a rate-limiting enzyme in steroid hormone synthesis pathway was significant differently expressed between the groups. The expression levels of fdft1 gene were further verified by RT-PCR and Western-blot methods. CONCLUSIONS: The results suggest that fdft1 gene plays an important role in responding to acute hypoxic injury by regulating steroid hormone biosynthesis.


Assuntos
Farnesil-Difosfato Farnesiltransferase/genética , Hipóxia/genética , Lesão Pulmonar/genética , Pulmão/metabolismo , Altitude , Animais , Perfilação da Expressão Gênica , Hormônios/biossíntese , Hormônios/genética , Hipóxia/fisiopatologia , Lipogênese/genética , Pulmão/fisiopatologia , Lesão Pulmonar/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley/genética , Ratos Sprague-Dawley/fisiologia , Esteroides/biossíntese , Transcriptoma/genética
7.
Horm Metab Res ; 52(8): 551-552, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32791541

RESUMO

About 3000 thousand years ago Marcus Aurelius said: "Time is a sort of river of passing events, and strong is its current; no sooner is a thing brought to sight than it is swept by and another takes its place, and this too will be swept away…" 1.


Assuntos
Pesquisa Biomédica/história , Hormônios/história , Neoplasias/história , Publicações Periódicas como Assunto/história , Receptores de Esteroides/metabolismo , História do Século XX , História do Século XXI , Hormônios/genética , Hormônios/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo
8.
Anim Reprod Sci ; 221: 106569, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32861119

RESUMO

Supplementation of N-carbamylglutamate (NCG) improves gestation outcomes, with increased piglet within-litter uniformity of birth weight and reduced peripheral steroid concentrations in pregnant sows and ewes. It was hypothesized that the effect of NCG on placental function results from direct effects on the placental trophoblasts. There, therefore, was investigation of the effects of NCG on pig placental trophoblast (pTr) steroidogenesis, mRNA transcript abundance, and cell proliferation in vitro. The pTr were treated with NCG in serum-free medium for 24-48 h. Treatment with NCG inhibited pTr progesterone, androstenedione, testosterone (all P <  0.01), and estradiol (P <  0.05) production, whereas it promoted (P <  0.05) pTr proliferation. Treatment with NCG suppressed (P <  0.05) the relative abundances of CYP11A1, CYP19A1, and CASP3 and increased abundances of CCDN1 (P <  0.01) and CDK4 (P <  0.05) mRNA transcripts in pTr, whereas NCG treatment had no effect (P >  0.10) on relative abundances of StAR, HSD17B4, or HSD3B mRNA transcripts. Treatments with NCG can increase pTr cell numbers of sows through upregulating CCND1 and CDK4 and suppressing CASP3 mRNA transcript abundances, while modulating steroidogenesis through effects on CYP11A1 and CYP19A1 mRNA transcript abundances. It is concluded that NCG may have a direct action on pTr and may regulate placental function by suppressing pTr differentiation as a consequence of lesser steroid synthesis while promoting pTr proliferation and inhibiting apoptosis in sows.


Assuntos
Glutamatos/farmacologia , RNA Mensageiro/metabolismo , Suínos/fisiologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Suplementos Nutricionais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hormônios/genética , Hormônios/metabolismo , Gravidez , RNA Mensageiro/genética , Suínos/genética
9.
FASEB J ; 34(8): 11243-11256, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32648604

RESUMO

In contrast to most rhodopsin-like G protein-coupled receptors, the glycoprotein hormone receptors (GPHR) have a large extracellular N-terminus for hormone binding. The hormones do not directly activate the transmembrane domain but mediate their action via a, thus, far only partially known Tethered Agonistic LIgand (TALI). The existence of such an intramolecular agonist was initially indicated by site-directed mutation studies and activating peptides derived from the extracellular hinge region. It is still unknown precisely how TALI is involved in intramolecular signal transmission. We combined systematic mutagenesis studies at the luteinizing hormone receptor and the thyroid-stimulating hormone receptor (TSHR), stimulation with a drug-like agonist (E2) of the TSHR, and structural homology modeling to unravel the functional and structural properties defining the TALI region. Here, we report that TALI (a) is predisposed to constitutively activate GPHR, (b) can by itself rearrange GPHR into a fully active conformation, (c) stabilizes active GPHR conformation, and (d) is not involved in activation of the TSHR by E2. In the active state conformation, TALI forms specific interactions between the N-terminus and the transmembrane domain. We show that stabilization of an active state is dependent on TALI, including activation by hormones and constitutively activating mutations.


Assuntos
Glicoproteínas/metabolismo , Hormônios/metabolismo , Glicoproteínas/genética , Células HEK293 , Hormônios/genética , Humanos , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutagênese/genética , Mutagênese Sítio-Dirigida/métodos , Mutação/genética , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica/genética , Domínios Proteicos/genética , Domínios Proteicos/fisiologia , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo , Transdução de Sinais/genética
10.
Cancer ; 126(17): 4059-4066, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32614992

RESUMO

BACKGROUND: Physical activity (PA) is recommended for women with breast cancer (BC); however, data are sparse on the association of PA with quality of life (QOL) and patient-reported symptoms for women on adjuvant endocrine therapy (AET). METHODS: Women with hormone receptor-positive BC who were taking AET completed standardized surveys about their health-related QOL, AET-related symptoms, and levels of PA using validated measures. A Wald chi-square test and an analysis of variance were used to assess associations with PA and independent variables. Generalized linear regression analyses assessed associations between PA, QOL, and AET-related symptoms. RESULTS: The analytic cohort included 485 Black and White women. Black race, a high body mass index (BMI), and being on aromatase inhibitors (vs tamoxifen) were associated with lower PA in a bivariate analysis. In a multivariate analysis, lower self-reported PA was associated with a high BMI (P = .02) and chemotherapy uptake (P = .006). Better health-related QOL (P = .01), less severe overall AET-related symptoms (P = .02), and less severe gynecological symptoms (P = .03) were associated with increasing levels of moderate PA. CONCLUSIONS: Among women taking AET, moderate levels of PA may be associated with fewer medication-related symptoms and overall better ratings of health-related QOL. Because of the low levels of PA observed in the sample overall and particularly for Black women, identifying successful strategies to promote PA are needed.


Assuntos
Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Exercício Físico , Hormônios/genética , Tamoxifeno/uso terapêutico , Adulto , Negro ou Afro-Americano/genética , Idoso , Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/efeitos adversos , Índice de Massa Corporal , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Quimioterapia Adjuvante/efeitos adversos , Terapia Combinada , Feminino , Humanos , Adesão à Medicação , Pessoa de Meia-Idade , Qualidade de Vida , Tamoxifeno/efeitos adversos , População Branca/genética
11.
Life Sci ; 256: 117922, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32522569

RESUMO

To test the hypothesis of STC-1 participation in maintenance of glucose homeostasis in fed and fasting (48 h) rats, we investigated that this hormone may be implicated in the regulation of renal gluconeogenesis pathway from lactate and lactate oxidation in renal cortex and medulla. Our results demonstrate the hSTC-1 role on lactate metabolism in the renal cortex and medulla from fed and fasting rats. hSTC-1 increased the gluconeogenesis activity in fed state in renal cortex, and this increase was induced by raise in Pck1 gene expression. In fasting animals hSTC-1 increase the renal medulla gluconeogenesis activity, but Pck1 gene expression was not alter. The stimulatory effect of hSTC-1 on 14C-lactate oxidation occurred only in the renal cortex from fed rats. These findings show the hSTC-1 contribution to lactate homeostasis and supplies glucose to other tissues. This response may represent a strategy of action of STC-1 in response to fasting stress as postulated by different authors. On the other hand, hSTC-1 acts downstream of adenylcyclase pathway, decreasing the gluconeogenesis activity induced by cAMP intracellular increase or stimulating the phosphodiesterase activity in the renal cortex. However, no hSTC-1 effect on 14C-lactate oxidation was found after increase in the intracellular cAMP. The findings also revealed that the renal cortex and medulla respond differently to hSTC-1, possibly due to the higher level of STC-1 gene expression in inner renal medulla than in renal cortex.


Assuntos
Gluconeogênese/efeitos dos fármacos , Glicoproteínas/metabolismo , Hormônios/metabolismo , Rim/metabolismo , Lactatos/metabolismo , Proteínas Recombinantes/metabolismo , Adenilil Ciclases/metabolismo , Animais , Dióxido de Carbono/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Glicoproteínas/genética , Hormônios/genética , Humanos , Córtex Renal/metabolismo , Medula Renal/metabolismo , Masculino , Oxirredução , Diester Fosfórico Hidrolases/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/genética , Transdução de Sinais
12.
Orv Hetil ; 161(25): 1028-1034, 2020 06.
Artigo em Húngaro | MEDLINE | ID: mdl-32516120

RESUMO

During conception (fusion of maternal and paternal germ cells) a new entity is formed, which has individual structure and functions. From its gene-pool (genome) epigenetic regulation selects those genes which are durably or acutely working, supplying a theoretically lifelong program. However, this program could be changed spontaneously or artificially, and there are life periods when the spontaneous changes are especially frequent and the sensitivity to physiological or artificial (man-made) factors is high. The basic sensitive (most vulnerable) period is the perinatal one, when the program agglomerated (and this is manifested in the faulty hormonal imprinting and DOHaD theories: perinatal adverse effects can cause diseases in adults), however, later periods are also sensitive. Such rather sensitive periods are the puberty and periadolescence as well as weaning, nevertheless, in any periods of life, cells or cell groups could be epigenetically imprinted, if the cells are in the state of differentiation, independent of the age or developmental state of the complete organism. Earlier mainly natural molecules (products of volcanic eruptions or phytoestrogens, mykotoxins, tobacco) threatened the program, today man-made artificial molecules (endocrine disruptors) can reprogram the visibly stable program, by a single encounter with hormone receptors at the periods of sensitivity (faulty hormonal imprinting) with life-long consequences (altered cell functions, inclination to diseases, manifestation of diseases, etc., provoked by functional teratogens). The deformed program is inherited to the offspring generations, where further program transformations are taking place on the inherited (transformed) program. As the amount and variants of man-made endocrine disruptors are enormously growing in the human environment and its important parts act during the developmentally most sensitive periods, the diseases provoked by them in adults expectedly will be enormously accruing. Orv Hetil. 2020; 161(25): 1028-1034.


Assuntos
Disruptores Endócrinos/efeitos adversos , Epigênese Genética , Impressão Genômica , Hormônios/metabolismo , Anormalidades Congênitas , Feminino , Hormônios/genética , Humanos , Gravidez , Maturidade Sexual
13.
Aging (Albany NY) ; 10(10): 2557-2569, 2018 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-30375982

RESUMO

Although there are numerous hypotheses explaining the nature of aging and associated processes, two concepts are dominant: (i) aging is a result of cell-autonomous processes, such as the accumulation of DNA mutations, aberrant methylations, protein defects, and shortening of telomeres, leading to either inhibition of cellular proliferation and death of non-dividing terminally differentiated cells or tumor development; (ii) aging is a result of a central program that is switched on at a specific stage of organismic development. The microRNA-based endocrine regulation hypothesis combines the two above concepts by proposing central regulation of cell death occurrences via hypothalamus-pituitary gland (PG)-secreted miRNA hormones, the expression and/or secretion of which are regulated by sex hormones. This hypothesis explains such well-known phenomena as inverse comorbidity of either cancer or Alzheimer's (AD) and other neurodegenerative diseases; higher AD morbidity and lower frequency of many common types of cancer in women vs. men; higher risk of early AD and lower risk of cancer in subjects with Down syndrome; longer life expectancy in women vs. men and much lower sex-dependent differences, if any, in other mammals; increased lifespans due to hypophysectomy or PG hypofunction; and parabiotic effects of blood or plasma transfusions between young and old animals.


Assuntos
Envelhecimento/metabolismo , Senescência Celular , Sistema Endócrino/metabolismo , Hormônios/metabolismo , MicroRNAs/metabolismo , Fatores Etários , Envelhecimento/genética , Envelhecimento/patologia , Animais , Sistema Endócrino/patologia , Regulação da Expressão Gênica no Desenvolvimento , Hormônios/genética , Humanos , Modelos Biológicos , Via Secretória , Transdução de Sinais
14.
BMC Med Genet ; 19(1): 149, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30134857

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) is the main cause of female infertility. Interactions among genetic, biochemical, and immunological factors can affect the pathogenesis of PCOS. As a proinflammatory cytokine, tumor necrosis factor-α (TNF-α) plays an important role in this regard. The present study aimed to evaluate the association of the rs361525 gene single-nucleotide polymorphism (SNP) and TNF-α serum levels with the hormonal and biochemical characteristics of PCOS in Iranian individuals. METHODS: The SNP rs361525 in the TNF-α gene was analyzed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) in a total of 111 PCOS patients and 105 healthy females. Serum levels of TNF-α, lipid and hormone profiles, and biochemical factors were measured using enzyme-linked immunosorbent assay (ELISA) and calorimetric methods, as appropriate. RESULTS: The TNF-α serum level was higher in women with PCOS compared with the control group (p <  0.0001), and it was significantly correlated with the homeostasis model assessment (HOMA) factor (r = 0.138, p <  0.05). No significant differences were found in the genotype and allelic frequencies between the two groups (p >  0.05). Higher levels and significant differences were found for the HOMA factor, luteinizing hormone/follicle-stimulating hormone (LH/FSH), testosterone, and body mass index (BMI) in the PCOS group compared with the control group (p <  0.0001). High LH/FSH ratios (odds ratio [OR] = 1.98, 95% confidence interval [CI] = 1.20-3.28, p <  0.01), and high HOMA factor (OR = 5.04, 95% CI = 2.82-9.01, p <  0.001) were significantly associated with an increased risk of PCOS. CONCLUSIONS: Despite the lack of significant difference between rs361525 polymorphism of the TNF-α gene and PCOS, the serum level of TNF-α was increased in PCOS patients and positively correlated with the HOMA factor. Elevation of the LH/FSH ratio and HOMA for insulin resistance (HOMA-IR) increased the risk of PCOS. Therefore, TNF-α could indirectly contribute to PCOS progression.


Assuntos
Predisposição Genética para Doença/genética , Síndrome do Ovário Policístico/genética , Polimorfismo de Nucleotídeo Único/genética , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , Feminino , Frequência do Gene/genética , Hormônios/genética , Humanos , Resistência à Insulina/genética , Irã (Geográfico) , Polimorfismo de Fragmento de Restrição/genética
15.
Poult Sci ; 97(8): 2926-2933, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-29750260

RESUMO

The major objective of this study was to assess the expression of mitochondrial hormone receptors for progesterone (PR), estrogen (ER), glucocorticoid (GR), thyroid (TR), and insulin (IR) in avian muscle cells (quail muscle 7, QM7) and in breast muscle of quail and broilers. Visualization of receptor location in QM7 cells was accomplished by immunofluorescence. QM7 cells were stained with Mito Tracker Deep Red CMX, fixed in methanol, immune stained with anti-PR, -GR, -TR, -IR, and -ER primary antibodies overnight at 4°C, and visualized with Alexa Fluor 488-conjugated secondary antibody. After staining the nucleus with 4',6-diamidino-2-phenylindole, images were obtained by immunofluorescence microscopy. Merged images revealed the presence of all 5 hormone receptors on mitochondria in QM7 cells. Western blot analysis identified; (a) the ß-isoform of the PR, (b) the α-isoform of GR, (c) the α-receptor of TR, (d) the ß-subunit of IR, and (e) the α-isoform of the ER on mitochondria isolated from broiler breast muscle. Similar results were obtained in quail breast muscle mitochondria with the exception that the α-isoform of the GR was not detected. To our knowledge, this is the first report of hormone receptors (PR, TR, GR, IR, and ER) on mitochondria in avian cells. We hypothesize that these receptors could play important roles in regulating mitochondrial function in avian muscle cells.


Assuntos
Proteínas Aviárias/genética , Galinhas/genética , Coturnix/genética , Hormônios/genética , Mitocôndrias/genética , Receptores Citoplasmáticos e Nucleares/genética , Animais , Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Coturnix/metabolismo , Hormônios/metabolismo , Masculino , Mitocôndrias/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo
16.
Poult Sci ; 97(6): 2189-2202, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29554306

RESUMO

The present study was conducted to determine the changes in concentrations of hormones and growth factors and their related receptor gene expressions in crop tissue, relative organ weight, and serum biochemical parameters in male and female pigeons during incubation and chick-rearing periods under artificial farming conditions. Seventy-eight pairs of 60-week-old White King pigeons with 2 fertile eggs per pair were randomly divided into 13 groups by different breeding stages. Serum prolactin and insulin-like growth factor-1 (IGF-1) concentrations in crop tissue homogenates were the highest in both male and female pigeons at 1 d of chick-rearing (R1), while epidermal growth factor (EGF) in female pigeons peaked at d 17 of incubation (I17) (P < 0.05). mRNA expression of the prolactin and EGF receptors in the crop tissue increased at the end of incubation and the early chick-rearing stage in both sexes. However, estrogen, progesterone, and growth hormone receptor expression each decreased during the early chick-rearing stage (P < 0.05). In male pigeons, IGF-1 receptor gene expression reached its peak at R7, while in female pigeons, it increased at the end of incubation. The relative weight of breast and abdominal fat in both sexes and thighs in the males was lowest at R7, and then gradually increased to the incubation period level. Serum total protein, albumin, and globulin concentrations increased to the highest levels at I17 (P < 0.05). Total cholesterol, triglyceride, and low-density lipoprotein reached their highest values at I17 in male pigeons and R25 in female pigeons (P < 0.05). In conclusion, hormones, growth factors, and their receptors potentially underlie pigeon crop tissue development. Changes in organs and serum biochemical profiles suggested their different breeding-cycle patterns with sexual effects.


Assuntos
Columbidae/anatomia & histologia , Columbidae/fisiologia , Expressão Gênica , Comportamento de Nidação , Criação de Animais Domésticos , Animais , China , Columbidae/sangue , Papo das Aves/metabolismo , Feminino , Hormônios/genética , Hormônios/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Tamanho do Órgão/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
17.
BMC Cancer ; 18(1): 280, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29530003

RESUMO

BACKGROUND: Reproductive factors are well known risk factors for breast cancer; however, little is known about how genetic variants in hormonal pathways interact with that relationship. METHODS: One thousand one hundred thirty nine cases of breast cancer in women and 1322 frequency-matched controls were compared. Genetic variants in hormonal pathways (identified in the Kyoto Encyclopedia of Genes and Genomes) were screened according to their relationship with breast cancer using the Cochran-Armitage statistic. Information on reproductive factors was obtained using a face-to-face questionnaire. The interaction among the selected genetic variants and reproductive factors was tested with logistic regression. RESULTS: Concerning C allele in rs2229712, compared to nulliparity in non-carriers the ORs for 1-2 and > 2 deliveries were 0.48 (0.28-0.81) and 0.34 (0.19-0.59), and in C carriers they were 0.92 (0.42-1.98) and 0.71 (0.31-1.61). Similar results were found in women carrying the C allele in rs1269851. Carriers of Allele T in rs35652107 and allele C in rs6018027 had the delivery number effect more pronounced. CONCLUSIONS: The number of deliveries had a dose-response protective effect on breast cancer; women carrying C allele in rs2229712 did not benefit from this protective effect.


Assuntos
Neoplasias da Mama/genética , Predisposição Genética para Doença , Paridade/genética , Reprodução/genética , Fator 6 Ativador da Transcrição , Adulto , Idoso , Alelos , Fatores de Transcrição de Zíper de Leucina Básica/genética , Neoplasias da Mama/complicações , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Feminino , Hormônios/genética , Humanos , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Mutação , Proteínas do Tecido Nervoso/genética , Polimorfismo de Nucleotídeo Único/genética , Gravidez , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Fatores de Risco , Espanha , Inquéritos e Questionários , Quinases da Família src/genética
18.
J Dairy Sci ; 101(1): 690-704, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29128220

RESUMO

Knowledge of the molecules used by the maternal reproductive tract to regulate development of the preimplantation embryo is largely incomplete. The goal of the present experiment was to identify candidates for this function. The approach was to assess expression patterns in the endometrium and oviduct of 93 genes encoding for hormones, growth factors, chemokines, cytokines, and WNT-related molecules. Results show that all of the genes were expressed in the reproductive tract. Expression in oviduct was affected by day of the estrous cycle for 21 genes with 11 genes having highest expression at estrus (CCL21, CTGF, CXCL10, CXCL16, DKK3, FGF10, IL18, IL33, IL34, PGF, and SFRP2), 1 gene at d 3 (WNT4), 8 at d 5 (BMP7, HGF, IL6, SFRP1, TGFB1, WIF1, WNT2, and WNT5A), and 1 at d 7 (IK). For endometrium, expression of 34 genes was affected by day of the estrous cycle with 11 having highest expression at d 0 (BMP7, CCL14, CCL21, CCL26, CTGF, CXCL12, IGF2, IL16, IL33, SFRP2, and WIF1), 2 at d 3 (HDGF, IL15), 14 at d 5 (CSF2, CX3CL1, CXCL3, FGF1, FGF2, GRO1, HGF, IGF1, IL1B, IL8, SFRP1, SFRP4, WNT5A, and WNT16), and 7 at d 7 (CXCL16, FGF13, HDGFRP2, TDGF1, VEGFB, WNT7A, and WNT11). Results are consistent with a set of genes regulated by estradiol early in the estrous cycle and another set regulated by progesterone later in the cycle. The cell-signaling genes identified here as being expressed in the oviduct and endometrium could serve to regulate early embryonic development in a stage-of-pregnancy-specific manner.


Assuntos
Bovinos/fisiologia , Desenvolvimento Embrionário/genética , Endométrio/metabolismo , Tubas Uterinas/metabolismo , Animais , Blastocisto/fisiologia , Quimiocinas/genética , Citocinas/genética , Desenvolvimento Embrionário/fisiologia , Estradiol/fisiologia , Ciclo Estral , Estro , Feminino , Expressão Gênica , Hormônios/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Gravidez , Transdução de Sinais/genética , Proteínas Wnt/genética
19.
Sci Rep ; 7(1): 14156, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29074860

RESUMO

The study explored differences in the steroidogenic pathway between obese and nonobese women with polycystic ovary syndrome (PCOS) using liquid chromatography-tandem mass spectrometry (LC-MS/MS). 1044 women with PCOS (including 350 lean, 312 overweight and 382 obese) and 366 control women without PCOS (including 203 lean, 32 overweight and 131 obese) were enrolled. The differences in steroid hormones were amplified in lean PCOS versus lean controls compared with obese PCOS versus obese controls. Compared with obese PCOS, lean PCOS demonstrated increased dehydroepiandrosterone sulfate (P = 0.015), 17-hydropregnenolone (P = 0.003), 17-hydroprogesterone (17-OHP) (P < 0.001), progesterone (P < 0.001) and estrone (P < 0.001) levels. Enzyme activity evaluation showed that lean PCOS had increased activity of P450c17 (17-hydropregnenolone/pregnenolone, P < 0.001), P450aro (P < 0.001), 3ßHSD2 (progesterone/ pregnenolone and 17-OHP/17-hydropregnenolone, both P < 0.001) and decreased activity of P450c21(11-deoxycorticorsterone/progesterone and 11-deoxycortisol/17-OHP, P < 0.001). Moreover, we found higher frequencies of CYP21A2- (encoding P450c21) c.552 C > G (p. D184E) in lean PCOS compared with obese PCOS patients (P = 0.006). In conclusion, this study demonstrated for the first time that the adrenal-specific enzyme P450c21 showed decreased activity in lean PCOS patients, and that the adrenal androgen excess may play different roles in lean and obese PCOS patients, which represents as different enzyme activity in the steroidogenic pathway.


Assuntos
Hormônios/sangue , Obesidade/sangue , Síndrome do Ovário Policístico/sangue , Esteroides/sangue , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , Estudos Transversais , Sulfato de Desidroepiandrosterona/sangue , Feminino , Hormônios/genética , Humanos , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Progesterona/sangue , Esteroide 17-alfa-Hidroxilase/genética , Esteroide 21-Hidroxilase/genética
20.
Toxicol Appl Pharmacol ; 336: 49-54, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29032082

RESUMO

Triclosan (TCS), a member of the class of compounds called pharmaceutical and personal care products (PPCPs), is a broad antibacterial and antifungal agent found in a lot of consumer products. However, TCS hormone effect mechanism in teleost female fish is not clear. Female Yellow River carp (Cyprinus carpio) were exposed to 1/20, 1/10 and 1/5 LC50 TCS (96h LC50 of TCS to carp) under semi-static conditions for 42days. Vitellogenin (Vtg), 17ß-estradiol (E2), testosterone(T), estrogen receptor (Er), gonadotropin (GtH), and gonadotropin-releasing hormone (GnRH) levels were measured by enzyme-linked immunosorbent assay (ELISA). Meanwhile, we also examined the mRNA expressions of aromatase, GtHs-ß, GnRH, and Er by quantitative real-time PCR (qRT-PCR). The results indicated that 1/5 LC50 TCS induced Vtg in hepatopancreas of female carps by interference with the hypothalamic-pituitary-gonadal (HPG) axis at multiple potential loci through three mechanisms: (a) TCS exposure enhanced the mRNA expression of hypothalamus and gonadal aromatase which converts androgens into estrogens, subsequently increasing serum concentrations of E2 to induce Vtg in hepatopancreas; (b) TCS treatment increased GnRH and GtH-ß mRNA expression and secretion, causing the disturbance of reproductive endocrine and the increase of E2 to induce Vtg in hepatopancreas; (c) TCS exposure enhanced synthesis and secretion of Er, then it bound to Er to active Vtg synthesis. These mechanisms showed that TCS may induce Vtg production in female Yellow River carp by Er-mediated and non-Er-mediated pathways.


Assuntos
Carpas/metabolismo , Disruptores Endócrinos/toxicidade , Estrogênios/toxicidade , Gônadas/efeitos dos fármacos , Hormônios/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Triclosan/toxicidade , Animais , Aromatase/genética , Aromatase/metabolismo , Carpas/genética , Estradiol/metabolismo , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/genética , Gonadotropinas Hipofisárias/metabolismo , Gônadas/metabolismo , Hepatopâncreas/efeitos dos fármacos , Hepatopâncreas/metabolismo , Hormônios/genética , Sistema Hipotálamo-Hipofisário/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Testosterona/metabolismo , Fatores de Tempo , Vitelogeninas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA