Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
2.
Gene ; 895: 148016, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-37981083

RESUMO

Understanding the pathophysiology of idiopathic central precocious puberty (ICPP) is essential, in view of its consequences on reproductive health and metabolic disorders in later life. Towards this, estimation of circulating levels of the neuropeptides, viz; Kisspeptin (Kp-10), Neurokinin B (NKB) and Neuropeptide Y (NPY), acting upstream to Gonadotropin-Releasing Hormone (GnRH), has shown promise. Insights can also be gained from functional studies on genetic variations implicated in ICPP. This study investigated the pathophysiology of ICPP in a girl by exploring the therapeutic relevance of the circulating levels of Kp-10, NKB, NPY and characterizing the nonsynonymous KISS1R variant, L364H, that she harbours, in a homozygous condition. Plasma levels of Kp-10, NKB and NPY before and after GnRH analog (GnRHa) treatment, were determined by ELISA. It was observed that GnRHa treatment resulted in suppression of circulating levels of Kp-10, NKB and NPY. Further, the H364 variant in KISS1R was generated by site directed mutagenesis. Post transient transfection of either L364 or H364 KISS1R variant in CHO cells, receptor expression was ascertained by western blotting, indirect immunofluorescence and flow cytometry. Kp-10 stimulated signalling response was also determined by phospho-ERK and inositol phosphate production. Structure-function studies revealed that, although the receptor expression in H364 KISS1R was comparable to L364 KISS1R, there was an enhanced signalling response through this variant at high doses of Kp-10. Thus, elevated levels of Kp-10, acting through H364 KISS1R, contributed to the manifestation of ICPP, providing further evidence that dysregulation of Kp-10/KISS1R axis impacts the onset of puberty.


Assuntos
Puberdade Precoce , Animais , Cricetinae , Feminino , Humanos , Cricetulus , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/genética , Neurocinina B/genética , Neurocinina B/metabolismo , Puberdade Precoce/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1/genética
3.
J Assist Reprod Genet ; 40(7): 1509-1522, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37338750

RESUMO

The endometrium is a dynamic tissue that undergoes extensive remodeling during the menstrual cycle and further gets modified during pregnancy. Different kinds of stem cells are reported in the endometrium. These include epithelial stem cells, endometrial mesenchymal stem cells, side population stem cells, and very small embryonic-like stem cells. Stem cells are also reported in the placenta which includes trophoblast stem cells, side population trophoblast stem cells, and placental mesenchymal stem cells. The endometrial and placental stem cells play a pivotal role in endometrial remodeling and placental vasculogenesis during pregnancy. The dysregulation of stem cell function is reported in various pregnancy complications like preeclampsia, fetal growth restriction, and preterm birth. However, the mechanisms by which it does so are yet elusive. Herein, we review the current knowledge of the different type of stem cells involved in pregnancy initiation and also highlight how their improper functionality leads to pathological pregnancy.


Assuntos
Placenta , Nascimento Prematuro , Recém-Nascido , Gravidez , Feminino , Humanos , Placenta/patologia , Nascimento Prematuro/patologia , Endométrio/patologia , Trofoblastos , Células-Tronco/fisiologia
4.
Gene ; 877: 147548, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37279863

RESUMO

GPER is a seven transmembrane G-protein-coupled estrogen receptor that mediates rapid estrogen actions. Large volumes of data have revealed its association with clinicopathological variables in breast tumors, role in epidermal growth factor (EGF)-like effects of estrogen, potential as a therapeutic target or a prognostic marker, and involvement in endocrine resistance in the face of tamoxifen agonism. GPER cross-talks with estrogen receptor alpha (ERα) in cell culture models implicating its role in the physiology of normal or transformed mammary epithelial cells. However, discrepancies in the literature have obfuscated the nature of their relationship, its significance, and the underlying mechanism. The purpose of this study was to assess the relationship between GPER, and ERα in breast tumors, to understand the mechanistic basis, and to gauge its clinical significance. We mined The Cancer Genome Atlas (TCGA)-BRCA data to examine the relationship between GPER and ERα expression. GPER mRNA, and protein expression were analyzed in ERα-positive or -negative breast tumors from two independent cohorts using immunohistochemistry, western blotting, or RT-qPCR. The Kaplan-Meier Plotter (KM) was employed for survival analysis. The influence of estrogen in vivo was studied by examining GPER expression levels in estrus or diestrus mouse mammary tissues, and the impact of 17ß-estradiol (E2) administration in juvenile or adult mice. The effect of E2, or propylpyrazoletriol (PPT, an ERα agonist) stimulation on GPER expression was studied in MCF-7 and T47D cells, with or without tamoxifen or ERα knockdown. ERα-binding to the GPER locus was explored by analysing ChIP-seq data (ERP000380), in silico prediction of estrogen response elements, and chromatin immunoprecipitation (ChIP) assay. Clinical data revealed significant positive association between GPER and ERα expression in breast tumors. The median GPER expression in ERα-positive tumors was significantly higher than ERα-negative tumors. High GPER expression was significantly associated with longer overall survival (OS) of patients with ERα-positive tumors. In vivo experiments showed a positive effect of E2 on GPER expression. E2 induced GPER expression in MCF-7 and T47D cells; an effect mimicked by PPT. Tamoxifen or ERα-knockdown blocked the induction of GPER. Estrogen-mediated induction was associated with increased ERα occupancy in the upstream region of GPER. Furthermore, treatment with 17ß-estradiol or PPT significantly reduced the IC50 of the GPER agonist (G1)-mediated loss of MCF-7 or T47D cell viability. In conclusion, GPER is positively associated with ERα in breast tumors, and induced by estrogen-ERα signalling axis. Estrogen-mediated induction of GPER makes the cells more responsive to GPER ligands. More in-depth studies are warranted to establish the significance of GPER-ERα co-expression, and their interplay in breast tumor development, progression, and treatment.


Assuntos
Receptor alfa de Estrogênio , Neoplasias Mamárias Animais , Animais , Feminino , Camundongos , Linhagem Celular Tumoral , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Ligação ao GTP/genética , Neoplasias Mamárias Animais/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico
5.
Int Immunopharmacol ; 119: 110250, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37163922

RESUMO

Triple-negative Breast cancer (TNBC) is a subtype of breast cancer (BC) that lacks expression for ER/PR/Her2 receptors and is associated with aggressive disease pathogenesis and the worst prognosis among other subtypes of BC. Accumulating evidence-based studies indicate the high immunogenic ability of TNBC tumors and the applicability of immunotherapeutic strategies to overcome therapy resistance and tumor recurrence in TNBC patients. However, not all TNBC patients respond equally well to current immunotherapies that mainly target the adaptive immune system for tumor rejection. Recent studies are contemplating the efficacy of tumor-associated macrophage (TAM) targeted therapies since these subpopulations of cells comprise one of the major components of tumor-infiltrating immune cells (TIIs) in the TNBC tumor microenvironment (TME) and play an essential role in priming the adaptive immune response mediators towards both antitumorigenic and pro-tumorigenic response facilitated by intercellular cross-talk between tumor cells and TAM populations present within TNBC-TME. The present review discusses these molecular mechanisms and their consequence on the progression of TNBC tumors. Also, the therapeutic strategies targeting candidate genes/pathways involved in molecular cross-talk between TAM-TNBC cells and their impact on the development and progression of TNBC tumors are also discussed.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Macrófagos Associados a Tumor/metabolismo , Recidiva Local de Neoplasia , Imunoterapia , Prognóstico , Microambiente Tumoral
6.
Am J Reprod Immunol ; 89(2): e13563, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35587052

RESUMO

A failure to achieve pregnancy after three or more embryo transfer cycles with high-quality blastocysts is referred to as recurrent implantation failure (RIF). RIF can be due to altered uterine factors or male factors or embryo factors. Disrupted endometrial receptivity, altered expression of genes in several pathways, immunologic disturbances in the peripheral blood and/or the endometrium, and epigenetic alterations are associated with RIF. Amongst the immunologic disturbances, altered Th1/Th2 ratio, altered NK cell and macrophage numbers are observed in women with RIF. However, not all women with RIF have the same kind of immune dysfunction suggesting that RIF is a heterogeneous condition associated with varied immune responses and one size may not fit all. Thus, personalized therapies based on the immune status of the patient are being tested in women with RIF. In general, women with a high Th1/Th2 ratio are offered Tacrolimus, while intravenous IgG is recommended in women with high NK cell numbers/HLA mismatch. Women with hyperactivated immune status in the uterus are offered progesterone support, prednisolone, vitamin E, and intralipid treatment to suppress inflammation and oxidative stress, while endometrial scratching and intrauterine hCG administration are offered to women with hypo-active immune status. There is a need for standardized tests for evaluation of immune status in patients and sufficiently powered randomized controlled trials for personalized therapies to determine which of these will be beneficial in women with RIF. Till then, the ART community should limit the use of such add-on interventions in women with RIF.


Assuntos
Implantação do Embrião , Infertilidade Feminina , Gravidez , Humanos , Masculino , Feminino , Implantação do Embrião/fisiologia , Útero/metabolismo , Endométrio/metabolismo , Transferência Embrionária
8.
J Mol Endocrinol ; 69(3): 431-444, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35917434

RESUMO

Endometrial cancer is the fourth most common malignancy in women and the precursor lesion is endometrial hyperplasia. HOXA10 is a transcription factor that plays key roles in endometrial functions such as the endowment of receptivity, embryo implantation, and trophoblast invasion. Herein, using testicular transgenesis, we developed transgenic mice that expressed a shRNA against HOXA10 and there was a nearly 70% reduction in the expression of HOXA10 in these animals. We observed that downregulation of HOXA10 led to the development of endometrial hyperplasia in the young animals (3 months), and as they aged (>1 year), most animals developed well-differentiated endometrial adenocarcinoma. In the endometrium of animals with reduced HOXA10, there was increased proliferation and elevated levels of ERα and ERß. In parallel, there was increased expression of Wnt4 and ß-Catenin, SOX9, and YAP1. We propose that chronic reduction in HOXA10 expression disrupts multiple pathways in the uterus that aids in the development of endometrial hyperplasia which progresses to endometrial cancer with age.


Assuntos
Hiperplasia Endometrial , Neoplasias do Endométrio , Animais , Implantação do Embrião/fisiologia , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/metabolismo , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Feminino , Proteínas Homeobox A10 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos
9.
Tissue Cell ; 73: 101670, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34710830

RESUMO

Endometrium is a dynamic tissue that undergoes extensive remodelling to attain a receptive state which is further modulated in presence of an embryo for successful initiation of pregnancy. Cadherins are the proteins of the junctional complex of which E-cadherin (E-Cad) is crucial for maintaining epithelial cell state and integrity of the epithelial barrier; gain of N-cadherin (N-Cad) in epithelial cells leads to epithelial to mesenchymal transition (EMT). In the present study, we investigated the expression of E-Cad and N-Cad in the mouse endometrial luminal epithelium and its modulation by estrogen, progesterone, and embryonic stimuli. We observed that E-Cad is diffusely expressed in the luminal epithelium of mouse endometrium during the estrus stage and upon estrogen treatment. It is apico-laterally and basolaterally sorted at the diestrus stage and in response to the combined treatment of estrogen and progesterone. In 3D spheroids of human endometrial epithelial cells, combined treatment with estrogen and progesterone led to lateral sorting of E-Cad without any effects on its mRNA levels. at the time of embryo implantation, there is loss of E-Cad along with the gain of N-Cad and SNAIL expression suggestive of EMT in the luminal epithelium. This EMT is possibly driven by embryonic stimuli as treatment with estrogen and progesterone did not lead to the gain of N-Cad expression in the mouse endometrium in vivo or in human endometrial epithelial cells in vitro. In conclusion, the present study demonstrates that steroid hormones directly affect E-Cad sorting in the endometrial epithelium which undergo EMT in response to embryonic stimuli.


Assuntos
Caderinas/metabolismo , Embrião de Mamíferos/metabolismo , Ovário/metabolismo , Esteroides/metabolismo , Animais , Membrana Celular/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Ciclo Estral , Feminino , Camundongos Endogâmicos C57BL
10.
Gene ; 794: 145746, 2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34062258

RESUMO

The expression of HOXB2, a homeobox transcription factor, is altered in a variety of solid tumors. Using an in vivo screen to identify regulators of breast tumor growth in murine mammary fat pads, Boimel and co-workers recently identified HOXB2 as a tumor suppressor. However, the mechanistic underpinnings of its role in breast cancer is not understood. Given the emerging interaction of estrogen-regulated gene expression and altered HOX gene expression network in the pathophysiology of breast cancer, this study addressed the relationship between estrogen signaling and HOXB2 expression. Using a mouse model and human breast cancer cell lines, we show that estrogen suppresses HOXB2 expression. Suppression of HOXB2 by PPT, a known ERα agonist, in MCF-7 and T47D cells indicated the involvement of ERα, which was confirmed by siRNA-mediated ERα knockdown experiments. In-silico analysis of the upstream promoter region revealed the presence of three putative EREs. Chromatin immunoprecipitation experiments showed that upon estrogen binding, ERα engaged with EREs in the 5' upstream region of HOXB2 in MCF-7 and T47D cells. Future investigations should address the implications of estrogen-mediated suppression on the proposed tumor suppressor function of HOXB2.


Assuntos
Neoplasias da Mama/genética , Regulação para Baixo , Receptor alfa de Estrogênio/genética , Proteínas de Homeodomínio/genética , Fenóis/administração & dosagem , Pirazóis/administração & dosagem , Fatores de Transcrição/genética , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos , Transplante de Neoplasias , Fenóis/farmacologia , Regiões Promotoras Genéticas , Pirazóis/farmacologia
11.
Mol Biotechnol ; 63(9): 828-839, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34089481

RESUMO

Monoclonal antibodies (mAbs) are one of the most significant molecules in protein therapeutics. They are employed in the field of immunology, oncology and organ transplant. They have been also been employed for alleviating several bacterial and viral infections. Moreover, they have revolutionized the area of targeted therapy and improved the quality of treatments, as compared to other cytotoxic drugs and therapies. mAbs bind to specific molecules on the antigen and exhibit specificity towards that molecule, i.e. epitope. Thus, mAbs have immense opportunity to be explored for personalized therapy. The introduction of targeted mAb-based therapeutics has promoted many important scientific achievements in rheumatology. This has warranted additional investigations for developing newer mAb producing clones, to supplement the limited industrial production of certain mAb therapeutics. In this investigation, an integrative approach comprising optimized expression, selection and expansion was adopted to develop a mammalian cell line expressing mAb against TNF-α.The resulting stable clone is anticipated to serve as an economic alternative to the industrial clones, especially for research purposes. The clone was constructed for development of biosimilar of the highly valued therapeutic antibody, Humira.


Assuntos
Adalimumab/biossíntese , Antirreumáticos/imunologia , Plasmídeos/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adalimumab/genética , Adalimumab/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/genética , Antirreumáticos/metabolismo , Bioensaio , Células CHO , Cricetulus , Expressão Gênica , Humanos , Plasmídeos/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
12.
Bioorg Med Chem Lett ; 44: 128132, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34022413

RESUMO

In our previous study, we had identified a 9-mer peptide (FSHß (89-97)) derived from seat belt loop of human FSHß and demonstrated its ability to function as FSHR antagonist in vivo. Structure analysis revealed that the four central residues 91STDC94 within this peptide may not be critical for receptor binding. In the present study, 91STDC94 residues were substituted with alanine to generate ΔFSHß 89-97(91STDC94/AAAA) peptide. Analogous to the parent peptide, ΔFSHß 89-97(91STDC94/AAAA) peptide inhibited binding of iodinated FSH to rat FSHR and reduced FSH-induced cAMP production. The peptide could impede granulosa cell proliferation leading to reduction in FSH-mediated ovarian weight gain in immature female rats. In these rats, peptide administration further downregulated androgen receptor and estrogen receptor-alpha expression and upregulated estrogen receptor-beta expression. The results indicate that substitution of 91STDC94 with alanine did not significantly alter FSHR antagonist activity of FSHß (89-97) peptide implying that these residues are not critical for FSH-FSHR interaction and can be replaced with non-peptidic moieties for development of more potent peptidomimetics.


Assuntos
Desenho de Fármacos , Hormônio Foliculoestimulante/farmacologia , Peptídeos/farmacologia , Peptidomiméticos , Receptores do FSH/antagonistas & inibidores , Animais , Sítios de Ligação/efeitos dos fármacos , Feminino , Hormônio Foliculoestimulante/química , Humanos , Modelos Moleculares , Ovário/efeitos dos fármacos , Peptídeos/química
14.
Am J Reprod Immunol ; 85(2): e13360, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33064348

RESUMO

Implantation of the embryo is a rate-limiting step for a successful pregnancy, and it requires an intricate crosstalk between the embryo and the endometrium. Extracellular vesicles (EVs) are membrane-enclosed, nano-sized structures produced by cells to mediate cell to cell communication and modulate a diverse set of biological processes. Herein, we review the involvement of EVs in the process of embryo implantation and endometrial diseases. EVs have been isolated from uterine fluid, cultured endometrial epithelial/stromal cells and trophectodermal cells. The endometrial epithelial and stromal/decidual cell-derived EVs and its cargo are internalized bythe trophoblast cells, and they regulate a diverse set of genes involved in adhesion, invasion and migration. Conversely, the embryo-derived EVs and its cargo are internalized by epithelial and immune cells of the endometrium for biosensing and immunomodulation required for successful implantation. EVs have also been shown to play a role in infertility, recurrent implantation failure, endometriosis, endometritis and endometrial cancer. Further research should set a stage for EVs as non-invasive "liquid biopsy" tools for assessment of endometrial health.


Assuntos
Endométrio/patologia , Vesículas Extracelulares/imunologia , Placenta/imunologia , Complicações na Gravidez/imunologia , Gravidez/imunologia , Trofoblastos/imunologia , Animais , Implantação do Embrião , Embrião de Mamíferos , Feminino , Humanos , Imunomodulação , Nanoestruturas , Complicações na Gravidez/patologia
15.
J Biosci ; 452020.
Artigo em Inglês | MEDLINE | ID: mdl-32975232

RESUMO

Endometriosis is a common disorder of unknown etiology, and non-surgical therapies are still a challenge. To understand the pathogenesis and preclinical testing of drugs for endometriosis, animal models are highly desirous. Herein, we carried out longitudinal characterization of a mouse model for endometriosis where uterine tissue was transplanted onto the intestinal mesentery. During the course of lesion development from day 15 to 60 post-induction, the ectopic endometrium became pale, fluid-filled and the animals developed peritoneal adhesions. Most lesions resembled a well-differentiated type of endometriosis and ~13% of animals had mixed type of lesions. There was extensive stromal compaction in the ectopic tissue. During the progression of endometriosis, there was increased proliferation of epithelial and stromal cells as evident by PCNA staining. Cyp19a1 (aromatase) mRNA was detected in the ectopic lesions on day 15 and 30 post-induction of endometriosis, by day 60 the expression was reduced. As compared to the control endometrium, the mRNA levels of Esr1 progressively reduced while the levels of inflammation associated genes (Esr2, Ifng, Tnf and Il1b) increased in the ectopic lesions. Infiltration of macrophages and polymorphonuclear leucocytes was also observed in the ectopic lesions indicative of inflammation. As compared to control, there was no change in levels of Cytokeratin and E-cadherin in the epithelial cells of ectopic endometrium. We did not observe excessive collagen deposition or α -SMA positive myofibroblasts in the stroma of the ectopic endometrium. Thus, epithelial-to-mesenchymal transition and fibrosis are not detected in the mouse model of endometriosis. Our results show that the mouse model of endometriosis mimics some but not all the features of human endometriosis.


Assuntos
Coristoma/genética , Endometriose/genética , Endométrio/metabolismo , Células Epiteliais/metabolismo , Células Estromais/metabolismo , Animais , Aromatase/genética , Aromatase/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proliferação de Células , Coristoma/metabolismo , Coristoma/patologia , Modelos Animais de Doenças , Endometriose/metabolismo , Endometriose/patologia , Endométrio/patologia , Endométrio/cirurgia , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Fibrose , Regulação da Expressão Gênica , Humanos , Inflamação , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Queratinas/genética , Queratinas/metabolismo , Mesentério/cirurgia , Camundongos , Miofibroblastos , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Células Estromais/patologia , Transplante Autólogo , Transplante Heterotópico , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Int J Dev Biol ; 64(1-2-3): 227-237, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32659011

RESUMO

The eutherian species evolved an elaborate uterus to allow viviparity. For successful pregnancy, the uterus must not only be differentiated, but must also function optimally and any defects in uterus differentiation and/or function can lead to infertility. The homoebox gene HOXA10 has emerged to be a key player in both uterine development and its optimal functioning in adulthood. Within the Abd-B family, the posterior Hoxa genes play a dominant role in anterio-posterior segmentation of the Müllerian ducts in mammals, with Hoxa10 having a central role in uterine segmentation. In the adult endometrium, HOXA10 is expressed by endometrial cells and is regulated in a cyclic manner under the influence of ovarian steroids. During embryo implantation, expression of HOXA10 is increased in endometrial stromal cells by signals from the embryo to govern stromal cell transformation to decidual cells. Once decidualization is initiated, HOXA10 is rapidly downregulated to activate expression of pro-invasive factors to promote trophoblast invasion. We propose that HOXA10 governs embryo implantation in a three-step process: 1) acquisition of endometrial receptivity, 2) responding to signals from the blastocyst to modify receptive endometrium for decidualization 3) making the decidua conductive for trophoblast invasion and placentation. There is currently ample evidence that expression of HOXA10 is deregulated in a variety of "endometriopathies" such as endometriosis and endometrial cancers. Overall, HOXA10 appears to be the master regulator of endometrial health and a central determinant of fertility in mammals.


Assuntos
Decídua/citologia , Implantação do Embrião , Endométrio/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Homeobox A10/metabolismo , Animais , Diferenciação Celular , Decídua/metabolismo , Endométrio/metabolismo , Feminino , Proteínas Homeobox A10/genética , Humanos , Placentação , Gravidez
18.
Peptides ; 132: 170367, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32645381

RESUMO

FSH-FSHR interaction is critical for folliculogenesis, spermatogenesis and progression of several cancers. Therefore, FSHR is an attractive target for fertility regulation and cancer therapeutics. Based on homology and structural analysis of hFSH-FSHR(ECD) complex, a minimal continuous stretch within FSHß seat-belt loop (FSHß (89-97)) was identified to be crucial for FSHR interaction. The ability of FSHß (89-97) peptide to neutralize FSHR activity was evaluated by a panel of in vitro and in vivo experiments. The synthetic peptide significantly inhibited binding of [125I]-FSH to rat Fshr as well as FSH-induced cAMP production. In immature rats, FSHß (89-97) peptide administration reduced FSH-mediated increase in ovarian weight. The peptide inhibited transition of follicles from pre-antral to antral stage and hindered the cell cycle progression of granulosa cells beyond G0/G1 phase. In adult rats, administration of the peptide inhibited estradiol synthesis and significantly perturbed folliculogenesis.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/metabolismo , Células da Granulosa/efeitos dos fármacos , Oligopeptídeos/farmacologia , Folículo Ovariano/efeitos dos fármacos , Ovário/metabolismo , Receptores do FSH/antagonistas & inibidores , Animais , Cristalografia por Raios X/métodos , Estradiol/biossíntese , Feminino , Células da Granulosa/metabolismo , Oligopeptídeos/química , Folículo Ovariano/metabolismo , Ovário/efeitos dos fármacos , Elementos Estruturais de Proteínas , Ratos , Ratos Sprague-Dawley , Receptores do FSH/metabolismo , Relação Estrutura-Atividade
19.
J Assist Reprod Genet ; 37(5): 1069-1081, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32152908

RESUMO

PURPOSE: Endometriosis is recognized as a steroid hormone-dependent disorder. However, controversies exist regarding the status of the steroid hormone receptor expression in endometriotic tissues. The purpose of this study was to determine the ontogeny of cellular changes in the expression of estrogen receptors (ERα, ERß), G protein-coupled estrogen receptor 1 (GPER1), and progesterone receptors (PRs) in endometriosis using a mouse model. METHODS: We used the autologous uterine tissue transfer mouse model and studied the mRNA and protein expression of ERα, ERß, GPER1, and PR in ectopic lesions at 2, 4, and 8 weeks of induction of endometriosis. RESULT: As compared to endometrium of controls, in the ectopic endometrium, ERα is reduced while ERß was elevated in stromal cells; however, Gper1 and PR levels are reduced in both stromal and epithelial cells in a time-specific manner. There is a high inter-animal variation in the levels of these receptors in ectopic endometrium as compared to controls; the levels also varied by almost 100-fold within the same lesion resulting in "micro-heterogeneity." The expression of all these receptors also deferred between two lesions from the same animal. CONCLUSION: In the endometriotic tissue, there is extensive inter-animal and intra-lesion heterogeneity in the expression of ERα, ERß, GPER1, and PR. These changes are not due to the influence of the peritoneal environment but appear to be tissue intrinsic. We propose that the variable outcomes in hormonal therapy for endometriosis could be possibly due to heterogeneity in the expression of steroid hormone receptors in the ectopic endometrium.


Assuntos
Endometriose/genética , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Progesterona/genética , Animais , Modelos Animais de Doenças , Endometriose/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Hormônios Esteroides Gonadais/genética , Humanos , Camundongos
20.
Transpl Infect Dis ; 22(2): e13193, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31605666

RESUMO

BACKGROUND: Crusted scabies (CS) is a rare, severe and highly contagious form of scabies, which has been reported in immunosuppressed patients. A high index of suspicion and awareness of CS is essential to treat this infestation. CASE: A 13-year-old boy presented with pruritic hyperkeratotic squamous plaques located on both inner wrists, the web spaces of both his hands and his feet, and the genital area of 12 months duration. He was diagnosed with focal segmental glomerulosclerosis at the age of 5 and received a kidney transplant at the age of 9. He has been on a maintenance dose of prednisone (5 mg/d) and mycophenolate mofetil (250 mg/d) for the past 2 years. He had a contact history with a school friend with similar lesions. A skin punch biopsy demonstrated the presence of multiple mites in the stratum corneum confirming the diagnosis of CS. Ivermectin, the recommended drug of choice for crusted scabies, is not available in South Africa. The patient was commenced on topical benzoyl benzoate lotion but discontinued its use because of intolerable irritation. We subsequently prescribed the daily application of topical 5% sulfur in petrolatum to which his pruritus subsided significantly after 2 weeks with resolution of all skin lesions at the end of 8 weeks. CONCLUSION: This case is the first documented report of CS in a pediatric renal transplant patient. Our management highlights that classic formularies of magistral drugs are still effective treatment options and can be used especially when standard therapies cannot be tolerated or when optimum treatments are not available.


Assuntos
Hospedeiro Imunocomprometido , Escabiose/diagnóstico , Transplantados , Adolescente , Benzoatos/efeitos adversos , Benzoatos/uso terapêutico , Biópsia , Humanos , Transplante de Rim , Masculino , Prednisona/efeitos adversos , Prednisona/uso terapêutico , Prurido/diagnóstico , Prurido/etiologia , Escabiose/tratamento farmacológico , Pele/patologia , Enxofre/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA