Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Adv Sci (Weinh) ; : e2402393, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39229959

ABSTRACT

Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an ß2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.

2.
Exp Dermatol ; 33(5): e15091, 2024 May.
Article in English | MEDLINE | ID: mdl-38711220

ABSTRACT

KIT ligand and its associated receptor KIT serve as a master regulatory system for both melanocytes and mast cells controlling survival, migration, proliferation and activation. Blockade of this pathway results in cell depletion, while overactivation leads to mastocytosis or melanoma. Expression defects are associated with pigmentary and mast cell disorders. KIT ligand regulation is complex but efficient targeting of this system would be of significant benefit to those suffering from melanocytic or mast cell disorders. Herein, we review the known associations of this pathway with cutaneous diseases and the regulators of this system both in skin and in the more well-studied germ cell system. Exogenous agents modulating this pathway will also be presented. Ultimately, we will review potential therapeutic opportunities to help our patients with melanocytic and mast cell disease processes potentially including vitiligo, hair greying, melasma, urticaria, mastocytosis and melanoma.


Subject(s)
Mast Cells , Mastocytosis , Melanocytes , Proto-Oncogene Proteins c-kit , Stem Cell Factor , Humans , Stem Cell Factor/metabolism , Melanocytes/metabolism , Mast Cells/metabolism , Mastocytosis/drug therapy , Mastocytosis/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Melanoma/metabolism , Melanoma/drug therapy , Vitiligo/metabolism , Vitiligo/drug therapy , Vitiligo/therapy , Pigmentation Disorders/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/drug therapy , Animals
3.
Anat Histol Embryol ; 52(6): 1010-1015, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37694739

ABSTRACT

In mammals, activation of primordial follicles to primary follicle is a progressive and highly regulated process. There is evidence in mice that phosphatase and tensin homologue deleted on Chromosome 10 (PTEN) silencing is an important negative regulator of phosphatidylinositol 3-kinase (PI3K), which initiates activation of dormant follicles. The objective of the study was to evaluate the effect of the addition of PTEN inhibitor (bpV(HOpic)) (10 µM) and/or Kit Ligand (KL) (100 ng/mL) on the in vitro activation and survival of alpaca primordial follicles. Ovarian cortical fragments from 11 adult alpacas were cultured for 24 h in tissue culture medium (α-MEM+ ) supplemented with KL and bpV or the association of both. Subsequently, each sample was processed by classical histology and follicular counting and classification were performed. The results obtained show a reduction (p < 0.05) of primordial follicles in more than 50% in follicular tissue cultured in vitro in α-MEM+ or supplemented with bpV and/or KL versus the control (not cultured). Further, >25% increase in primary follicles in follicular tissue cultured in vitro in α-MEM+ or supplemented with KL and/or bpV versus control. However, the follicular survival rate showed a decrease of 20% in the cultured tissues, except for the α-MEM+ supplemented with KL and bpV. In conclusion, supplementation of bpV (HOpic) (10 µM) and KL (100 ng/mL) increased the activation in vitro of primordial follicles and survival after in vitro culture of alpaca ovarian tissue.


Subject(s)
Camelids, New World , Female , Animals , Mice , Stem Cell Factor/pharmacology , Phosphatidylinositol 3-Kinases/pharmacology , Ovarian Follicle/physiology
4.
Cryobiology ; 96: 30-36, 2020 10.
Article in English | MEDLINE | ID: mdl-32871128

ABSTRACT

Different factors, such as basic fibroblast growth factor (bFGF) and kit ligand (KL), are used in ovarian cortical culture to promote activation of primordial follicles. In the present study, the effects of bFGF and KL, alone and in combination, were evaluated on human follicular activation and growth during in-situ cortical culture. Slow frozen-thawed human ovarian cortical tissues (n = 6) were cultured in 4 different groups: 1) control (base medium), 2) KL (base medium; BM + 100 ng/ml KL), 3) bFGF (BM + 100 ng/ml bFGF) and 4) bFGF + KL (BM + 100 ng/ml KL + 100 ng/ml bFGF) for a week. The proportion of morphologically normal and degenerated follicles at different developmental stages, secreted hormonal levels and specific gene expressions were compared. Although the proportion of growing follicles was higher than primordial counterpart in all cultured groups, no significant differences were observed among the cultured groups. In all cultured groups, anti-Müllerian hormone (AMH), progesterone and estradiol hormones levels increased after 7 days of culture; however, this increase was only significant for estradiol in the bFGF + KL group. The expression of Ki67 gene indicated an increase in ovarian cell proliferation in the three experimental groups compared to the control group, however this increment was only significant for the bFGF + KL group. It can be concluded that KL and bFGF factors individually have no beneficial effects on in-situ follicular growth, but their combination positively influences steroidogenesis of granulosa cells without significantly increasing the number of growing follicles.


Subject(s)
Fibroblast Growth Factor 2 , Stem Cell Factor , Cell Proliferation , Cryopreservation/methods , Female , Fibroblast Growth Factor 2/genetics , Granulosa Cells , Humans , Stem Cell Factor/genetics
5.
Immunology ; 159(4): 441-449, 2020 04.
Article in English | MEDLINE | ID: mdl-31957000

ABSTRACT

Signaling by Kit has been extensively studied in hematopoietic cells and is essential for the survival, proliferation and maintenance of hematopoietic stem and progenitor cells. In addition to the activation of intrinsic signaling pathways, Kit has been shown to interact with lineage-restricted type I cytokine receptors and produce cross signals, e.g. erythropoietin receptor, interleukin-7 receptor (IL-7R), IL-3R. Based on the earlier studies, we hypothesize that Kit activate other type I cytokine receptors in a cell-specific manner and execute cell-specific function. To investigate other Kit-activated receptors, we tested Kit and IL-4R cross-receptor activation in murine bone-marrow-derived mast cells, which express both Kit and IL-4R at the surface level. Kit upon activation by Kit ligand (KL), activated IL-4Rα, γC , and signal transducer and activator of transcription 6 independent of its cognate ligand IL-4. Though KL and IL-4 are individually mitogenic, combinations of KL and IL-4 synergistically promoted mast cell proliferation. Furthermore, inhibition of lipid raft formation by methyl-ß-cyclodextrin resulted in loss of synergistic proliferation. Together the data suggest IL-4R as a novel Kit-activated receptor. Such cross-receptor activations are likely to be a universal mechanism of Kit signaling in hematopoiesis.


Subject(s)
Interleukin-4/pharmacology , Mast Cells/drug effects , Proto-Oncogene Proteins c-kit/genetics , Receptors, Interleukin-4/genetics , STAT6 Transcription Factor/genetics , Stem Cell Factor/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Proliferation/drug effects , Gene Expression Regulation , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mast Cells/cytology , Mast Cells/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proto-Oncogene Proteins c-kit/immunology , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-4/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , STAT6 Transcription Factor/immunology , Signal Transduction , Stem Cell Factor/genetics , Stem Cell Factor/immunology , beta-Cyclodextrins/pharmacology
6.
G3 (Bethesda) ; 10(1): 311-319, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31757930

ABSTRACT

The body coloration of animals is due to pigment cells derived from neural crest cells, which are multipotent and differentiate into diverse cell types. Medaka (Oryzias latipes) possesses four distinct types of pigment cells known as melanophores, xanthophores, iridophores, and leucophores. The few melanophore (fm) mutant of medaka is characterized by reduced numbers of melanophores and leucophores. We here identify kit-ligand a (kitlga) as the gene whose mutation gives rise to the fm phenotype. This identification was confirmed by generation of kitlga knockout medaka and the findings that these fish also manifest reduced numbers of melanophores and leucophores and fail to rescue the fm mutant phenotype. We also found that expression of sox5, pax7a, pax3a, and mitfa genes is down-regulated in both fm and kitlga knockout medaka, implicating c-Kit signaling in regulation of the expression of these genes as well as the encoded transcription factors in pigment cell specification. Our results may provide insight into the pathogenesis of c-Kit-related pigmentation disorders such as piebaldism in humans, and our kitlga knockout medaka may prove useful as a tool for drug screening.


Subject(s)
Fish Proteins/genetics , Melanophores/metabolism , Oryzias/genetics , Skin Pigmentation/genetics , Animals , Fish Proteins/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
7.
Curr Med Sci ; 39(5): 702-706, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31612386

ABSTRACT

It has been reported that c-KIT ligand (KITLG) gene polymorphisms may be associated with testicular germ cell tumors (TGCT). Owing to mixed and inconclusive results, we conducted a systematic review and meta-analysis to summarize and clarify this association. A systematic search of studies on the association between KITLG gene polymorphisms and TGCT susceptibility was conducted in databases. Odds ratios and 95% confidence intervals were used to pool the effect size. Six articles were included in our systematic review and meta-analysis. Compared with adenine (A), KITLG rs995030 guanine (G) might be associated with increased risk of TGCT. There are insufficient data to fully confirm the association between KITLG rs4474514 and TGCT susceptibility. Well-designed studies with larger sample size and more subgroups are required to validate the risk identified in the current meta-analysis.


Subject(s)
Genetic Predisposition to Disease , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , Polymorphism, Single Nucleotide , Stem Cell Factor/genetics , Testicular Neoplasms/diagnosis , Testicular Neoplasms/genetics , Asian People , Gene Expression , Humans , Male , Neoplasms, Germ Cell and Embryonal/ethnology , Neoplasms, Germ Cell and Embryonal/pathology , Odds Ratio , Testicular Neoplasms/ethnology , Testicular Neoplasms/pathology , White People
8.
Theriogenology ; 123: 37-44, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30273739

ABSTRACT

The overall aim of this work was to study the influence of the hematopoietic growth factors erythropoietin (EPO) and kit ligand (KITL) during bovine oocyte in vitro maturation (IVM). The effect of adding different concentrations of EPO or KITL to maturation medium was evaluated analyzing oocyte nuclear maturation, cumulus cells apoptosis, embryo cleavage, reactive oxygen species (ROS) production in matured oocytes and cleaved embryos and the developmental competence to the blastocyst stage. No significant differences were observed in the percentage of oocytes that completed nuclear maturation among treatments, but the percentages of cleaved embryos and blastocysts obtained increased. With the addition of both hematopoietic growth factors the percentage of cumulus cells undergoing apoptosis decreased, the number of blastomeres per cleaved embryo was larger and ROS production per cleaved embryo increased. In conclusion, although the addition of EPO and KITL hematopoietic growth factors during bovine oocyte IVM had no impact on nuclear maturation, it had a positive effect on oocyte cytoplasmic maturation and developmental competence.


Subject(s)
Cattle/embryology , Embryo Culture Techniques/veterinary , Embryonic Development/drug effects , Erythropoietin/pharmacology , Stem Cell Factor/pharmacology , Animals , Fertilization in Vitro/veterinary , In Vitro Oocyte Maturation Techniques
9.
EMBO Rep ; 19(10)2018 10.
Article in English | MEDLINE | ID: mdl-30166337

ABSTRACT

Few studies report on the in vivo requirement for hematopoietic niche factors in the mammalian embryo. Here, we comprehensively analyze the requirement for Kit ligand (Kitl) in the yolk sac and aorta-gonad-mesonephros (AGM) niche. In-depth analysis of loss-of-function and transgenic reporter mouse models show that Kitl-deficient embryos harbor decreased numbers of yolk sac erythro-myeloid progenitor (EMP) cells, resulting from a proliferation defect following their initial emergence. This EMP defect causes a dramatic decrease in fetal liver erythroid cells prior to the onset of hematopoietic stem cell (HSC)-derived erythropoiesis, and a reduction in tissue-resident macrophages. Pre-HSCs in the AGM require Kitl for survival and maturation, but not proliferation. Although Kitl is expressed widely in all embryonic hematopoietic niches, conditional deletion in endothelial cells recapitulates germline loss-of-function phenotypes in AGM and yolk sac, with phenotypic HSCs but not EMPs remaining dependent on endothelial Kitl upon migration to the fetal liver. In conclusion, our data establish Kitl as a critical regulator in the in vivoAGM and yolk sac endothelial niche.


Subject(s)
Embryonic Development/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Stem Cell Factor/genetics , Animals , Aorta/growth & development , Cell Lineage/genetics , Cell Proliferation/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Erythropoiesis/genetics , Gene Expression Regulation, Developmental/genetics , Gonads/growth & development , Mesonephros/growth & development , Mice , Mice, Transgenic , Stem Cell Niche/genetics , Yolk Sac/growth & development
10.
Fish Physiol Biochem ; 44(6): 1599-1616, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30121735

ABSTRACT

The present study aimed to investigate whether the Gfra1/Gdnf and/or Kit/Kitlg regulatory pathways could be involved in the regulation of spermatogonial cell proliferation and/or differentiation in fish. Homologs of the mammalian gfra1, gdnf, kitr, and kitlg genes were identified in gnathostomes and reliable orthologous relationships were established using phylogenetic reconstructions and analyses of syntenic chromosomal fragments. Gene duplications and losses occurred specifically in teleost fish and members of the Salmoninae family including rainbow trout (Oncorhynchus mykiss) and Atlantic salmon (Salmo salar). Some duplicated genes exhibited distinct spatiotemporal expression profiles and were differently regulated by hormones in rainbow trout. Undifferentiated type A spermatogonia expressed higher levels of kitrb and kitra2 making them possible target cells for the gonadal kitlgb and somatic kitlga before the onset of spermatogenesis. Interestingly, gdnfa and gdnfb ohnologous genes were poorly expressed before the onset of spermatogenesis. The expression level of gdnfb was correlated with that of the vasa gene suggesting that the late increased abundance of gdnfb during spermatogenesis onset was due to the increased number of spermatogonial cells. gfra1a2 was expressed in undifferentiated type A spermatogonia whereas gfra1a1 was mainly detected in somatic cells. These observations indicate that the germinal gdnfb ligand could exert autocrine and paracrine functions on spermatogonia and on testicular somatic cells, respectively. Fsh and androgens inhibited gfra1a2 and gdnfb whereas gfra1a1 was stimulated by Fsh, androgens, and 17α, 20ß progesterone. Finally, our data provide evidences that the molecular identity of the male germ stem cells changes during ontogenesis prior to spermatogenesis onset.


Subject(s)
Evolution, Molecular , Fish Proteins/genetics , Gene Expression Regulation , Hormones/pharmacology , Oncorhynchus mykiss/genetics , Testis/metabolism , Transcriptome , Animals , Male , Oncorhynchus mykiss/physiology , Phylogeny , Signal Transduction , Spatio-Temporal Analysis , Spermatogenesis , Testis/growth & development
11.
Stem Cell Reports ; 10(6): 1920-1934, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29779898

ABSTRACT

Understanding the molecular pathways controlling hematopoietic stem cell specification and expansion is a necessary milestone to perform regenerative medicine. Here, we used the zebrafish model to study the role of the ckit signaling pathway in this process. We show the importance of kitb/kitlgb signaling in the specification and expansion of hematopoietic stem cells (HSCs), in the hemogenic endothelium and caudal hematopoietic tissue (CHT), respectively. Moreover, we identified the zebrafish ortholog of Oncostatin M (osm) in the zebrafish genome. We show that the osm/osmr pathway acts upstream of kitb during specification of the hemogenic endothelium, while both pathways act synergistically to expand HSCs in the CHT. Moreover, we found that osm, in addition to its role in promoting HSC proliferation, inhibits HSC commitment to the lymphoid fate. Altogether, our data identified two cytokines, kitlgb and osm, secreted by the vascular niche, that control HSCs during early embryonic development.


Subject(s)
Embryonic Development/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Oncostatin M/pharmacology , Stem Cell Factor/pharmacology , Zebrafish , Animals , Biomarkers , Gene Expression , Hematopoietic Stem Cells/drug effects , Immunohistochemistry , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Models, Biological , Oncostatin M Receptor beta Subunit/metabolism , Signal Transduction/drug effects
12.
Curr Osteoporos Rep ; 16(2): 95-104, 2018 04.
Article in English | MEDLINE | ID: mdl-29492879

ABSTRACT

PURPOSE OF REVIEW: Mesenchymal stem cells (MSCs) located in the bone marrow have the capacity to differentiate into multiple cell lineages, including osteoblast and adipocyte. Adipocyte density within marrow is inversely associated with bone mass during aging and in some pathological conditions, contributing to the prevailing view that marrow adipocytes play a largely negative role in bone metabolism. However, a negative association between marrow adipocytes and bone balance is not universal. Although MAT levels appear tightly regulated, establishing the precise physiological significance of MAT has proven elusive. Here, we review recent literature aimed at delineating the function of MAT. RECENT FINDINGS: An important physiological function of MAT may be to provide an expandable/contractible fat depot, which is critical for minimization of energy requirements for sustaining optimal hematopoiesis. Because the energy requirements for storing fat are negligible compared to those required to maintain hematopoiesis, even small reductions in hematopoietic tissue volume to match a reduced requirement for hematopoiesis could represent an important reduction in energy cost. Such a physiological function would require tight coupling between hematopoietic stem cells and MSCs to regulate the balance between MAT and hematopoiesis. Kit-ligand, an important regulator of proliferation, differentiation, and survival of hematopoietic cells, may function as a prototypic factor coupling MAT and hematopoiesis. Crosstalk between hematopoietic and mesenchymal cells in the bone marrow may contribute to establishing the balance between MAT levels and hematopoiesis.


Subject(s)
Adipose Tissue/physiology , Bone Marrow/physiology , Hematopoiesis/physiology , Adipocytes/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Aging/metabolism , Aging/physiology , Bone Marrow/metabolism , Bone Remodeling/physiology , Cell Differentiation , Cell Proliferation , Cell Survival , Hematopoietic Stem Cells , Humans , Mesenchymal Stem Cells , Osteoblasts , Stem Cell Factor/metabolism
13.
Dev Comp Immunol ; 84: 327-336, 2018 07.
Article in English | MEDLINE | ID: mdl-29522790

ABSTRACT

The development of mammalian megakaryocytes and platelets is regulated by numerous cytokine signals, primarily through the thrombopoietin (TPO)/c-MPL axis. Although non-mammalian vertebrates are known to possess nucleated thrombocytes functionally equivalent to mammalian platelets, the dynamics of the thrombocyte development remains unclear. Here we identified TPO and a splice variant (TPO-v) caused by the intron retention in common carp (Cyprinus carpio). Both the tpo and its variant transcripts were highly expressed in heart and liver. Recombinant carp TPO (rcTPO) was produced and purified in HEK293T cells stably expressing tpo, but TPO-v was shown not to be secreted from the transfectants. rcTPO induced the formation of colony-forming unit-thrombocyte (CFU-T) colonies which were recognized by a monoclonal antibody against carp thrombocytes expressing c-mpl and cd41, in a dose-dependent manner. The combination of rcTPO and recombinant carp Kit ligand A (rcKITLA) exerted a significant synergistic effect on three types of colony formation: thrombocytic colonies, thrombocytic burst colonies and thrombocytic/erythroid colonies. Utilizing this colony assay to examine the distribution of thrombocytic progenitor cells in carp, we demonstrated that carp head and trunk kidney play a primary role in thrombopoiesis, while the spleen does not. Our results indicate that carp possess mechanisms of TPO- and KITLA-dependent thrombopoiesis similar to those in other vertebrates and the sites of thrombopoiesis are restricted to the kidney, the primary hematopoietic organ in the teleost fish.


Subject(s)
Blood Platelets/physiology , Carps/physiology , Fish Proteins/metabolism , Heart/physiology , Kidney/physiology , Liver/physiology , Thrombopoietin/metabolism , Animals , Bodily Secretions , Fish Proteins/genetics , HEK293 Cells , Humans , Ligands , Mammals/physiology , Myeloid Progenitor Cells , RNA Isoforms/genetics , Receptors, Thrombopoietin/metabolism , Signal Transduction , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Thrombopoiesis , Thrombopoietin/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Int J Reprod Biomed ; 16(12)2018 Dec.
Article in English | MEDLINE | ID: mdl-31417983

ABSTRACT

BACKGROUND: Peritoneal fluid (PF) from infertile women with endometriosis contains inflammatory mediators that may interfere with folliculogenesis. OBJECTIVE: The aim was to evaluate the effects of curcumin on growth factors expression by evaluating Growth Differentiation Factor-9 (GDF-9), Kit Ligand (KitL), and Tumor Necrosis Factor α (TNF α ) expressions in bovine cumulus-oocyte complexes (COC)s cultured with PF from infertile women with endometriosis. MATERIALS AND METHODS: In this experimental study, 21 infertile women (aged between 20 and 40 years) who refered to Dr Sutomo Hospital from January to July 2015 were enrolled. COCs were aspirated from antral follicles of bovine ovaries. PF was collected from infertile women with endometriosis undergoing laparoscopy for infertility evaluation. Curcumin, a strong anti-inflammatory turmeric, was added in Tissue Culture Medium 199 (TCM199) and PF for culture medium. Bovine COCs were cultured into three groups of the medium: 1. TCM199, 2. TCM199 + PF, and 3. TCM199 + PF + curcumin. GDF-9, KitL, and TNF α expressions were examined using immunohistochemistry technique. RESULTS: GDF-9 expression of bovine COCs cultured in PF with curcumin addition (2.67 ± 0.98) was found to increase compared to those cultured without curcumin (0.50 ± 0.67) (p ≤ 0.001). It was similar to KitL expression of bovine COCs cultured with curcumin (2.67 ± 1.23), which increased compared to those without curcumin (0.33 ± 0.49) (p ≤ 0.001). A significant difference in TNF α expression was noted between groups with or without curcumin (p ≤ 0.001). CONCLUSION: In the culture of PF from infertile women with endometriosis, curcumin addition improves the growth factors expression of bovine COCs. The increase of GDF-9 and KitL expressions will improve folliculogenesis.

15.
Proc Natl Acad Sci U S A ; 114(38): E8035-E8044, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28878021

ABSTRACT

Casein kinase 1α (CK1α), a component of the ß-catenin destruction complex, is a critical regulator of Wnt signaling; its ablation induces both Wnt and p53 activation. To characterize the role of CK1α (encoded by Csnk1a1) in skin physiology, we crossed mice harboring floxed Csnk1a1 with mice expressing K14-Cre-ERT2 to generate mice in which tamoxifen induces the deletion of Csnk1a1 exclusively in keratinocytes [single-knockout (SKO) mice]. As expected, CK1α loss was accompanied by ß-catenin and p53 stabilization, with the preferential induction of p53 target genes, but phenotypically most striking was hyperpigmentation of the skin, importantly without tumorigenesis, for at least 9 mo after Csnk1a1 ablation. The number of epidermal melanocytes and eumelanin levels were dramatically increased in SKO mice. To clarify the putative role of p53 in epidermal hyperpigmentation, we established K14-Cre-ERT2 CK1α/p53 double-knockout (DKO) mice and found that coablation failed to induce epidermal hyperpigmentation, demonstrating that it was p53-dependent. Transcriptome analysis of the epidermis revealed p53-dependent up-regulation of Kit ligand (KitL). SKO mice treated with ACK2 (a Kit-neutralizing antibody) or imatinib (a Kit inhibitor) abrogated the CK1α ablation-induced hyperpigmentation, demonstrating that it requires the KitL/Kit pathway. Pro-opiomelanocortin (POMC), a precursor of α-melanocyte-stimulating hormone (α-MSH), was not activated in the CK1α ablation-induced hyperpigmentation, which is in contrast to the mechanism of p53-dependent UV tanning. Nevertheless, acute sunburn effects were successfully prevented in the hyperpigmented skin of SKO mice. CK1α inhibition induces skin-protective eumelanin but no carcinogenic pheomelanin and may therefore constitute an effective strategy for safely increasing eumelanin via UV-independent pathways, protecting against acute sunburn.


Subject(s)
Casein Kinase I/metabolism , Keratinocytes/metabolism , Skin Pigmentation , Sunburn/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/genetics , Epidermis/metabolism , Epidermis/pathology , Keratinocytes/pathology , Melanins/biosynthesis , Melanins/genetics , Melanocytes/metabolism , Melanocytes/pathology , Mice , Mice, Knockout , Sunburn/genetics , Sunburn/pathology , Tumor Suppressor Protein p53/genetics , beta Catenin/genetics , beta Catenin/metabolism
16.
J Cell Commun Signal ; 11(4): 297-307, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28656507

ABSTRACT

The stem cell factor (SCF) is a cytokine that specifically binds the tyrosine kinase receptor c-KIT. The SCF/c-KIT interaction leads to receptor dimerization, activation of kinase activity and initiation of several signal transduction pathways that control cell proliferation, apoptosis, differentiation and migration in several tissues. The activity of SCF/c-KIT system is linked with the phosphatidylinositol 3-kinase (PI3-K), the Src, the Janus kinase/signal transducers and activators of transcription (JAK/STAT), the phospholipase-C (PLC-γ) and the mitogen-activated protein kinase (MAPK) pathways. Moreover, it has been reported that cancer cases display an overactivation of c-KIT due to the presence of gain-of-function mutations or receptor overexpression, which renders c-KIT a tempting target for cancer treatment. In the case of male cancers the most documented activated pathways are the PI3-K and Src, both enhancing abnormal cell proliferation. It is also known that the Src activity in prostate cancer cases depends on the presence of tr-KIT, the cytoplasmic truncated variant of c-KIT that is specifically expressed in tumour tissues and, thus, a very interesting target for drug development. The present review provides an overview of the signalling pathways activated by SCF/c-KIT and discusses the potential application of c-KIT inhibitors for treatment of testicular and prostatic cancers.

17.
Proteins ; 85(7): 1362-1370, 2017 07.
Article in English | MEDLINE | ID: mdl-28370370

ABSTRACT

Kit ligand (KITL) plays important roles in cell proliferation, differentiation, and survival via interaction with its receptor Kit. The previous studies demonstrated that KITL formed a noncovalent homodimer through transmembrane (TM) domain; however, the undergoing mechanism of transmembrane association that determines KITL TM dimerization is still not clear. Herein, molecular dynamics (MD) simulation strategy and TOXCAT assay were combined to characterize the dimerization interface and structure of KITL TM in details. KITL TM formed a more energetically favorable noncovalent dimer through a conserved SxxxGxxxG motif in the MD simulation. Furthermore, the TOXCAT results demonstrated that KITL TM self-associated strongly in the bilayer membrane environment. Mutating any one of the small residues Ser11, Gly15 or Gly19 to Ile disrupted KITL TM dimerization dramatically, which further validated our MD simulation results. In addition, our results showed that Tyr22 could help to stabilize the TM interactions via interacting with the phosphoric group in the bilayer membrane. Pro7 did not induce helix kinks or swivel angles in KITL TM, but it was related with the pitch of the turn around this residue so as to affect the dimer formation. Combining the results of computer modeling and experimental mutagenesis studies on the KITL TM provide new insights for the transmembrane helix association of KITL dimerization. Proteins 2017; 85:1362-1370. © 2017 Wiley Periodicals, Inc.


Subject(s)
Chloramphenicol O-Acetyltransferase/metabolism , Molecular Dynamics Simulation , Recombinant Fusion Proteins/chemistry , Stem Cell Factor/chemistry , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cattle , Chloramphenicol O-Acetyltransferase/genetics , Cloning, Molecular , Dogs , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genes, Reporter , Humans , Kinetics , Lipid Bilayers/chemistry , Mice , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Multimerization , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Substrate Specificity , Swine , Thermodynamics
18.
Eur J Immunol ; 47(1): 206-212, 2017 01.
Article in English | MEDLINE | ID: mdl-27739072

ABSTRACT

Long-term proliferating, DH JH -rearranged mouse precursor B-cell lines have previously been established in serum- and IL-7-containing media from fetal liver, but not from bone marrow. Serum and stromal cells expose these pre-B cells to undefined factors, hampering accurate analyses of ligand-dependent signaling, which controls pre-B cell proliferation, survival, residence and migration. Here, we describe a novel serum-free, stromal cell-free culture system, which allows us to establish and maintain pre-B cells not only from fetal liver, but also from bone marrow with practically identical efficiencies in proliferation, cloning and differentiation. Surprisingly, recombinant kit-ligand, also called stem cell factor, produced as a kit-ligand-Fc fusion protein, suffices to replace stromal cells and serum, provided that it is presented to cultured pre-B cells in an optimal density in plate-bound, insolubilized, potentially crosslinking form. Additional recombinant CXCL12 and fibronectin have a minor influence on the establishment and maintenance of pre-B cell lines and clones from fetal liver, but are necessary to establish such cell lines from bone marrow.


Subject(s)
Interleukin-7/metabolism , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Stem Cell Factor/immunology , Animals , Cell Culture Techniques , Cell Line , Clone Cells , Culture Media , Interleukin-7/pharmacology , Mice , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/drug effects , Stem Cell Factor/pharmacology
19.
Theriogenology ; 86(1): 41-53, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27155734

ABSTRACT

This review focuses on the role of the dialog between the oocyte and its companion somatic cells in driving folliculogenesis from the primordial to the preovulatory follicle stage. Mouse and sheep genetic models have brought complementary evidence of these cell interactions and their consequences for ovarian function. In mouse, the deletion of genes encoding connexins has shown that functional gap junction channels between oocytes and granulosa cells and between granulosa cells themselves maintain the follicle in a functionally integrated state. Targeted deletions in oocytes or granulosa cells have revealed the cell- and stage-specific role of ubiquist factors belonging to the phosphatidylinositol 3 kinase signaling pathway in primordial follicle activation, oocyte growth and follicle survival. Various models of transgenic mice and sheep carrying natural loss-of-function mutations associated with sterility have established that the oocyte-derived factors, bone morphogenetic protein (BMP) 15 and growth differentiation factor 9 orchestrate follicle development, support cumulus metabolism and maturation and participate in oocyte meiosis arrest. Unexpectedly in sheep, mutations resulting in the attenuation of BMP signaling lead to enhanced ovulation rate, likely resulting from a lowered follicular atresia rate and the enhancement of FSH-regulated follicular maturation. Both the activation level of BMP signaling and an adequate equilibrium between BMP15 and growth differentiation factor 9 determine follicle survival, maturation, and development toward ovulation. The physiological approaches which were implemented on genetic animal models during the last 20 years have opened up new perspectives for female fertility by identifying the main signaling pathways of the oocyte-somatic cell dialog.


Subject(s)
Models, Biological , Oocytes/physiology , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Animals , Female
20.
Mol Cell Endocrinol ; 430: 33-48, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27045358

ABSTRACT

The early reproductive events starting with folliculogenesis and ending with blastocyst implantation into the uterine endometrium are regulated by a complex interplay among endocrine, paracrine and autocrine factors. This review examines the spatiotemporal integration of these maternal and embryonic signals that are required for successful reproduction. In coordination with hypothalamic-pituitary-gonadal (HPG) hormones, an intraovarian HPG-like axis regulates folliculogenesis, follicular quiescence, ovulation, follicular atresia, and corpus luteal functions. Upon conception and passage of the zygote through the fallopian tube, the contribution of maternal hormones in the form of paracrine secretions from the endosalpinx to embryonic development declines, with autocrine and paracrine signaling becoming increasingly important as instructional signals for the differentiation of the early zygote/morula into a blastocyst. These maternal and embryonic signals include activin and gonadotropin-releasing hormone 1 (GnRH1) that are crucial for the synthesis and secretion of the 'pregnancy' hormone human chorionic gonadotropin (hCG). hCG in turn signals pre-implantation embryonic cell division and sex steroid production required for stem cell differentiation, and subsequent blastulation, gastrulation, cavitation and blastocyst formation. Upon reaching the uterus, blastocyst hatching occurs under the influence of decreased activin signaling, while the attachment and invasion of the trophoblast into the endometrium appears to be driven by a decrease in activin signaling, and by increased GnRH1 and hCG signaling that allows for tissue remodeling and the controlled invasion of the blastocyst into the uterine endometrium. This review demonstrates the importance of integrative endocrine, paracrine, and autocrine signaling for successful human reproduction.


Subject(s)
Embryo Implantation , Embryonic Development , Hormones/metabolism , Organogenesis , Ovarian Follicle/embryology , Female , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL