Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Part Fibre Toxicol ; 18(1): 33, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34479598

ABSTRACT

BACKGROUND: Metal oxide nanoparticles (NPs) are increasingly used in many industrial and biomedical applications, hence their impact on occupational and public health has become a concern. In recent years, interest on the effect that exposure to NPs may exert on human reproduction has grown, however data are still scant. In the present work, we investigated whether different metal oxide NPs interfere with mouse cumulus cell-oocyte complex (COC) expansion. METHODS: Mouse COCs from pre-ovulatory follicles were cultured in vitro in the presence of various concentrations of two types of TiO2 NPs (JRC NM-103 and NM-104) and four types of ZnO NPs (JRC NM-110, NM-111, and in-house prepared uncoated and SiO2-coated NPs) and the organization of a muco-elastic extracellular matrix by cumulus cells during the process named cumulus expansion was investigated. RESULTS: We show that COC expansion was not affected by the presence of both types of TiO2 NPs at all tested doses, while ZnO NM-110 and NM-111 induced strong toxicity and inhibited COCs expansion at relatively low concentration. Medium conditioned by these NPs showed lower toxicity, suggesting that, beside ion release, inhibition of COC expansion also depends on NPs per se. To further elucidate this, we compared COC expansion in the presence of uncoated or SiO2-coated NPs. Differently from the uncoated NPs, SiO2-coated NPs underwent slower dissolution, were not internalized by the cells, and showed an overall lower toxicity. Gene expression analysis demonstrated that ZnO NPs, but not SiO2-coated ZnO NPs, affected the expression of genes fundamental for COC expansion. Dosimetry analysis revealed that the delivered-to-cell mass fractions for both NPs was very low. CONCLUSIONS: Altogether, these results suggest that chemical composition, dissolution, and cell internalization are all responsible for the adverse effects of the tested NPs and support the importance of a tailored, safer-by-design production of NPs to reduce toxicity.


Subject(s)
Metal Nanoparticles , Zinc Oxide , Animals , Cumulus Cells , Female , Metal Nanoparticles/toxicity , Mice , Oocytes , Silicon Dioxide/toxicity , Zinc Oxide/toxicity
2.
Int J Mol Sci ; 21(7)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218212

ABSTRACT

In the mammalian ovary, the hyaluronan (HA)-rich cumulus extracellular matrix (ECM) organized during the gonadotropin-induced process of oocyte maturation is essential for ovulation of the oocyte-cumulus complex (OCC) and fertilization. Versican is an HA-binding proteoglycan that regulates cell function and ECM assembly. Versican cleavage and function remain to be determined in ovarian follicle. We investigated versican expression in porcine ovarian follicles by real-time (RT)-PCR and western blotting. The aims of the present work were to determine whether 1) versican was produced and cleaved by porcine OCCs during gonadotropin stimulation; 2) these processes were autonomous or required the participation of mural granulosa cells (MGCs); and 3) versican cleavage was involved in the formation or degradation of expanded cumulus ECM. We demonstrate two cleavage products of G1 domain of versican (V1) accumulated in the HA-rich cumulus ECM. One of them, a G1-DPEAAE N-terminal fragment (VG1) of ~70 kDa, was generated from V1 during organization of HA in in vivo and in vitro expanded porcine OCCs. Second, the V1-cleaved DPEAAE-positive form of ~65 kDa was the only species detected in MGCs. No versican cleavage products were detected in OCCs cultured without follicular fluid. In summary, porcine OCCs are autonomous in producing and cleaving V1; the cleaved fragment of ~70 kDa VG1 is specific for formation of the expanded cumulus HA-rich ECM.


Subject(s)
Oocytes/metabolism , Versicans/metabolism , Animals , Cell Differentiation , Cells, Cultured , Epitopes/immunology , Female , Oocytes/cytology , Oocytes/immunology , Swine , Versicans/genetics
3.
Front Immunol ; 11: 622772, 2020.
Article in English | MEDLINE | ID: mdl-33584725

ABSTRACT

The Long Pentraxin 3 (PTX3) is a multifunctional glycoprotein released by peripheral blood leukocytes and myeloid dendritic cells in response to primary pro-inflammatory stimuli, that acts as a non-redundant component of the humoral arm of innate immunity. In addition to the primary role in the acute inflammatory response, PTX3 seems to be involved in other physiological and pathological processes. Indeed, PTX3 seems to play a pivotal role in the deposition and remodeling of bone matrix during the mineralization process, promoting osteoblasts differentiation and activity. Recently, PTX3 was seen to be involved in the ectopic calcifications' formation in breast cancer disease. In this regard, it has been observed that breast cancer tumors characterized by high expression of PTX3 and high amount of Breast Osteoblast Like Cells (BOLCs) showed several Hydroxyapatite (HA) microcalcifications, suggesting a likely role for PTX3 in differentiation and osteoblastic activity in both bone and extra-bone sites. Furthermore, given its involvement in bone metabolism, several studies agree with the definition of PTX3 as a molecule significantly involved in the pathogenesis of age-related bone diseases, such as osteoporosis, both in mice and humans. Recent results suggest that genetic and epigenetic mechanisms acting on PTX3 gene are also involved in the progression of these diseases. Based on these evidences, the aim of our systemic review was to offer an overview of the variety of biological processes in which PTX3 is involved, focusing on bone mineralization, both in a physiological and pathological context.


Subject(s)
Aging/immunology , C-Reactive Protein/immunology , Calcification, Physiologic/immunology , Nerve Tissue Proteins/immunology , Osteoporosis/immunology , Serum Amyloid P-Component/immunology , Aging/pathology , Animals , Humans , Mice , Osteoporosis/pathology
4.
Matrix Biol ; 78-79: 11-23, 2019 05.
Article in English | MEDLINE | ID: mdl-29408277

ABSTRACT

Successful ovulation and oocyte fertilization are essential prerequisites for the beginning of life in sexually reproducing animals. In mammalian fertilization, the relevance of the protein coat surrounding the oocyte plasma membrane, known as zona pellucida, has been widely recognized, while, until not too long ago, the general belief was that the cumulus oophorus, consisting of follicle cells embedded in a hyaluronan rich extracellular matrix, was not essential. This opinion was based on in vitro fertilization procedures, in which a large number of sperms are normally utilized and the oocyte can be fertilized even if depleted of cumulus cells. Conversely, in vivo, only very few sperm cells reach the fertilization site, arguing against the possibility of a coincidental encounter with the oocyte. In the last two decades, proteins required for HA organization in the cumulus extracellular matrix have been identified and the study of fertility in mice deprived of the corresponding genes have provided compelling evidence that this jelly-like coat is critical for fertilization. This review focuses on the advances in understanding the molecular interactions making the cumulus environment suitable for oocyte and sperm encounter. Most of the studies on the molecular characterization of the cumulus extracellular matrix have been performed in the mouse and we will refer essentially to findings obtained in this animal model.


Subject(s)
Cumulus Cells/metabolism , Fertilization , Hyaluronic Acid/metabolism , Oocytes/metabolism , Animals , Extracellular Matrix/metabolism , Female , Humans , Male , Mammals , Mice , Oocytes/chemistry , Signal Transduction , Spermatozoa/physiology
5.
Cell Death Differ ; 26(4): 779, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30127514

ABSTRACT

Following publication of their article, the authors reported that the name of the fifth author had been formatted incorrectly in PubMed. Instead of "Rella FD" it should be "Di Rella F".

6.
Front Immunol ; 9: 2808, 2018.
Article in English | MEDLINE | ID: mdl-30555480

ABSTRACT

Follicular development is a highly coordinated process that in humans takes more than 6 months. Pituitary gonadotropins and a variety of locally produced growth factors and cytokines are involved in determining a precise sequence of changes in cell metabolism, proliferation, vascularization, and matrix remodeling in order to obtain a follicle with full ovulatory and steroidogenic capability. A low-grade inflammation can alter such processes leading to premature arrest of follicular growth and female reproductive failure. On the other hand, factors that are involved in inflammatory response as well as in innate immunity are physiologically upregulated in the follicle at the final stage of maturation and play an essential role in ovulation and fertilization. The generation of pentraxin 3 (PTX3) deficient mice provided the first evidence that this humoral pattern recognition molecule of the innate immunity has a non-redundant role in female fertility. The expression, localization, and molecular interactions of PTX3 in the periovulatory follicle have been extensively studied in the last 10 years. In this review, we summarize findings demonstrating that PTX3 is synthesized before ovulation by cells surrounding the oocyte and actively participates in the organization of the hyaluronan-rich provisional matrix required for successful fertilization. Data in humans tend to confirm these findings, indicating PTX3 as a biomarker of oocyte quality. Moreover, we discuss the emerging evidence that in humans altered PTX3 systemic levels, determined by genetic variations and/or low-grade chronic inflammation, can also impact the growth and development of the follicle and affect the incidence of ovarian disorders.


Subject(s)
C-Reactive Protein/immunology , Fertility/immunology , Immunity, Innate , Oocytes/immunology , Ovarian Diseases/immunology , Ovarian Follicle/immunology , Serum Amyloid P-Component/immunology , Animals , Extracellular Matrix/immunology , Female , Humans
7.
Minerva Ginecol ; 70(5): 549-560, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29999289

ABSTRACT

During a woman's reproductive life, only about 400 primordial follicles will develop into a preovulatory follicle and undergo ovulation, releasing an oocyte available for fertilization. The process of formation and selection of these follicles is complex and involves a multistep process characterized by a balance between survival and death of the oocytes and the surrounding follicular cells. Although the mechanisms underlying such process are not completely clarified yet, it is common idea that they can occur through various types of programmed cellular death (PCD). Since atresia is the principal destiny of the ovarian follicles, it is relevant to understand how this process takes place and how it is regulated. In this review, after a summary description of folliculogenesis in humans, the main mechanisms of atresia reported to occur during folliculogenesis from birth to adult age, in the human ovary and in other mammals when appropriate, are described.


Subject(s)
Oocytes/cytology , Ovarian Follicle/physiology , Ovary/physiology , Adult , Animals , Apoptosis/physiology , Cell Death/physiology , Female , Follicular Atresia/physiology , Humans , Ovulation/physiology
8.
J Endocrinol ; 236(1): 1-12, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28993437

ABSTRACT

A clinical association between thyroid dysfunction and pregnancy complications has been extensively reported; however, the molecular mechanisms through which TH might regulate key events of pregnancy have not been elucidated yet. In this respect, we performed in vivo studies in MMI-induced hypothyroid pregnant mice, evaluating the effect of hypothyroidism on the number of implantation sites, developing embryos/resorptions and pups per litter, at 4.5, 10.5, 18.5 days post-coitum (dpc) and at birth. We also studied the expression of major molecules involved in implantation and placentation, such as the proteases ISPs, MMPs, TIMPs and Notch pathway-related genes. Our results demonstrate that hypothyroidism may have a dual effect on pregnancy, by initially influencing implantation and by regulating placental development at later stages of gestation. To further elucidate the role of TH in implantation, we performed in vitro studies by culturing 3.5 dpc blastocysts in the presence of TH, with or without endometrial cells used as the feeder layer, and studied their ability to undergo hatching and outgrowth. We observed that, in the presence of endometrial feeder cells, TH is able to anticipate blastocyst hatching by upregulating the expression of blastocyst-produced ISPs, and to enhance blastocyst outgrowth by upregulating endometrial ISPs and MMPs. These results clearly indicate that TH is involved in the bidirectional crosstalk between the competent blastocyst and the receptive endometrium at the time of implantation.


Subject(s)
Embryo Implantation/genetics , Embryonic Development/genetics , Hypothyroidism/genetics , Peptide Hydrolases/genetics , Receptors, Notch/genetics , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/metabolism , Cells, Cultured , Coculture Techniques , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Enzyme Activation , Female , Gene Expression Regulation, Developmental , Hypothyroidism/chemically induced , Hypothyroidism/metabolism , Male , Methimazole , Mice , Peptide Hydrolases/metabolism , Pregnancy , Receptors, Notch/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/genetics , Thyroid Hormones/blood , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology
9.
Cell Death Dis ; 8(10): e3125, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29022895

ABSTRACT

Pentraxin 3 (PTX3) is a multifunctional glycoprotein regulating inflammatory response, cell proliferation and migration and deposition and remodelling of the extracellular matrix by a variety of cells. In this study, we investigated the possible role of PTX3 in bone homeostasis. To this end, we compared the expression and function of PTX3 in human osteoblasts of osteoporotic, osteoarthritic patients and young subjects not affected by bone diseases. Immunohistochemical analysis performed on bone head biopsies showed a close association between bone health and the number of osteoblasts expressing PTX3. Noteworthy, the proportion of PTX3-positive osteoblasts resulted to be significantly lower in osteoporotic patients compared with both young patients and osteoarthritic patients of the same age. Ex vivo culture of osteoblasts isolated from the three groups of patients confirmed in vivo observation. Specifically, we observed rare runt-related transcription factor 2 (RUNX2) immunopositive osteoblasts expressing PTX3 in cell cultures derived from osteoporotic patients and western blotting analysis showed 80% reduction of PTX3 in the corresponding culture extracts compared with young and osteoarthritic patients. The treatment of human osteoblast primary cultures derived from young patients with anti-PTX3 antibody dramatically affected osteoblast behaviour. Indeed, they lost the morphological and molecular features typical of mature osteoblasts, acquiring fibroblast-like shape and highly decreasing nuclear factor kappa-B ligand (RANKL) and RUNX2 expression. Also, the inhibition of PTX3 negatively affected osteoblast proliferation and their ability to form cell clusters and microhydroxyapatite crystals. Altogether, these results suggest a central role of PTX3 in bone homeostasis showing its involvement in osteoblast proliferation, differentiation and function.


Subject(s)
Bone Density/physiology , C-Reactive Protein/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Osteoporosis/pathology , Serum Amyloid P-Component/metabolism , Aged , Bone and Bones/metabolism , C-Reactive Protein/biosynthesis , C-Reactive Protein/immunology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , Osteoarthritis/pathology , RANK Ligand/metabolism , Serum Amyloid P-Component/biosynthesis , Serum Amyloid P-Component/immunology
10.
Cell Death Differ ; 24(1): 72-82, 2017 01.
Article in English | MEDLINE | ID: mdl-27689876

ABSTRACT

Premature ovarian failure and female infertility are frequent side effects of anticancer therapies, owing to the extreme sensitivity of the ovarian reserve oocytes to the damaging effects of irradiation and chemotherapy on DNA. We report here a robust protective effect of luteinizing hormone (LH) on the primordial follicle pool of prepubertal ovaries against the cisplatin (Cs)-induced apoptosis. In vitro LH treatment of prepubertal ovarian fragments generated anti-apoptotic signals by a subset of ovarian somatic cells expressing LH receptor (LHR) through cAMP/PKA and Akt pathways. Such signals, reducing the oocyte level of pro-apoptotic TAp63 protein and favoring the repair of the Cs-damaged DNA in the oocytes, prevented their apoptosis. Noteworthy, in vivo administration to prepubertal female mice of a single dose of LH together with Cs inhibited the depletion of the primordial follicle reserve caused by the drug and preserved their fertility in reproductive age, preventing significant alteration in the number of pregnancy and of delivered pups. In conclusion, these findings establish a novel ovoprotective role for LH and further support the very attracting prospective to use physiological 'fertoprotective' approaches for preventing premature infertility and risks linked to precocious menopause in young patients who survived cancer after chemotherapy.


Subject(s)
Apoptosis/drug effects , Cisplatin/toxicity , Luteinizing Hormone/pharmacology , Androstadienes/pharmacology , Animals , Cells, Cultured , Chromones/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Damage/drug effects , Female , Fertility/drug effects , Follicle Stimulating Hormone/pharmacology , Mice , Mice, Transgenic , Morpholines/pharmacology , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, LH/metabolism , Signal Transduction , Trans-Activators/metabolism , Wortmannin
11.
Biophys J ; 110(12): 2779-2789, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27332136

ABSTRACT

The cumulus cell-oocyte complex (COC) matrix is an extended coat that forms around the oocyte a few hours before ovulation and plays vital roles in oocyte biology. Here, we analyzed the micromechanical response of mouse COC matrix by colloidal-probe atomic force microscopy. We found that the COC matrix is elastic insofar as it does not flow and its original shape is restored after force release. At the same time, the COC matrix is extremely soft. Specifically, the most compliant parts of in vivo and in vitro expanded COC matrices yielded Young's modulus values of 0.5 ± 0.1 Pa and 1.6 ± 0.3 Pa, respectively, suggesting both high porosity and a large mesh size (≥100 nm). In addition, the elastic modulus increased progressively with indentation. Furthermore, using optical microscopy to correlate these mechanical properties with ultrastructure, we discovered that the COC is surrounded by a thick matrix shell that is essentially devoid of cumulus cells and is enhanced upon COC expansion in vivo. We propose that the pronounced nonlinear elastic behavior of the COC matrix is a consequence of structural heterogeneity and serves important functions in biological processes such as oocyte transport in the oviduct and sperm penetration.


Subject(s)
Elasticity/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Hyaluronic Acid/metabolism , Oocytes/metabolism , Oocytes/ultrastructure , Animals , Female , Mice , Microscopy, Atomic Force , Optical Imaging , Viscosity
12.
J Biol Chem ; 291(8): 3821-36, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26694612

ABSTRACT

Cumulus cells sustain the development and fertilization of the mammalian oocyte. These cells are retained around the oocyte by a hyaluronan-rich extracellular matrix synthesized before ovulation, a process called cumulus cell-oocyte complex (COC) expansion. Hyaluronan release and dispersion of the cumulus cells progressively occur after ovulation, paralleling the decline of oocyte fertilization. We show here that, in mice, postovulatory changes of matrix are temporally correlated to cumulus cell death. Cumulus cell apoptosis and matrix disassembly also occurred in ovulated COCs cultured in vitro. COCs expanded in vitro with FSH or EGF underwent the same changes, whereas those expanded with 8-bromo-adenosine-3',5'-cyclic monophosphate (8-Br-cAMP) maintained integrity for a longer time. It is noteworthy that 8-Br-cAMP treatment was also effective on ovulated COCs cultured in vitro, prolonging the vitality of the cumulus cells and the stability of the matrix from a few hours to >2 days. Stimulation of endogenous adenylate cyclase with forskolin or inhibition of phosphodiesterase with rolipram produced similar effects. The treatment with selective cAMP analogues suggests that the effects of cAMP elevation are exerted through an EPAC-independent, PKA type II-dependent signaling pathway, probably acting at the post-transcriptional level. Finally, overnight culture of ovulated COCs with 8-Br-cAMP significantly counteracted the decrease of fertilization rate, doubling the number of fertilized oocytes compared with control conditions. In conclusion, these studies suggest that cAMP-elevating agents prevent cumulus cell senescence and allow them to continue to exert beneficial effects on oocyte and sperm, thereby extending in vitro the time frame of oocyte fertilizability.


Subject(s)
Cumulus Cells/metabolism , Cyclic AMP/metabolism , Epidermal Growth Factor/pharmacology , Fertilization/drug effects , Follicle Stimulating Hormone/pharmacology , Hyaluronic Acid/metabolism , Oocytes/metabolism , Adenylyl Cyclases/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cumulus Cells/cytology , Extracellular Matrix/metabolism , Female , Mice , Oocytes/cytology , Rolipram/pharmacology , Signal Transduction/drug effects
13.
J Biol Chem ; 290(48): 28708-23, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26468290

ABSTRACT

The matrix polysaccharide hyaluronan (HA) has a critical role in the expansion of the cumulus cell-oocyte complex (COC), a process that is necessary for ovulation and fertilization in most mammals. Hyaluronan is organized into a cross-linked network by the cooperative action of three proteins, inter-α-inhibitor (IαI), pentraxin-3, and TNF-stimulated gene-6 (TSG-6), driving the expansion of the COC and providing the cumulus matrix with its required viscoelastic properties. Although it is known that matrix stabilization involves the TSG-6-mediated transfer of IαI heavy chains (HCs) onto hyaluronan (to form covalent HC·HA complexes that are cross-linked by pentraxin-3) and that this occurs via the formation of covalent HC·TSG-6 intermediates, the underlying molecular mechanisms are not well understood. Here, we have determined the tertiary structure of the CUB module from human TSG-6, identifying a calcium ion-binding site and chelating glutamic acid residue that mediate the formation of HC·TSG-6. This occurs via an initial metal ion-dependent, non-covalent, interaction between TSG-6 and HCs that also requires the presence of an HC-associated magnesium ion. In addition, we have found that the well characterized hyaluronan-binding site in the TSG-6 Link module is not used for recognition during transfer of HCs onto HA. Analysis of TSG-6 mutants (with impaired transferase and/or hyaluronan-binding functions) revealed that although the TSG-6-mediated formation of HC·HA complexes is essential for the expansion of mouse COCs in vitro, the hyaluronan-binding function of TSG-6 does not play a major role in the stabilization of the murine cumulus matrix.


Subject(s)
Cell Adhesion Molecules , Cumulus Cells/metabolism , Extracellular Matrix , Hyaluronic Acid , Oocytes/metabolism , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Mice
14.
Matrix Biol ; 30(5-6): 330-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21619930

ABSTRACT

Pentraxin 3 (PTX3) plays a key role in the formation of the hyaluronan-rich matrix of the cumulus oophorus surrounding ovulated eggs that is required for successful fertilization and female fertility. PTX3 is a multimeric protein consisting of eight identical protomers held together by a combination of non-covalent interactions and disulfide bonds. Recent findings suggest that the oligomeric status of PTX3 is important for stabilizing the cumulus matrix. Because the role of PTX3 in the cumulus resides in the unique N-terminal sequence of the protomer, we investigated further this issue by testing the ability of distinct Cys/Ser mutants of recombinant N-terminal region of PTX3 (N(_)PTX3) with different oligomeric arrangement to promote in vitro normal expansion in cumuli from Ptx3-null mice. Here we report that the dimer of the N(_)PTX3 is unable to rescue cumulus matrix organization, and that the tetrameric assembly of the protein is the minimal oligomeric state required for accomplishing this function. We have previously demonstrated that PTX3 binds to HCs of IαI and TSG-6, which are essential for cumulus matrix formation and able to interact with hyaluronan. Interestingly, here we show by solid-phase binding experiments that the dimer of the N(_)PTX3 retains the ability to bind to both IαI and TSG-6, suggesting that the octameric structure of PTX3 provides multiple binding sites for each of these ligands. These findings support the hypothesis that PTX3 contributes to cumulus matrix organization by cross-linking HA polymers through interactions with multiple HCs of IαI and/or TSG-6. The N-terminal PTX3 tetrameric oligomerization was recently reported to be also required for recognition and inhibition of FGF2. Given that this growth factor has been detected in the mammalian preovulatory follicle, we wondered whether FGF2 negatively influences cumulus expansion and PTX3 may also serve in vivo to antagonize its activity. We found that a molar excess of FGF2, above PTX3 binding capacity, does not affect in vitro cumulus matrix formation thus ruling out this possibility. In conclusion, the data strength the view that PTX3 acts as a nodal molecule in cross-linking HA in the matrix.


Subject(s)
C-Reactive Protein/metabolism , Cell Adhesion Molecules/metabolism , Cumulus Cells/metabolism , Oocytes/metabolism , Serum Amyloid P-Component/metabolism , Animals , Binding Sites , C-Reactive Protein/genetics , Chromatography, Gel , Cloning, Molecular , Cumulus Cells/cytology , Extracellular Matrix/metabolism , Female , Fibroblast Growth Factor 2/metabolism , HEK293 Cells , Humans , Hyaluronic Acid/metabolism , Mice , Oocytes/cytology , Protein Binding , Protein Conformation , Recombinant Proteins/metabolism , Serum Amyloid P-Component/genetics
15.
Mol Reprod Dev ; 78(6): 391-402, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21520325

ABSTRACT

Several lines of evidence suggest that in mice the activation of SMAD2/3 signaling by oocyte secreted factors, together with epidermal growth factor receptor (EGFR) activation, is essential to induce cumulus expansion. Here we show that inhibition of EGFR kinase in follicle stimulating hormone (FSH)-stimulated porcine oocyte-cumulus cell complex (OCCs) strongly decreases hyaluronan (HA) synthesis and its retention in the matrix, as well as progesterone synthesis. Although porcine cumulus cells undergo expansion independently of oocytes, we use biochemical and gene expression analyses to show that they do require activation of SMAD2/3 for optimal stimulation of HA synthesis and proteins involved in the organization of this polymer in the expanded matrix. Furthermore, FSH-induced progesterone synthesis by porcine cumulus cells was increased by blocking SMAD2/3 activation. In conclusion, these results support the hypothesis that an FSH-EGF autocrine loop is active in porcine OCCs, and provide the first evidence that the SMAD2/3 signaling pathway is induced by paracrine/autocrine factors in porcine cumulus cells and is involved in the control of both cumulus expansion and steroidogenesis.


Subject(s)
Cumulus Cells/metabolism , ErbB Receptors/metabolism , Hyaluronic Acid/biosynthesis , Isoquinolines/pharmacology , Oocytes/enzymology , Progesterone/biosynthesis , Pyridines/pharmacology , Pyrroles/pharmacology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Animals , Benzamides/pharmacology , C-Reactive Protein/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Dioxoles/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental/drug effects , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Meiosis/drug effects , Mice , Oocytes/physiology , Quinazolines/pharmacology , Serum Amyloid P-Component/metabolism , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad3 Protein/antagonists & inhibitors , Swine , Tyrphostins/pharmacology
16.
Hum Mol Genet ; 19(24): 4886-94, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20881015

ABSTRACT

Sam68 is a multifunctional RNA-binding protein highly expressed in the gonads, whose ablation causes male infertility. Herein, we have investigated Sam68 expression in the adult ovary and its function in female fertility. Immunohistochemistry showed that Sam68 was localized in the nucleus of oocytes and follicular cells at all stages of folliculogenesis. Sam68(-/-) females were severely subfertile, and they showed a delay in the age of first pregnancy, increased breeding time for successful pregnancy and yielded smaller litters. Morphological analyses indicated a significant reduction in the number of secondary and pre-antral follicles in the ovary. These defects were associated with alteration of oestrous cycles and a reduced number of ovulated oocytes, which were only partially restored by the administration of exogenous gonadotropins. Crosslinking/immunoprecipitation experiments showed that Sam68 directly binds the mRNAs for the follicle-stimulating hormone (FSH) and the luteinizing hormone receptors (Fshr and Lhcgr), which were downregulated in ovaries of adult knockout females. Stimulation of immature females with FSH-like pregnant mare serum gonadotropin (PMSG), or of follicular cells with the FSH second messenger analogue 8Br-cAMP, caused the upregulation of Sam68. The increase in Sam68 levels paralleled that of the Fshr and Lhcgr mRNAs in the pre-ovulatory follicle and was required to allow accumulation of these transcripts in follicular cells. These studies identify a new crucial function for Sam68 in the regulation of female fertility and indicate that this protein is required to insure proper expression of the gonadotropin receptor transcripts in pre-ovulatory follicles in adult ovary.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Deletion , Gonadotropins, Equine/pharmacology , Infertility, Female/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , RNA-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Estrous Cycle/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Infertility, Female/genetics , Male , Mice , Ovarian Follicle/pathology , Ovarian Follicle/physiopathology , Pregnancy , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism , Up-Regulation/drug effects
17.
Biol Reprod ; 78(5): 903-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18256330

ABSTRACT

We have previously shown that the heavy chains (HCs) of inter-alpha-trypsin inhibitor (IalphaI) become covalently linked to hyaluronan (HA) during in vivo and in vitro expansion of porcine oocyte-cumulus cell complexes (OCCs). We have now studied by immunoblotting the synthesis of tumor necrosis factor alpha-induced protein 6 (TNFAIP6), which is essential for catalyzing this reaction in expanding mouse OCCs. Expanding OCCs were collected from preovulatory follicles of naturally cycling pigs and also after in vitro culture (24 or 42 h) in medium supplemented with FSH and pig serum. After isolation, OCCs were treated with Streptomyces hyaluronidase or Chondroitinase ABC. Matrix, cell pellet, and total extracts were analyzed by Western blotting. A band of about 35 kDa and a doublet of about 120 kDa, corresponding to the molecular weight of the native and HC-linked forms of TNFAIP6, respectively, were detected by a rabbit anti-human TNFAIP6 polyclonal antibody in matrix extracts of expanded cumuli. Moreover, we found by using a cell-free assay that porcine follicular fluid collected from follicles at 24 h after hCG stimulation contains HC-HA coupling activity. This activity was abolished by the rat anti-human monoclonal antibody A38, which has an epitope within the Link module domain of TNFAIP6. These experiments suggest that free TNFAIP6 protein was present in follicular fluid aspirated from porcine follicles 24 h after hCG stimulation. In contrast to mouse, we show that the A38 monoclonal antibody does not affect in vitro cumulus expansion of porcine OCCs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Adhesion Molecules/metabolism , Follicular Phase/metabolism , Ovarian Follicle/metabolism , Alpha-Globulins/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/immunology , Cells, Cultured , Epitopes/metabolism , Female , Follicular Fluid/metabolism , Hyaluronic Acid/metabolism , Mice , Mice, Inbred Strains , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Swine , Time Factors
18.
J Biol Chem ; 283(15): 10147-61, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18223257

ABSTRACT

PTX3 is an acute phase glycoprotein that plays key roles in resistance to certain pathogens and in female fertility. PTX3 exerts its functions by interacting with a number of structurally unrelated molecules, a capacity that is likely to rely on its complex multimeric structure stabilized by interchain disulfide bonds. In this study, PAGE analyses performed under both native and denaturing conditions indicated that human recombinant PTX3 is mainly composed of covalently linked octamers. The network of disulfide bonds supporting this octameric assembly was resolved by mass spectrometry and Cys to Ser site-directed mutagenesis. Here we report that cysteine residues at positions 47, 49, and 103 in the N-terminal domain form three symmetric interchain disulfide bonds stabilizing four protein subunits in a tetrameric arrangement. Additional interchain disulfide bonds formed by the C-terminal domain cysteines Cys(317) and Cys(318) are responsible for linking the PTX3 tetramers into octamers. We also identified three intrachain disulfide bonds within the C-terminal domain that we used as structural constraints to build a new three-dimensional model for this domain. Previously it has been shown that PTX3 is a key component of the cumulus oophorus extracellular matrix, which forms around the oocyte prior to ovulation, because cumuli from PTX3(-/-) mice show defective matrix organization. Recombinant PTX3 is able to restore the normal phenotype ex vivo in cumuli from PTX3(-/-) mice. Here we demonstrate that PTX3 Cys to Ser mutants, mainly assembled into tetramers, exhibited wild type rescue activity, whereas a mutant, predominantly composed of dimers, had impaired functionality. These findings indicate that protein oligomerization is essential for PTX3 activity within the cumulus matrix and implicate PTX3 tetramers as the functional molecular units required for cumulus matrix organization and stabilization.


Subject(s)
C-Reactive Protein/chemistry , Disulfides/chemistry , Extracellular Matrix/chemistry , Serum Amyloid P-Component/chemistry , Amino Acid Substitution , Animals , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , CHO Cells , Cricetinae , Cricetulus , Cumulus Cells/metabolism , Disulfides/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Fertility/physiology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oocytes/chemistry , Oocytes/metabolism , Ovulation/physiology , Protein Structure, Quaternary/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism
19.
J Biol Chem ; 282(41): 30161-70, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17675295

ABSTRACT

Pentraxin 3 (PTX3) and heavy chains (HCs) of inter-alpha-trypsin inhibitor (IalphaI) are essential for hyaluronan (HA) organization within the extracellular matrix of the cumulus oophorus, which is critical for in vivo oocyte fertilization and female fertility. In this study, we examined the possibility that these molecules interact and cooperate in this function. We show that HCs and PTX3 colocalize in the cumulus matrix and coimmunoprecipitate from cumulus matrix extracts. Coimmunoprecipitation experiments and solid-phase binding assays performed with purified human IalphaI and recombinant PTX3 demonstrate that their interaction is direct and not mediated by other matrix components. PTX3 does not bind to IalphaI subcomponent bikunin and, accordingly, bikunin does not compete for the binding of PTX3 to IalphaI, indicating that PTX3 interacts with IalphaI subcomponent HC only. Recombinant PTX3-specific N-terminal region, but not the PTX3-pentraxin C-terminal domain, showed the same ability as full-length protein to bind to HCs and to enable HA organization and matrix formation by Ptx3(-/-) cumulus cell oocyte complexes cultured in vitro. Furthermore, a monoclonal antibody raised against PTX3 N terminus, which inhibits PTX3/IalphaI interaction, also prevents recombinant full-length PTX3 from restoring a normal phenotype to in vitro-cultured Ptx3(-/-) cumuli. These results indicate that PTX3 directly interacts with HCs of IalphaI and that such interaction is essential for organizing HA in the viscoelastic matrix of cumulus oophorus, highlighting a direct functional link between the two molecules.


Subject(s)
Alpha-Globulins/metabolism , C-Reactive Protein/metabolism , Serum Amyloid P-Component/metabolism , Alpha-Globulins/chemistry , Animals , C-Reactive Protein/chemistry , Dose-Response Relationship, Drug , Female , Fertility , Fertilization , Humans , Hyaluronic Acid/chemistry , Mice , Mice, Transgenic , Models, Biological , Ovarian Follicle/metabolism , Protein Binding , Recombinant Proteins/chemistry , Serum Amyloid P-Component/chemistry
20.
Semin Reprod Med ; 24(4): 217-27, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16944419

ABSTRACT

The mammalian oocyte is surrounded by several layers of cumulus granulosa cells that nurture the oocyte during its development and actively participate in the process of ovulation. After the ovulatory luteinizing hormone surge, a distinctive program of extracellular matrix production is initiated in the cumulus-oocyte complex. This process known as cumulus expansion or mucification involves synthesis of a backbone of long hyaluronan oligosaccharide chains that are cross-linked by a complex of hyaluronan binding cell surface and extracellular matrix proteins and proteoglycans. Active components of the cumulus matrix are synthesized directly by cumulus cells under the control of endocrine- and oocyte-derived factors, secreted by mural granulosa cells, or enter the follicle in blood plasma. Appropriate composition and assembly of the cumulus matrix is essential for ovulation, efficient passage of the oocyte through the oviduct, and for fertilization. This review describes the critical components and their functional roles in the cumulus matrix, as well as the molecular regulation of cumulus matrix gene expression.


Subject(s)
Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Oocytes/chemistry , Animals , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Female , Fertilization , Gene Expression Regulation , Humans , Hyaluronic Acid/biosynthesis , Kinetics , Models, Biological , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovulation , Proteoglycans/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...