Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
FASEB J ; 35(3): e21397, 2021 03.
Article in English | MEDLINE | ID: mdl-33565176

ABSTRACT

Sperm develop from puberty in the seminiferous tubules, inside the blood-testis barrier to prevent their recognition as "non-self" by the immune system, and it is widely assumed that human sperm-specific proteins cannot access the circulatory or immune systems. Sperm-specific proteins aberrantly expressed in cancer, known as cancer-testis antigens (CTAs), are often pursued as cancer biomarkers and therapeutic targets based on the assumption they are neoantigens absent from the circulation in healthy men. Here, we identify a wide range of germ cell-derived and sperm-specific proteins, including multiple CTAs, that are selectively deposited by the Sertoli cells of the adult mouse and human seminiferous tubules into testicular interstitial fluid (TIF) that is "outside" the blood-testis barrier. From TIF, the proteins can access the circulatory- and immune systems. Disruption of spermatogenesis decreases the abundance of these proteins in mouse TIF, and a sperm-specific CTA is significantly decreased in TIF from infertile men, suggesting that exposure of certain CTAs to the immune system could depend on fertility status. The results provide a rationale for the development of blood-based tests useful in the management of male infertility and indicate CTA candidates for cancer immunotherapy and biomarker development that could show sex-specific and male-fertility-related responses.


Subject(s)
Antigens, Neoplasm/analysis , Proteins/analysis , Seminiferous Tubules/metabolism , Spermatozoa/chemistry , Animals , Blood-Testis Barrier , Extracellular Fluid/chemistry , Humans , Immunotherapy , Infertility, Male/metabolism , Male , Mice , Neoplasms/therapy , Proteome , Sertoli Cells/physiology , Spermatogenesis , Testis/metabolism
2.
Reproduction ; 157(2): R53-R72, 2019 02.
Article in English | MEDLINE | ID: mdl-30576284

ABSTRACT

As germ cells progress through spermatogenesis, they undergo a dramatic transformation, wherein a single, diploid spermatogonial stem cell ultimately produces thousands of highly specialised, haploid spermatozoa. The cytoskeleton is an integral aspect of all eukaryotic cells. It concomitantly provides both structural support and functional pliability, performing key roles in many fundamental processes including, motility, intracellular trafficking, differentiation and cell division. Accordingly, cytoskeletal dynamics underlie many key spermatogenic processes. This review summarises the organisational and functional aspects of the four major cytoskeletal components (actin, microtubules, intermediate filaments and septins) during the various spermatogenic phases in mammals. We focus on the cytoskeletal machinery of both germ cells and Sertoli cells, and thus, highlight the critical importance of a dynamic and precisely regulated cytoskeleton for male fertility.


Subject(s)
Cytoskeleton/physiology , Spermatogenesis/physiology , Animals , Humans , Male , Microtubules/physiology , Spermatozoa/physiology , Spermatozoa/ultrastructure
3.
Semin Cell Dev Biol ; 59: 166-173, 2016 11.
Article in English | MEDLINE | ID: mdl-27353840

ABSTRACT

The purpose of this review is to describe the endocrine and local testicular factors that contribute to the regulation of the blood-testis barrier (BTB), using information gained from in vivo and in vitro models of BTB formation during/after puberty, and from the maintenance of BTB function during adulthood. In vivo the BTB, in part comprised of tight junctions between adjacent somatic Sertoli cells, compartmentalizes meiotic spermatocytes and post-meiotic spermatids away from the vasculature, and therefore prevents autoantibody production by the immune system against these immunogenic germ cells. This adluminal compartment also features a unique biochemical milieu required for the completion of germ cell development. During the normal process of spermatogenesis, earlier germ cells continually cross into the adluminal compartment, but the regulatory mechanisms and changes in junctional proteins that allow this translocation step without causing a 'leak' remain poorly understood. Recent data describing the roles of FSH and androgen on the regulation of Sertoli cell tight junctions and tight junction proteins will be discussed, followed by an examination of the role of paracrine factors, including members of the TGFß superfamily (TGFß3, activin A) and retinoid signalling, as potential mediators of junction assembly and disassembly during the translocation process.


Subject(s)
Blood-Testis Barrier/metabolism , Animals , Endocrine System/metabolism , Humans , Models, Biological , Paracrine Communication
4.
Reproduction ; 154(3): 293-305, 2017 09.
Article in English | MEDLINE | ID: mdl-28667125

ABSTRACT

Experimental autoimmune orchitis (EAO) is a rodent model of chronic testicular inflammation that mimics the pathology observed in some types of human infertility. In a previous study, testicular expression of the inflammatory/immunoregulatory cytokine, activin A, was elevated in adult mice during the onset of EAO, indicating a potential role in the regulation of the disease. Consequently, we examined the development of EAO in mice with elevated levels of follistatin, an endogenous activin antagonist, as a potential therapeutic approach to testicular inflammation. Prior to EAO induction, mice received a single intramuscular injection of a non-replicative recombinant adeno-associated viral vector carrying a gene cassette of the circulating form of follistatin, FST315 (FST group). Serum follistatin levels were increased 5-fold in the FST group compared with the control empty vector (EV) group at 30 and 50 days of EAO, but intra-testicular levels of follistatin or activin A were not significantly altered. Induction of EAO was reduced, but not prevented, with mild-to-severe damage in 75% of the EV group and 40% of the FST group, at 50 days following immunisation with testicular homogenate. However, the EAO damage score (based on disruption of the blood-testis barrier, apoptosis, testicular damage and fibrosis) and extent of intratesticular inflammation (expression of inflammatory mediators) were directly proportional to the levels of activin A measured in the testis at 50 days. These data implicate activin A in the progression of EAO, thereby providing a potential therapeutic target; however, elevating circulating follistatin levels were not sufficient to prevent EAO development.


Subject(s)
Apoptosis , Autoimmune Diseases/physiopathology , Disease Models, Animal , Follistatin/blood , Orchitis/physiopathology , Testis/metabolism , Up-Regulation , Activins/antagonists & inhibitors , Activins/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Biomarkers/blood , Biomarkers/metabolism , Blood-Testis Barrier/immunology , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Blood-Testis Barrier/physiopathology , Disease Progression , Fibrosis , Follistatin/administration & dosage , Follistatin/genetics , Follistatin/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Orchitis/immunology , Orchitis/metabolism , Orchitis/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Testis/immunology , Testis/pathology
5.
Reprod Fertil Dev ; 29(3): 609-620, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26488911

ABSTRACT

A dynamic partnership between follicle-stimulating hormone (FSH) and activin is required for normal Sertoli cell development and fertility. Disruptions to this partnership trigger Sertoli cells to deviate from their normal developmental pathway, as observed in inhibin α-knockout (Inha-KO) mice, which feature Sertoli cell tumours in adulthood. Here, we identified the developmental windows by which adult Sertoli cell tumourigenesis is most FSH sensitive. FSH was suppressed for 7 days in Inha-KO mice and wild-type littermates during the 1st, 2nd or 4th week after birth and culled in the 5th week to assess the effect on adult Sertoli cell development. Tumour growth was profoundly reduced in adult Inha-KO mice in response to FSH suppression during Weeks 1 and 2, but not Week 4. Proliferative Sertoli cells were markedly reduced in adult Inha-KO mice following FSH suppression during Weeks 1, 2 or 4, resulting in levels similar to those in wild-type mice, with greatest effect observed at the 2 week time point. Apoptotic Sertoli cells increased in adult Inha-KO mice after FSH suppression during Week 4. In conclusion, acute FSH suppression during the 1st or 2nd week after birth in Inha-KO mice profoundly suppresses Sertoli cell tumour progression, probably by inhibiting proliferation in the adult, with early postnatal Sertoli cells being most sensitive to FSH action.


Subject(s)
Inhibins/metabolism , Sertoli Cell Tumor/pathology , Spermatogenesis/genetics , Testicular Neoplasms/pathology , Activins/blood , Animals , Follicle Stimulating Hormone/blood , Inhibins/genetics , Male , Mice , Mice, Knockout , Sertoli Cell Tumor/genetics , Sertoli Cell Tumor/metabolism , Sertoli Cells/metabolism , Sertoli Cells/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testis/metabolism , Testis/pathology
6.
Proteomics ; 16(17): 2391-402, 2016 09.
Article in English | MEDLINE | ID: mdl-27324652

ABSTRACT

Communication between the testicular somatic (Sertoli, Leydig, peritubular myoid, macrophage) and germ cell types is essential for sperm production (spermatogenesis), but the communicating factors are poorly understood. We reasoned that identification of proteins in the testicular interstitial fluid (TIF) that bathes these cells could provide a new means to explore spermatogenic function. The aim of this study was to map the proteome of TIF from normal adult rats. Low-abundance proteins in TIF were enriched using ProteoMiner beads and identified by MALDI-MS/MS, recognizing 276 proteins. Comparison with proteomic and genomic databases showed these proteins originated from germ cells, somatic cells (Sertoli, peritubular myoid, Leydig), and blood plasma. In silico analysis revealed homologues of >80% TIF proteins in the human plasma proteome, suggesting ready exchange between these fluids. Only 36% of TIF proteins were common with seminiferous tubule fluid that transports mature spermatids to the epididymis, indicating these two fluids are quite different. This TIF proteome provides an important new resource for the study of intercellular communication in the testis.


Subject(s)
Extracellular Fluid/chemistry , Proteome/analysis , Testis/chemistry , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Male , Proteomics , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
7.
Front Endocrinol (Lausanne) ; 15: 1327800, 2024.
Article in English | MEDLINE | ID: mdl-38654926

ABSTRACT

Introduction: Azoospermia, characterized by an absence of sperm in the ejaculate, represents the most severe form of male infertility. While surgical sperm retrieval in obstructive azoospermia (OA) is successful in the majority of cases, patients with non-obstructive azoospermia (NOA) show retrieval rates of only about 50% and thus frequently have unnecessary surgery. Surgical intervention could be avoided if patients without preserved spermatogenesis are identified preoperatively. This prospective study aimed to discover biomarkers in seminal plasma that could be employed for a non-invasive differential diagnosis of OA/NOA in order to rationalize surgery recommendations and improve success rates. Methods: All patients signed written informed consent, underwent comprehensive andrological evaluation, received human genetics to exclude relevant pathologies, and patients with azoospermia underwent surgical sperm retrieval. Using label-free LC-MS/MS, we compared the proteomes of seminal plasma samples from fertile men (healthy controls (HC), n=8) and infertile men diagnosed with 1) OA (n=7), 2) NOA with successful sperm retrieval (mixed testicular atrophy (MTA), n=8), and 3) NOA without sperm retrieval (Sertoli cell-only phenotype (SCO), n=7). Relative abundance changes of two candidate markers of sperm retrieval, HSPA2 and LDHC, were confirmed by Western Blot. Results: We found the protein expression levels of 42 proteins to be significantly down-regulated (p ≤ 0.05) in seminal plasma from SCO NOA patients relative to HC whereas only one protein was down-regulated in seminal plasma from MTA patients. Analysis of tissue and cell expression suggested that the testis-specific proteins LDHC, PGK2, DPEP3, and germ-cell enriched heat-shock proteins HSPA2 and HSPA4L are promising biomarkers of spermatogenic function. Western blotting revealed a significantly lower abundance of LDHC and HSPA2 in the seminal plasma of men with NOA (SCO and MTA) compared to controls. Discussion: The results indicate that certain testis-specific proteins when measured in seminal plasma, could serve as indicators of the presence of sperm in the testis and predict the success of sperm retrieval. Used in conjunction with conventional clinical assessments, these proteomic biomarkers may assist in the non-invasive diagnosis of idiopathic male infertility.


Subject(s)
Azoospermia , Biomarkers , Proteomics , Semen , Humans , Male , Azoospermia/metabolism , Azoospermia/diagnosis , Semen/metabolism , Semen/chemistry , Biomarkers/metabolism , Biomarkers/analysis , Biomarkers/blood , Adult , Proteomics/methods , Prospective Studies , Sperm Retrieval , Case-Control Studies , Spermatogenesis/physiology
8.
Biol Reprod ; 88(3): 60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23303684

ABSTRACT

Tubulobulbar complexes are cytoskeleton-related membrane structures that develop at sites of intercellular attachment in mammalian seminiferous epithelium. At apical junctions between Sertoli cells and spermatids, the structures internalize adhesion junctions and are a component of the sperm release mechanism. Here we explore the possibility that tubulobulbar complexes that form at the blood-testis barrier are subcellular machines that internalize basal junction complexes. Using electron microscopy, we confirmed that morphologically identifiable tight and gap junctions are present in basal tubulobulbar complexes in rats. In addition, immunological probes for claudin-11 (CLDN11), connexin-43 (GJA1), and nectin-2 (PVRL2) react with linear structures at the light level that we interpret as tubulobulbar complexes, and probes for early endosome antigen 1 (EEA1) and Rab5 (RAB5A) react in similar locations. Significantly, fluorescence patterns for actin and claudin-11 indicate that the amount of junction present is dramatically reduced over the time period that tubulobulbar complexes are known to be most prevalent during spermatogenesis. We also demonstrated, using electron microscopy and fluorescence microscopy, that tubulobulbar complexes develop at basal junctions in primary cultures of Sertoli cells and that like their in vivo counterparts, the structures contain junction proteins. We use this culture system together with transfection techniques to show that junction proteins from one transfected cell occur in structures that project into adjacent nontransfected cells as predicted by the junction internalization hypothesis. On the basis of our findings, we present a new model for basal junction remodeling as it relates to spermatocyte translocation in the seminiferous epithelium.


Subject(s)
Intercellular Junctions/physiology , Seminiferous Epithelium/physiology , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Claudins/metabolism , Connexin 43/metabolism , Endocytosis , Intercellular Junctions/ultrastructure , Male , Nectins , Rats , Rats, Sprague-Dawley , Sertoli Cells/physiology , Vesicular Transport Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism
9.
Front Cell Dev Biol ; 11: 1237273, 2023.
Article in English | MEDLINE | ID: mdl-37564373

ABSTRACT

Adult male fertility depends on spermatogonial stem cells (SSCs) which undergo either self-renewal or differentiation in response to microenvironmental signals. Activin A acts on Sertoli and Leydig cells to regulate key aspects of testis development and function throughout life, including steroid production. Recognising that activin A levels are elevated in many pathophysiological conditions, this study investigates effects of this growth factor on the niche that determines spermatogonial fate. Although activin A can promote differentiation of isolated spermatogonia in vitro, its impacts on SSC and spermatogonial function in vivo are unknown. To assess this, we examined testes of Inha KO mice, which feature elevated activin A levels and bioactivity, and develop gonadal stromal cell tumours as adults. The GFRA1+ SSC-enriched population was more abundant and proliferative in Inha KO compared to wildtype controls, suggesting that chronic elevation of activin A promotes a niche which supports SSC self-renewal. Intriguingly, clusters of GFRA1+/EOMES+/LIN28A- cells, resembling a primitive SSC subset, were frequently observed in tubules adjacent to tumour regions. Transcriptional analyses of Inha KO tumours, tubules adjacent to tumours, and tubules distant from tumour regions revealed disrupted gene expression in each KO group increased in parallel with tumour proximity. Modest transcriptional changes were documented in Inha KO tubules with complete spermatogenesis. Importantly, tumours displaying upregulation of activin responsive genes were also enriched for factors that promote SSC self-renewal, including Gdnf, Igf1, and Fgf2, indicating the tumours generate a supportive microenvironment for SSCs. Tumour cells featured some characteristics of adult Sertoli cells but lacked consistent SOX9 expression and exhibited an enhanced steroidogenic phenotype, which could arise from maintenance or acquisition of a fetal cell identity or acquisition of another somatic phenotype. Tumour regions were also heavily infiltrated with endothelial, peritubular myoid and immune cells, which may contribute to adjacent SSC support. Our data show for the first time that chronically elevated activin A affects SSC fate in vivo. The discovery that testis stromal tumours in the Inha KO mouse create a microenvironment that supports SSC self-renewal but not differentiation offers a strategy for identifying pathways that improve spermatogonial propagation in vitro.

10.
PLoS One ; 18(9): e0290846, 2023.
Article in English | MEDLINE | ID: mdl-37656709

ABSTRACT

Sertoli cells support the development of sperm and the function of various somatic cells in the interstitium between the tubules. Sertoli cells regulate the function of the testicular vasculature and the development and function of the Leydig cells that produce testosterone for fertility and virility. However, the Sertoli cell-derived factors that regulate these cells are largely unknown. To define potential mechanisms by which Sertoli cells could support testicular somatic cell function, we aimed to identify Sertoli cell-enriched proteins in the testicular interstitial fluid (TIF) between the tubules. We previously resolved the proteome of TIF in mice and humans and have shown it to be a rich source of seminiferous tubule-derived proteins. In the current study, we designed bioinformatic strategies to interrogate relevant proteomic and genomic datasets to identify Sertoli cell-enriched proteins in mouse and human TIF. We analysed proteins in mouse TIF that were significantly reduced after one week of acute Sertoli cell ablation in vivo and validated which of these are likely to arise primarily from Sertoli cells based on relevant mouse testis RNASeq datasets. We used a different, but complementary, approach to identify Sertoli cell-enriched proteins in human TIF, taking advantage of high-quality human testis genomic, proteomic and immunohistochemical datasets. We identified a total of 47 and 40 Sertoli cell-enriched proteins in mouse and human TIF, respectively, including 15 proteins that are conserved in both species. Proteins with potential roles in angiogenesis, the regulation of Leydig cells or steroidogenesis, and immune cell regulation were identified. The data suggests that some of these proteins are secreted, but that Sertoli cells also deposit specific proteins into TIF via the release of extracellular vesicles. In conclusion, we have identified novel Sertoli cell-enriched proteins in TIF that are candidates for regulating somatic cell-cell communication and testis function.


Subject(s)
Sertoli Cells , Testis , Humans , Male , Animals , Mice , Extracellular Fluid , Proteomics , Semen
11.
Biol Reprod ; 87(1): 13, 1-11, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22492971

ABSTRACT

New data have challenged the convention that the adult Sertoli cell population is fixed and unmodifiable. The Sertoli cell has two distinct functions: 1) formation of the seminiferous cords and 2) provision of nutritional and structural support to developing germ cells. For these to occur successfully, Sertoli cells must undergo many maturational changes between fetal and adult life, the main switches occurring around puberty, including the loss of proliferative activity and the formation of the blood-testis barrier. Follicle-stimulating hormone plays a key role in promoting Sertoli cell proliferation, while thyroid hormone inhibits proliferative activity in early postnatal life. Together these regulate the Sertoli-germ cell complement and sperm output in adulthood. By puberty, the Sertoli cell population is considered to be stable and unmodifiable by hormones. But there is mounting evidence that the size of the adult Sertoli cell population and its maturational status is modifiable by hormones and that Sertoli cells can gain proliferative ability in the spermatogenically disrupted hamster and human model. This new information demonstrates that the adult Sertoli cell population, at least in the settings of testicular regression in the hamster and impaired fertility in humans in vivo and from mice and men in vitro, is not a terminally differentiated population. Data from the hamster now show that the adult Sertoli cell population size is regulated by hormones. This creates exciting prospects for basic and clinical research in testis biology. The potential to replenish an adult Sertoli-germ cell complement to normal in a setting of infertility may now be realized.


Subject(s)
Sertoli Cells/cytology , Animals , Autocrine Communication/physiology , Cell Cycle Proteins/physiology , Cell Differentiation/physiology , Cell Proliferation , Humans , Intercellular Junctions/physiology , Male , Models, Biological , Paracrine Communication/physiology , Puberty/physiology , Seasons , Sertoli Cells/physiology , Sexual Maturation/physiology , Signal Transduction/physiology , Spermatogenesis/physiology
12.
Biol Reprod ; 87(2): 38, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22623623

ABSTRACT

Sertoli cell tight junctions (TJs) form at puberty as a major component of the blood-testis barrier (BTB), which is essential for spermatogenesis. This study characterized the hormonal induction of functional Sertoli cell TJ formation in vivo using the gonadotropin-deficient hypogonadal (hpg) mouse that displays prepubertal spermatogenic arrest. Androgen actions were determined in hpg mice treated for 2 or 10 days with dihydrotestosterone (DHT). Follicle-stimulating hormone (FSH) actions were studied in hpg mice expressing transgenic human FSH (hpg+tgFSH) with or without DHT treatment. TJ formation was examined by mRNA expression and immunolocalization of TJ proteins claudin-3 and claudin-11, and barrier functionality was examined by biotin tracer permeability. Immunolocalization of claudin-3 and claudin-11 was extensive at wild-type (wt) Sertoli cell TJs, which functionally excluded permeability tracer. In contrast, seminiferous tubules of hpg testes lacked claudin-3, but claudin-11 protein was present in adluminal regions of Sertoli cells. Biotin tracer permeated throughout these tubules, demonstrating dysfunctional TJs. In hpg+tgFSH testes, claudin-3 was generally absent, but claudin-11 had redistributed basally toward the TJs, where function was variable. In hpg testes, DHT treatment stimulated the redistribution of claudin-11 protein toward the basal region of Sertoli cells by Day 2, increased Cldn3 and Cldn11 mRNA expression, then induced the formation of functional TJs containing both proteins by Day 10. In hpg+tgFSH testes, TJ protein redistribution was accelerated and functional TJs formed by Day 2 of DHT treatment. We conclude that androgen stimulates initial Sertoli cell TJ formation and function in mice, whereas FSH activity is insufficient alone, but augments androgen-induced TJ function.


Subject(s)
Androgens/physiology , Follicle Stimulating Hormone/physiology , Sertoli Cells/physiology , Tight Junctions/physiology , Animals , Connexins/metabolism , Dihydrotestosterone , Disease Models, Animal , Humans , Hypogonadism , Male , Mice , Mice, Transgenic , Organ Size , RNA, Messenger/metabolism , Rats
13.
Hum Reprod ; 26(10): 2841-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21840908

ABSTRACT

BACKGROUND: Human trophoblast invasion and differentiation are essential for a successful pregnancy outcome. Dysregulation of these processes can lead to placental pathologies such as pre-eclampsia. The molecular mechanisms; however, are poorly understood. Interleukin (IL)11--a cytokine that regulates endometrial epithelial cell adhesion, trophoblast motility and invasion during implantation--may be involved in some of these processes. METHODS AND RESULTS: The effect of IL11 on protein expression was investigated in trophoblastic HTR8/SVneo cells and primary extravillous trophoblasts (EVTs) purified from first- trimester placentas. Two-dimension (2D)-differential in-gel electrophoresis analyses revealed that 731 spots were significantly differentially regulated by IL11 in HTR8/SVneo cells: seven spots were analyzed by liquid chromatography-tandem mass spectrometry and 14 unique proteins identified. Protein disulfide isomerase family A, member 3 (PDIA3; endoplasmic reticulum p57) and glucose-regulated protein 78 (GRP78) were further validated to be regulated by IL11 in HTR8/SVneo and primary EVT. One dimension western blot analysis confirmed that PDIA3 was down-regulated in EVT. 2D western blot analysis revealed that GRP78 was post-translationally modified following IL11 treatment. Moreover, IL11 stimulated the secretion of GRP78 in EVT. CONCLUSIONS: Data suggest that IL11, possibly via signal transducers and activators of transcription 3 signaling pathway, regulates PDIA3 protein expression and modification/secretion of GRP78. This is the first study to identify PDIA3 and GRP78 as IL11 targets in invasive trophoblasts and identifies a possible mechanism by which IL11 regulates trophoblast function.


Subject(s)
Interleukin-11/physiology , Protein Disulfide-Isomerases/biosynthesis , Trophoblasts/cytology , Cell Culture Techniques , Chromatography, Liquid/methods , Electrophoresis, Gel, Two-Dimensional/methods , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression Profiling/methods , Heat-Shock Proteins/biosynthesis , Humans , Interleukin-11/metabolism , Mass Spectrometry/methods , Models, Biological , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Protein Processing, Post-Translational , Trophoblasts/metabolism
14.
Reprod Biol Endocrinol ; 9: 73, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21619711

ABSTRACT

BACKGROUND: During the peri-implantation period, the embryo adheres to an adequately prepared or receptive endometrial surface epithelium. Abnormal embryo adhesion to the endometrium results in embryo implantation failure and infertility. Endometrial epithelial cell plasma membrane proteins critical in regulating adhesion may potentially be infertility biomarkers or targets for treating infertility. Interleukin (IL) 11 regulates human endometrial epithelial cells (hEEC) adhesion. Its production is abnormal in women with infertility. The objective of the study was to identify IL11 regulated plasma membrane proteins in hEEC in vitro using a proteomic approach. METHODS: Using a 2D-differential in-gel electrophoresis (DIGE) electrophoresis combined with LCMS/MS mass spectrometry approach, we identified 20 unique plasma membrane proteins differentially regulated by IL11 in ECC-1 cells, a hEEC derived cell line. Two IL11 regulated proteins with known roles in cell adhesion, annexin A2 (ANXA2) and flotillin-1 (FLOT1), were validated by Western blot and immunocytochemistry in hEEC lines (ECC-1 and an additional cell line, Ishikawa) and primary hEEC. Flotilin-1 was further validated by immunohistochemistry in human endometrium throughout the menstrual cycle (n = 6-8/cycle). RESULTS: 2D-DIGE analysis identified 4 spots that were significantly different between control and IL11 treated group. Of these 4 spots, there were 20 proteins that were identified with LCMS/MS. Two proteins; ANXA2 and FLOT1 were chosen for further analyses and have found to be significantly up-regulated following IL11 treatment. Western blot analysis showed a 2-fold and a 2.5-fold increase of ANXA2 in hEEC membrane fraction of ECC-1 and Ishikawa cells respectively. Similarly, a 1.8-fold and a 2.3/2.4-fold increase was also observed for FLOT1 in hEEC membrane fraction of ECC-1 and Ishikawa cells respectively. In vitro, IL11 induced stronger ANXA2 expression on cell surface of primary hEEC and ECC-1 whilst, the lipid-raft protein FLOT1 demonstrated punctate staining in the apical surface of ECC-1 plasma membranes and was upregulated in the epithelium in the receptive phase of the menstrual cycle (p lower or equal 0.05). CONCLUSIONS: This is the first study to use a proteomics approach to identify hEEC plasma membrane proteins that may be useful as infertility markers or pharmacological targets for fertility regulation.


Subject(s)
Endometrium/drug effects , Epithelial Cells/drug effects , Interleukin-11/pharmacology , Membrane Proteins/isolation & purification , Proteomics , Adult , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Endometrium/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Humans , Interleukin-11/physiology , Mass Spectrometry , Membrane Proteins/analysis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteome/analysis , Two-Dimensional Difference Gel Electrophoresis
16.
Mol Cell Endocrinol ; 527: 111216, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33639219

ABSTRACT

The testis is a temperature-sensitive organ that needs to be maintained 2-7 °C below core body temperature to ensure the production of normal sperm. Failure to maintain testicular temperature in mammals impairs spermatogenesis and leads to low sperm counts, poor sperm motility and abnormal sperm morphology in the ejaculate. This review discusses the recent knowledge on the response of testicular somatic cells to heat stress and, specifically, regarding the relevant contributions of heat, germ cell depletion and inflammatory reactions on the functions of Sertoli and Leydig cells. It also outlines mechanisms of testicular thermoregulation, as well as the thermogenic factors that impact testicular function.


Subject(s)
Leydig Cells/metabolism , Sertoli Cells/metabolism , Sperm Motility , Spermatogenesis , Spermatozoa/metabolism , Animals , Heat-Shock Response , Humans , Male
17.
Reprod Sci ; 28(10): 2916-2928, 2021 10.
Article in English | MEDLINE | ID: mdl-34008157

ABSTRACT

Cryptorchidism causes spermatogenic failure and reduced serum androgen levels, as well as testicular oedema and fibrosis, which are hallmarks of inflammation. However, the role of inflammation and the effects of cryptorchidism on Sertoli cell and Leydig cell function at the molecular level remain ill-defined. Bilateral cryptorchidism was surgically induced in adult rats for 7 and 14 weeks. Testis weights decreased to 40% of normal within 7 weeks, due to loss of all developing spermatogenic cells except spermatogonia, but did not decrease further at 14 weeks. Serum FSH and LH were increased at both time points, consistent with a loss of feedback by inhibin and testosterone. This damage was accompanied by progressive accumulation of interstitial fluid and peritubular fibrosis, and a progressive decline of several critical Sertoli cell genes (Sox9, Inha (inhbin α-subunit), Cldn11 (claudin 11), Gja1 (connexin 43), and Il1a (interleukin-1α)) and the Leydig cell steroidogenic enzymes, Cyp11a1, Hsd3b1, and Hs17b3. Activin B and the activin-binding protein, follistatin, also declined, but the intratesticular concentration of activin A, which is a regulator of inflammatory responses, was largely unaffected at either time point. Expression of genes involved in inflammation (Tnf, Il10, Il1b, Mcp1) and fibrosis (Acta2, Col1a1) were considerably elevated at both time points. These data indicate that induction of experimental cryptorchidism, which causes complete failure of spermatogenesis in the adult rat, also induces chronic testicular inflammation, manifesting in oedema and fibrosis, and a progressive decline of Sertoli and Leydig cell gene expression and function.


Subject(s)
Cryptorchidism/metabolism , Disease Progression , Inflammation Mediators/metabolism , Leydig Cells/metabolism , Sertoli Cells/metabolism , Animals , Cryptorchidism/pathology , Leydig Cells/pathology , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/pathology , Testis/metabolism , Testis/pathology
18.
Proteomics ; 10(2): 332-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19899076

ABSTRACT

We have developed an optimized procedure using dual size exclusion/affinity hydrogel nanoparticles to capture and comparatively analyze low molecular mass proteins directly from biological samples. The method described facilitates charge- and size-dependent protein binding, direct analysis by MS or other means and is highly reproducible. A comparative analysis of the low molecular mass proteome of plasma following freeze-thaw immediately after venipuncture is used to illustrate proof-of-concept. The technique described is rapid and may be easily reproduced in any laboratory.


Subject(s)
Blood Proteins/analysis , Chromatography, Gel/methods , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Nanoparticles/chemistry , Proteomics/methods , Blood Proteins/chemistry , Humans
19.
J Proteome Res ; 9(3): 1437-49, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-19943703

ABSTRACT

Intestinal adaptation in response to the loss of the small intestine is essential to restore enteral autonomy in patients who have undergone massive small bowel resection (MSBR). In a proportion of patients, intestinal function is not restored, resulting in chronic intestinal failure (IF). Early referral of such patients for transplant provides the best prognosis; however, the molecular mechanisms underlying intestinal adaptation remain elusive and there is currently no convenient marker to predict whether patients will develop IF. We have investigated the adaptation response in a well-characterized porcine model of intestinal adaptation. 2D DIGE analysis of ileal epithelium from piglets recovering from massive small bowel resection (MSBR) identified over 60 proteins that changed specifically in MSBR animals relative to nonoperational or sham-operated controls. Three fatty acid binding proteins (L-FABP, FABP-6, and I-FABP) showed changes in MSBR animals. The expression changes and localization of each FABP were validated by immunoblotting and immunohistochemical analysis. FABP expression changes in MSBR animals occurred concurrently with altered triglyceride and bile acid metabolism as well as weight gain. The observed FABP expression changes in the ileal epithelium occur as part of the intestinal adaptation response and could provide a clinically useful marker to evaluate adaptation following MSBR.


Subject(s)
Adaptation, Biological/physiology , Fatty Acid-Binding Proteins/biosynthesis , Intestinal Mucosa/metabolism , Intestine, Small/physiology , Intestine, Small/surgery , Proteomics/methods , Adaptation, Biological/genetics , Animals , Cluster Analysis , Electrophoresis, Gel, Two-Dimensional , Fatty Acid-Binding Proteins/analysis , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation , Ileum/cytology , Ileum/metabolism , Ileum/physiology , Intestine, Small/metabolism , Reproducibility of Results , Short Bowel Syndrome/genetics , Short Bowel Syndrome/metabolism , Signal Transduction , Swine
20.
J Proteome Res ; 9(5): 2438-49, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20199104

ABSTRACT

Endometriosis is a chronic disorder affecting approximately 10% of women in whom endometrial tissue forms painful lesions outside the uterus. It has a major impact on their physical, mental and social well-being but has no known cure, and there is no nonsurgical means of diagnosis. We have used a proteomic approach to identify proteins with altered abundance in the eutopic endometrium of endometriosis patients in the midsecretory phase of the menstrual cycle. 2D-differential in gel electrophoresis (DIGE) and mass spectrometry identified 20 proteins that were present at different levels in endometriosis patients (p < 0.05), many of which have not previously been associated with endometriosis. Protein abundance changes did not correlate well with published gene array data, emphasizing the extensive post-translational modification that occurs in this tissue. Abundance or localization changes in endometrial tissue were validated by immunohistochemistry and Western blotting for three proteins, vimentin (VIM), peroxiredoxin 6 (PRDX6), and ribonuclease/angiogenin inhibitor 1 (RNH1), while observed changes could not be confirmed for coronin 1A (CORO1A) or transgelin (TAGLN2). In addition, multiple charge and size isoforms were observed for PDRX6 and vimentin (VIM), and an additional PDRX6 isoform was observed in endometriosis patients that was below the level of detection in healthy women. Biological pathway analysis identified that cytoskeletal remodeling via keratin intermediate filaments, processing of the cystic fibrosis transmembrane receptor (CFTR), the glucocorticoid receptor subunit alpha (GCR), and heat shock factor 1 (HSF1) were all significantly over-represented features in endometriosis patients. This study highlights the highly dynamic nature of endometrial tissue and suggests that considerable post-translational modification of proteins is a key factor in the pathology of endometriosis.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Endometriosis/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Blotting, Western , Carrier Proteins/metabolism , Cluster Analysis , Databases, Genetic , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Peroxiredoxin VI/metabolism , Protein Isoforms , Proteomics/methods , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL