Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36834705

ABSTRACT

In early 2020, the novel pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wuhan, China, and rapidly propagated worldwide causing a global health emergency. SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) protein for cell entry, followed by proteolytic cleavage of the Spike (S) protein by the transmembrane serine protease 2 (TMPRSS2), allowing fusion of the viral and cellular membranes. Interestingly, TMPRSS2 is a key regulator in prostate cancer (PCa) progression which is regulated by androgen receptor (AR) signaling. Our hypothesis is that the AR signaling may regulate the expression of TMPRSS2 in human respiratory cells and thus influence the membrane fusion entry pathway of SARS-CoV-2. We show here that TMPRSS2 and AR are expressed in Calu-3 lung cells. In this cell line, TMPRSS2 expression is regulated by androgens. Finally, pre-treatment with anti-androgen drugs such as apalutamide significantly reduced SARS-CoV-2 entry and infection in Calu-3 lung cells but also in primary human nasal epithelial cells. Altogether, these data provide strong evidence to support the use of apalutamide as a treatment option for the PCa population vulnerable to severe COVID-19.


Subject(s)
COVID-19 , Male , Humans , COVID-19/metabolism , SARS-CoV-2/metabolism , Peptidyl-Dipeptidase A/metabolism , Lung/metabolism , Epithelial Cells/metabolism , Virus Internalization
2.
Br J Cancer ; 128(5): 918-927, 2023 03.
Article in English | MEDLINE | ID: mdl-36550208

ABSTRACT

BACKGROUND: Neuroendocrine prostate cancer (NEPC) is a multi-resistant variant of prostate cancer (PCa) that has become a major challenge in clinics. Understanding the neuroendocrine differentiation (NED) process at the molecular level is therefore critical to define therapeutic strategies that can prevent multi-drug resistance. METHODS: Using RNA expression profiling and immunohistochemistry, we have identified and characterised a gene expression signature associated with the emergence of NED in a large PCa cohort, including 169 hormone-naïve PCa (HNPC) and 48 castration-resistance PCa (CRPC) patients. In vitro and preclinical in vivo NED models were used to explore the cellular mechanism and to characterise the effects of castration on PCa progression. RESULTS: We show for the first time that Neuropilin-1 (NRP1) is a key component of NED in PCa cells. NRP1 is upregulated in response to androgen deprivation therapies (ADT) and elicits cell survival through induction of the PKC pathway. Downmodulation of either NRP1 protein expression or PKC activation suppresses NED, prevents tumour evolution toward castration resistance and increases the efficacy of docetaxel-based chemotherapy in preclinical models in vivo. CONCLUSIONS: This study reveals the NRP1/PKC axis as a promising therapeutic target for the prevention of neuroendocrine castration-resistant variants of PCa and indicates NRP1 as an early transitional biomarker.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Neuropilin-1 , Prostatic Neoplasms, Castration-Resistant/pathology , Androgen Antagonists , Drug Resistance , Cell Differentiation , Cell Line, Tumor
3.
J Extracell Vesicles ; 11(10): e12269, 2022 10.
Article in English | MEDLINE | ID: mdl-36271885

ABSTRACT

Small Extracellular Vesicles (sEVs) are 50-200 nm in diameter vesicles delimited by a lipid bilayer, formed within the endosomal network or derived from the plasma membrane. They are secreted in various biological fluids, including airway nasal mucus. The goal of this work was to understand the role of sEVs present in the mucus (mu-sEVs) produced by human nasal epithelial cells (HNECs) in SARS-CoV-2 infection. We show that uninfected HNECs produce mu-sEVs containing SARS-CoV-2 receptor ACE2 and activated protease TMPRSS2. mu-sEVs cleave prefusion viral Spike proteins at the S1/S2 boundary, resulting in higher proportions of prefusion S proteins exposing their receptor binding domain in an 'open' conformation, thereby facilitating receptor binding at the cell surface. We show that the role of nasal mu-sEVs is to complete prefusion Spike priming performed by intracellular furin during viral egress from infected cells. This effect is mediated by vesicular TMPRSS2 activity, rendering SARS-CoV-2 virions prone to entry into target cells using the 'early', TMPRSS2-dependent pathway instead of the 'late', cathepsin-dependent route. These results indicate that prefusion Spike priming by mu-sEVs in the nasal cavity plays a role in viral tropism. They also show that nasal mucus does not protect from SARS-CoV-2 infection, but instead facilitates it.


Subject(s)
COVID-19 , Extracellular Vesicles , Humans , Spike Glycoprotein, Coronavirus/chemistry , Furin , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , Proviruses/metabolism , Lipid Bilayers , Virus Internalization , Epithelial Cells/metabolism , Extracellular Vesicles/metabolism , Cathepsins
4.
J Extracell Vesicles ; 11(5): e12210, 2022 05.
Article in English | MEDLINE | ID: mdl-35527349

ABSTRACT

Long noncoding (lnc)RNAs modulate gene expression alongside presenting unexpected source of neoantigens. Despite their immense interest, their ability to be transferred and control adjacent cells is unknown. Extracellular Vesicles (EVs) offer a protective environment for nucleic acids, with pro and antitumourigenic functions by controlling the immune response. In contrast to extracellular nonvesicular RNA, few studies have addressed the full RNA content within human fluids' EVs and have compared them with their tissue of origin. Here, we performed Total RNA-Sequencing on six Formalin-Fixed-Paraffin-Embedded (FFPE) prostate cancer (PCa) tumour tissues and their paired urinary (u)EVs to provide the first whole transcriptome comparison from the same patients. UEVs contain simplified transcriptome with intron-free cytoplasmic transcripts and enriched lnc/circular (circ)RNAs, strikingly common to an independent 20 patients' urinary cohort. Our full cellular and EVs transcriptome comparison within three PCa cell lines identified a set of overlapping 14 uEV-circRNAs characterized as essential for prostate cell proliferation in vitro and 28 uEV-lncRNAs belonging to the cancer-related lncRNA census (CLC2). In addition, we found 15 uEV-lncRNAs, predicted to encode 768 high-affinity neoantigens, and for which three of the encoded-ORF produced detectable unmodified peptides by mass spectrometry. Our dual analysis of EVs-lnc/circRNAs both in urines' and in vitro's EVs provides a fundamental resource for future uEV-lnc/circRNAs phenotypic characterization involved in PCa.


Subject(s)
Extracellular Vesicles , MicroRNAs , Prostatic Neoplasms , RNA, Long Noncoding , Extracellular Vesicles/genetics , Humans , Male , MicroRNAs/genetics , Prostatic Neoplasms/genetics , RNA, Circular , RNA, Long Noncoding/genetics , Transcriptome
5.
Int J Mol Sci ; 23(9)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35562881

ABSTRACT

Prostate cancer (PCa) is the second most frequent cancer and the fifth leading cause of cancer death in men worldwide. If local PCa presents a favorable prognosis, available treatments for advanced PCa display limiting benefits due to therapeutic resistances. Nucleolin (NCL) is a ubiquitous protein involved in numerous cell processes, such as ribosome biogenesis, cell cycles, or angiogenesis. NCL is overexpressed in several tumor types in which it has been proposed as a diagnostic and prognostic biomarker. In PCa, NCL has mainly been studied as a target for new therapeutic agents. Nevertheless, little data are available concerning its expression in patient tissues. Here, we investigated the expression of NCL using a new cohort from Mondor Hospital and data from published cohorts. Results were then compared with NCL expression using in vitro models. NCL was overexpressed in PCa tissues compared to the normal tissues, but no prognostic values were demonstrated. Nine genes were highly co-expressed with NCL in patient tissues and tumor prostate cell lines. Our data demonstrate that NCL is an interesting diagnostic biomarker and propose a signature of genes co-expressed with NCL.


Subject(s)
Prostatic Neoplasms , RNA-Binding Proteins , Biomarkers , Humans , Male , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prostatic Neoplasms/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Nucleolin
6.
Stem Cell Reports ; 17(4): 936-952, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35334216

ABSTRACT

Male infertility is responsible for approximately half of all cases of reproductive issues. Spermatogenesis originates in a small pool of spermatogonial stem cells (SSCs), which are of interest for therapy of infertility but remain not well defined in humans. Using multiparametric analysis of the side population (SP) phenotype and the α-6 integrin, THY1, and ß-2 microglobulin cell markers, we identified a population of human primitive undifferentiated spermatogonia with the phenotype ß-2 microglobulin (ß-2M)-SPα-6+THY1+, which is highly enriched in stem cells. By analyzing the expression signatures of this SSC-enriched population along with other germinal progenitors, we established an exhaustive transcriptome of human spermatogenesis. Transcriptome profiling of the human ß-2M-SPα-6+THY1+ population and comparison with the profile of mouse undifferentiated spermatogonia provide insights into the molecular networks and key transcriptional regulators regulating human SSCs, including the basic-helix-loop-helix (bHLH) transcriptional repressor HES1, which we show to be implicated in maintenance of SSCs in vitro.


Subject(s)
Adult Germline Stem Cells , Spermatogenesis , Animals , Gene Expression Profiling , Humans , Male , Mice , Spermatogenesis/genetics , Spermatogonia/metabolism , Stem Cells/metabolism , Testis/metabolism , Transcription Factors/metabolism
7.
Hum Mol Genet ; 31(1): 97-110, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34368842

ABSTRACT

Fanconi anemia (FA) is a rare human genetic disorder characterized by bone marrow failure, predisposition to cancer and developmental defects including hypogonadism. Reproductive defects leading to germ cell aplasia are the most consistent phenotypes seen in FA mouse models. We examined the role of the nuclear FA core complex gene Fancg in the development of primordial germ cells (PGCs), the embryonic precursors of adult gametes, during fetal development. PGC maintenance was severely impaired in Fancg-/- embryos. We observed a defect in the number of PGCs starting at E9.5 and a strong attrition at E11.5 and E13.5. Remarkably, we observed a mosaic pattern reflecting a portion of testicular cords devoid of PGCs in E13.5 fetal gonads. Our in vitro and in vivo data highlight a potential role of Fancg in the proliferation and in the intrinsic cell motility abilities of PGCs. The random migratory process is abnormally activated in Fancg-/- PGCs, altering the migration of cells. Increased cell death and PGC attrition observed in E11.5 Fancg-/- embryos are features consistent with delayed migration of PGCs along the migratory pathway to the genital ridges. Moreover, we show that an inhibitor of RAC1 mitigates the abnormal migratory pattern observed in Fancg-/- PGCs.


Subject(s)
Fanconi Anemia , Animals , Cell Movement/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group G Protein/metabolism , Germ Cells/metabolism , Gonads/metabolism , Mice , Signal Transduction
8.
Cancers (Basel) ; 13(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34359692

ABSTRACT

Prostate cancer (PCa) is the second most frequent cancer and the fifth leading cause of cancer death among men worldwide. At first, advanced PCa is treated by androgen deprivation therapy with a good initial response. Nevertheless, recurrences occur, leading to Castrate-Resistance Prostate Cancer (CRPC). During the last decade, new therapies based on inhibition of the androgen receptor pathway or taxane chemotherapies have been used to treat CRPC patients leading to an increase in overall survival, but the occurrence of resistances limits their benefits. Numerous studies have demonstrated the implication of extracellular vesicles (EVs) in different cancer cellular mechanisms. Thus, the possibility to isolate and explore EVs produced by tumor cells in plasma/sera represents an important opportunity for the deciphering of those mechanisms and the discovery of biomarkers. Herein, we summarized the role of EVs in therapeutic resistance of advanced prostate cancer and their use to find biomarkers able to predict these resistances.

9.
BMC Cancer ; 21(1): 394, 2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33845808

ABSTRACT

BACKGROUND: RNA-seq data are increasingly used to derive prognostic signatures for cancer outcome prediction. A limitation of current predictors is their reliance on reference gene annotations, which amounts to ignoring large numbers of non-canonical RNAs produced in disease tissues. A recently introduced kind of transcriptome classifier operates entirely in a reference-free manner, relying on k-mers extracted from patient RNA-seq data. METHODS: In this paper, we set out to compare conventional and reference-free signatures in risk and relapse prediction of prostate cancer. To compare the two approaches as fairly as possible, we set up a common procedure that takes as input either a k-mer count matrix or a gene expression matrix, extracts a signature and evaluates this signature in an independent dataset. RESULTS: We find that both gene-based and k-mer based classifiers had similarly high performances for risk prediction and a markedly lower performance for relapse prediction. Interestingly, the reference-free signatures included a set of sequences mapping to novel lncRNAs or variable regions of cancer driver genes that were not part of gene-based signatures. CONCLUSIONS: Reference-free classifiers are thus a promising strategy for the identification of novel prognostic RNA biomarkers.


Subject(s)
Biomarkers, Tumor , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Transcriptome , Algorithms , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Male , Prognosis , Prostatic Neoplasms/pathology , Recurrence , Reproducibility of Results , Supervised Machine Learning
10.
Oncogene ; 40(6): 1118-1127, 2021 02.
Article in English | MEDLINE | ID: mdl-33323967

ABSTRACT

Dysregulated androgen receptor (AR) plays a crucial role in prostate cancer (PCa) development, though further factors involved in its regulation remain to be identified. Recently, paradoxical results were reported on the implication of the MEN1 gene in PCa. To dissect its role in prostate luminal cells, we generated a mouse model with inducible Men1 disruption in Nkx3.1-deficient mice in which mouse prostatic intraepithelial neoplasia (mPIN) occur. Prostate glands from mutant and control mice were analyzed pathologically and molecularly; cellular and molecular analyses were carried out in PCa cell lines after MEN1 knockdown (KD) by siRNA. Double-mutant mice developed accelerated mPIN and later displayed microinvasive adenocarcinoma. Markedly, early-stage lesions exhibited a decreased expression of AR and its target genes, accompanied by reduced CK18 and E-cadherin expression, suggesting a shift from a luminal to a dedifferentiated epithelial phenotype. Intriguingly, over 60% of menin-deficient cells expressed CD44 at a later stage. Furthermore, MEN1 KD led to the increase in CD44 expression in PC3 cells re-expressing AR. Menin bound to the proximal AR promoter and regulated AR transcription via the H3K4me3 histone mark. Interestingly, the cell proliferation of AR-dependent cells (LNCaP, 22Rv1, and VCaP), but not of AR-independent cells (DU145, PC3), responded strongly to MEN1 silencing. Finally, menin expression was found reduced in some human PCa. These findings highlight the regulation of the AR promoter by menin and the crosstalk between menin and the AR pathway. Our data could be useful for better understanding the increasingly reported AR-negative/NE-negative subtype of PCa and the mechanisms underlying its development.


Subject(s)
Homeodomain Proteins/genetics , Hyaluronan Receptors/genetics , Prostatic Intraepithelial Neoplasia/genetics , Proto-Oncogene Proteins/genetics , Receptors, Androgen/genetics , Transcription Factors/genetics , Animals , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prostate/metabolism , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Signal Transduction
11.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198087

ABSTRACT

Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Infertility, Male/genetics , Quantitative Trait Loci/genetics , Acrosome/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/physiology , Epididymis/physiology , Female , Humans , Male , Mice , Phenotype , Spermatids/physiology , Spermatogenesis/genetics , Spermatozoa/physiology , Teratozoospermia/genetics
12.
Cancers (Basel) ; 12(10)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33049910

ABSTRACT

Numerous pan-genomic studies identified alterations in protein-coding genes and signaling pathways involved in bladder carcinogenesis, while non-coding somatic alterations remain weakly explored. The goal of this study was to identify clinical biomarkers in non-coding regions for bladder cancer patients. We have previously identified in bladder tumors two non-coding mutational hotspots occurring at high frequencies (≥30%). These mutations are located close to the GPR126 and PLEKHS1 genes, at the guanine or the cytosine of a TGAACA core motif flanked, on both sides, by a stretch of palindromic sequences. Here, we hypothesize that such a pattern of recurrent non-coding mutations could be a signature of somatic genomic instability specifically involved in bladder cancer. We analyzed 26 additional mutable non-coding sites with the same core motif in a cohort of 103 bladder cancers composed of 44 NMIBC cases and 59 MIBC cases using high-resolution melting (HRM) and Sanger sequencing. Five bladder cancers were additionally analyzed for protein-coding gene mutations using a targeted NGS panel composed of 571 genes. Expression levels of three members of the APOBEC3 family genes were assessed using real-time quantitative RT-PCR. Non-coding somatic mutations were observed for at least one TGAACA core motif locus in 62.1% (64/103) of bladder tumor samples. These non-coding mutations co-occurred in the bladder tumors but were absent in prostate tumor, HPV-positive Head and Neck Squamous Cell Carcinoma, and high microsatellite instability (MSI-H) colorectal tumor series. This signature of palindromic non-coding somatic mutations, specific to bladder tumors, was not associated with patients' outcome and was more frequent in females. Interestingly, this signature was associated with high tumor mutational burden (TMB) and high expression levels of APOBEC3B and interferon inducible genes. We identified a new type of somatic genomic instability targeting the TGAACA core motif loci flanked by palindromic sequences in bladder cancer. This mutational signature is a promising candidate clinical biomarker for the early detection of relapse and a major low-cost alternative to the TMB to monitor the response to immunotherapy for bladder cancer patients.

13.
Nature ; 577(7792): E10, 2020 01.
Article in English | MEDLINE | ID: mdl-31911658

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Life Sci Alliance ; 2(6)2019 12.
Article in English | MEDLINE | ID: mdl-31732695

ABSTRACT

The use of RNA-sequencing technologies held a promise of improved diagnostic tools based on comprehensive transcript sets. However, mining human transcriptome data for disease biomarkers in clinical specimens are restricted by the limited power of conventional reference-based protocols relying on unique and annotated transcripts. Here, we implemented a blind reference-free computational protocol, DE-kupl, to infer yet unreferenced RNA variations from total stranded RNA-sequencing datasets of tissue origin. As a bench test, this protocol was powered for detection of RNA subsequences embedded into putative long noncoding (lnc)RNAs expressed in prostate cancer. Through filtering of 1,179 candidates, we defined 21 lncRNAs that were further validated by NanoString for robust tumor-specific expression in 144 tissue specimens. Predictive modeling yielded a restricted probe panel enabling more than 90% of true-positive detections of cancer in an independent The Cancer Genome Atlas cohort. Remarkably, this clinical signature made of only nine unannotated lncRNAs largely outperformed PCA3, the only used prostate cancer lncRNA biomarker, in detection of high-risk tumors. This modular workflow is highly sensitive and can be applied to any pathology or clinical application.


Subject(s)
Prostatic Neoplasms/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Biomarkers, Tumor/genetics , Cohort Studies , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Prostate/pathology , Prostatic Neoplasms/diagnosis , RNA, Long Noncoding/genetics , Retrospective Studies
15.
Nature ; 569(7758): 672-678, 2019 05.
Article in English | MEDLINE | ID: mdl-31092925

ABSTRACT

Autonomic nerve fibres in the tumour microenvironment regulate cancer initiation and dissemination, but how nerves emerge in tumours is currently unknown. Here we show that neural progenitors from the central nervous system that express doublecortin (DCX+) infiltrate prostate tumours and metastases, in which they initiate neurogenesis. In mouse models of prostate cancer, oscillations of DCX+ neural progenitors in the subventricular zone-a neurogenic area of the central nervous system-are associated with disruption of the blood-brain barrier, and with the egress of DCX+ cells into the circulation. These cells then infiltrate and reside in the tumour, and can generate new adrenergic neurons. Selective genetic depletion of DCX+ cells inhibits the early phases of tumour development in our mouse models of prostate cancer, whereas transplantation of DCX+ neural progenitors promotes tumour growth and metastasis. In humans, the density of DCX+ neural progenitors is strongly associated with the aggressiveness and recurrence of prostate adenocarcinoma. These results reveal a unique crosstalk between the central nervous system and prostate tumours, and indicate neural targets for the treatment of cancer.


Subject(s)
Central Nervous System/pathology , Neural Stem Cells/pathology , Neurogenesis , Prostatic Neoplasms/pathology , Adenocarcinoma/pathology , Adrenergic Neurons/pathology , Animals , Carcinogenesis , Cell Differentiation , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Genes, myc , Humans , Lateral Ventricles/pathology , Male , Mice , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/metabolism , Neuropeptides/metabolism , Olfactory Bulb/pathology , Prognosis
16.
Cancer Lett ; 410: 100-111, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28935391

ABSTRACT

Extracellular vesicles released from cancer cells may play an important role in cancer progression by shuttling oncogenic information into recipient cells. However, our knowledge is still fragmentary and there remain numerous questions regarding the mechanisms at play and the functional consequences of these interactions. We have recently established a mesenchymal-like prostate cancer cell line (22Rv1/CR-1; Mes-PCa). In this study, we assessed the effects of the extracellular vesicles released by these cells on recipient androgen-dependent epithelial VCaP prostate cancer cells. Mes-PCa derived vesicles were found to promote mesenchymal features in the recipient epithelial-like prostate cancer cells. This transformation was accompanied by a modulation of androgen receptor signaling and activation of TGFß signaling pathway. Moreover, recipient cells acquiring mesenchymal traits displayed enhanced migratory and invasive features as well as increased resistance to the androgen receptor antagonist, enzalutamide. Our results suggest a previously unappreciated role for Mes-PCa secreted vesicles in cancer promotion by transferring cell-mediated signals and promoting phenotypic changes in recipient prostate cancer cells.


Subject(s)
Cell-Derived Microparticles/metabolism , Epithelial-Mesenchymal Transition , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Androgen Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Cell Movement , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Humans , Male , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Phenotype , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Signal Transduction/drug effects , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment
17.
Oncotarget ; 8(6): 10050-10063, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28052023

ABSTRACT

The male germinal lineage, which is defined as unipotent, produces sperm through spermatogenesis. However, embryonic primordial germ cells and postnatal spermatogonial stem cells (SSCs) can change their fate and convert to pluripotency in culture when they are not controlled by the testicular microenvironment. The mechanisms underlying these reprogramming processes are poorly understood. Testicular germ cell tumors, including teratoma, share some molecular characteristics with pluripotent cells, suggesting that cancer could result from an abnormal differentiation of primordial germ cells or from an abnormal conversion of SCCs to pluripotency in the testis. Here, we investigated whether the somatic reprogramming factors Oct3/4, Sox2, Klf4 and c-Myc (OSKM) could play a role in SSCs reprogramming and induce pluripotency using a doxycycline-inducible transgenic Col1a1-4F2A-OSKM mouse model. We showed that, in contrast to somatic cells, SSCs from adult mice are resistant to this reprogramming strategy, even in combination with small molecules, hypoxia, or p53 deficiency, which were previously described to favour the conversion of somatic cells to pluripotency. This finding suggests that adult SSCs have developed specific mechanisms to repress reprogramming by OSKM factors, contributing to circumvent testicular cancer initiation events.


Subject(s)
Adult Germline Stem Cells/metabolism , Cellular Reprogramming Techniques , Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/metabolism , Adult Germline Stem Cells/drug effects , Animals , Cell Hypoxia , Cell Lineage , Cells, Cultured , Cellular Reprogramming/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Gene Expression Regulation, Developmental , Genotype , Induced Pluripotent Stem Cells/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/drug effects , Octamer Transcription Factor-3/genetics , Phenotype , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Sci Rep ; 6: 36906, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27883081

ABSTRACT

B-lymphocytes in the bone marrow (BM) must generate a functional B-cell receptor and overcome the negative selection induced by reactivity with autoantigens. Two rounds of DNA recombination are required for the production of functional immunoglobulin heavy (Ig-HCs) and light (LCs) chains necessary for the continuation of B-lymphocyte development in the BM. Both rounds depend on the joint action of recombination activating gene-1 (RAG-1) and RAG-2 endonucleases with the DNA non-homologous end-joining pathway. Loss of the FANC gene leads to the chromosome breakage and cancer predisposition syndrome Fanconi anemia. Because the FANC proteins are involved in certain aspects of the recombination process, we sought to determine the impact of the FANC pathway on the Ig diversification process using Fanca-/- mice. In this work we demonstrated that Fanca-/- animals have a mild B-cell differentiation defect characterized by a specific alteration of the IgM- to IgM+ transition of the B220low B-cell population. Pre-B cells from Fanca-/- mice show evidence of impaired kLC rearrangement at the level of the Vk-Jk junction. Furthermore, Fanca-/- mice showed a skewed Vκ gene usage during formation of the LCs Vk-Jk junctions. Therefore, the Fanca protein appears as a yet unidentified factor involved in the primary diversification of Ig.


Subject(s)
B-Lymphocytes/immunology , Fanconi Anemia Complementation Group A Protein/immunology , Precursor Cells, B-Lymphoid/immunology , V(D)J Recombination/immunology , Animals , DNA End-Joining Repair/immunology , DNA-Binding Proteins/immunology , Fanconi Anemia/immunology , Fanconi Anemia Complementation Group Proteins/immunology , Homeodomain Proteins/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Mice , Receptors, Antigen, B-Cell/immunology
19.
Neoplasia ; 15(7): 761-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23814488

ABSTRACT

Castration-resistant prostate cancers (CRPCs) that relapse after androgen deprivation therapies (ADTs) are responsible for the majority of mortalities from prostate cancer (PCa). While mechanisms enabling recurrent activity of androgen receptor (AR) are certainly involved in the development of CRPC, there may be factors that contribute to the process including acquired neuroendocrine (NE) cell-like behaviors working through alternate (non-AR) cell signaling systems or AR-dependent mechanisms. In this study, we explore the potential relationship between the AR axis and a novel putative marker of NE differentiation, the human male protocadherin-PC (PCDH-PC), in vitro and in human situations. We found evidence for an NE transdifferentiation process and PCDH-PC expression as an early-onset adaptive mechanism following ADT and elucidate AR as a key regulator of PCDH-PC expression. PCDH-PC overexpression, in turn, attenuates the ligand-dependent activity of the AR, enabling certain prostate tumor clones to assume a more NE phenotype and promoting their survival under diverse stress conditions. Acquisition of an NE phenotype by PCa cells positively correlated with resistance to cytotoxic agents including docetaxel, a taxane chemotherapy approved for the treatment of patients with metastatic CRPC. Furthermore, knockdown of PCDH-PC in cells that have undergone an NE transdifferentiation partially sensitized cells to docetaxel. Together, these results reveal a reciprocal regulation between the AR axis and PCDH-PC signals, observed both in vitro and in vivo, with potential implications in coordinating NE transdifferentiation processes and progression of PCa toward hormonal and chemoresistance.


Subject(s)
Cadherins/metabolism , Cell Transdifferentiation , Drug Resistance, Neoplasm , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgens/metabolism , Antineoplastic Agents/pharmacology , Cadherins/genetics , Cell Line, Tumor , Cell Transdifferentiation/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Ligands , Male , Phenotype , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcriptional Activation
20.
Cell Stem Cell ; 11(5): 585-6, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23122284

ABSTRACT

The feasibility of using spermatogonial stem cells (SSCs) for cell-based infertility treatment has suffered from a lack of evidence in a preclinical nonhuman primate model. In this issue, Hermann et al. (2012) demonstrate that autologous and allogeneic transplantation of SSCs in testes of rhesus macaques produced functional sperm.


Subject(s)
Spermatogonia/transplantation , Spermatozoa/physiology , Testis/transplantation , Animals , Male
SELECTION OF CITATIONS
SEARCH DETAIL