Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 109(4): 533-551, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37552049

ABSTRACT

Niche-derived growth factors support self-renewal of mouse spermatogonial stem and progenitor cells through ERK MAPK signaling and other pathways. At the same time, dysregulated growth factor-dependent signaling has been associated with loss of stem cell activity and aberrant differentiation. We hypothesized that growth factor signaling through the ERK MAPK pathway in spermatogonial stem cells is tightly regulated within a narrow range through distinct intracellular negative feedback regulators. Evaluation of candidate extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK)-responsive genes known to dampen downstream signaling revealed robust induction of specific negative feedback regulators, including Spry4, in cultured mouse spermatogonial stem cells in response to glial cell line-derived neurotrophic factor or fibroblast growth factor 2. Undifferentiated spermatogonia in vivo exhibited high levels of Spry4 mRNA. Quantitative single-cell analysis of ERK MAPK signaling in spermatogonial stem cell cultures revealed both dynamic signaling patterns in response to growth factors and disruption of such effects when Spry4 was ablated, due to dysregulation of ERK MAPK downstream of RAS. Whereas negative feedback regulator expression decreased during differentiation, loss of Spry4 shifted cell fate toward early differentiation with concomitant loss of stem cell activity. Finally, a mouse Spry4 reporter line revealed that the adult spermatogonial stem cell population in vivo is demarcated by strong Spry4 promoter activity. Collectively, our data suggest that negative feedback-dependent regulation of ERK MAPK is critical for preservation of spermatogonial stem cell fate within the mammalian testis.


Subject(s)
Adult Stem Cells , Extracellular Signal-Regulated MAP Kinases , Male , Mice , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback , Cell Differentiation/physiology , Spermatogonia/metabolism , Adult Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mammals/metabolism
2.
Methods Mol Biol ; 2656: 211-225, 2023.
Article in English | MEDLINE | ID: mdl-37249874

ABSTRACT

Lentiviral vectors have been major tools for genetic manipulation of spermatogonial stem cells (SSCs) in vitro. Adeno-associated viral vectors are promising emerging tools for in vivo SSC transduction that are less invasive, compared to lentivirus, since AAV DNA is not integrated into the host genome and the host genome remains intact. In this chapter, we describe protocols using lentiviral and adeno-associated viral vectors to transduce SSCs in vitro and vivo, respectively.


Subject(s)
Genetic Techniques , Mammals , Animals , Male , Mammals/genetics , Stem Cells , Genetic Vectors/genetics , Transduction, Genetic , Lentivirus/genetics , Spermatogonia
3.
Biol Reprod ; 102(4): 950-962, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31836902

ABSTRACT

Germline stem and progenitor cells can be extracted from the adult mouse testis and maintained long-term in vitro. Yet, the optimal culture conditions for preserving stem cell activity are unknown. Recently, multiple members of the Eph receptor family were detected in murine spermatogonia, but their roles remain obscure. One such gene, Ephb2, is crucial for maintenance of somatic stem cells and was previously found enriched at the level of mRNA in murine spermatogonia. We detected Ephb2 mRNA and protein in primary adult spermatogonial cultures and hypothesized that Ephb2 plays a role in maintenance of stem cells in vitro. We employed CRISPR-Cas9 targeting and generated stable mutant SSC lines with complete loss of Ephb2. The characteristics of Ephb2-KO cells were interrogated using phenotypic and functional assays. Ephb2-KO SSCs exhibited reduced proliferation compared to wild-type cells, while apoptosis was unaffected. Therefore, we examined whether Ephb2 loss correlates with activity of canonical pathways involved in stem cell self-renewal and proliferation. Ephb2-KO cells had reduced ERK MAPK signaling. Using a lentiviral transgene, Ephb2 expression was rescued in Ephb2-KO cells, which partially restored signaling and proliferation. Transplantation analysis revealed that Ephb2-KO SSCs cultures formed significantly fewer colonies than WT, indicating a role for Ephb2 in preserving stem cell activity of cultured cells. Transcriptome analysis of wild-type and Ephb2-KO SSCs identified Dppa4 and Bnc1 as differentially expressed, Ephb2-dependent genes that are potentially involved in stem cell function. These data uncover for the first time a crucial role for Ephb2 signaling in cultured SSCs.


Subject(s)
Adult Stem Cells/metabolism , Cell Proliferation/physiology , Receptor, EphB2/metabolism , Spermatogonia/metabolism , Adult Stem Cells/cytology , Animals , CRISPR-Cas Systems , Cell Line , Cells, Cultured , Male , Mice , Mice, Knockout , Receptor, EphB2/genetics , Signal Transduction/physiology , Spermatogenesis/physiology , Spermatogonia/cytology
4.
PLoS Genet ; 15(5): e1008139, 2019 05.
Article in English | MEDLINE | ID: mdl-31050682

ABSTRACT

Accumulating evidence indicates that paternal age correlates with disease risk in children. De novo gain-of-function mutations in the FGF-RAS-MAPK signaling pathway are known to cause a subset of genetic diseases associated with advanced paternal age, such as Apert syndrome, achondroplasia, Noonan syndrome, and Costello syndrome. It has been hypothesized that adult spermatogonial stem cells with pathogenic mutations are clonally expanded over time and propagate the mutations to offspring. However, no model system exists to interrogate mammalian germline stem cell competition in vivo. In this study, we created a lineage tracing system, which enabled undifferentiated spermatogonia with endogenous expression of HrasG12V, a known pathogenic gain-of-function mutation in RAS-MAPK signaling, to compete with their wild-type counterparts in the mouse testis. Over a year of fate analysis, neither HrasG12V-positive germ cells nor sperm exhibited a significant expansion compared to wild-type neighbors. Short-term stem cell capacity as measured by transplantation analysis was also comparable between wild-type and mutant groups. Furthermore, although constitutively active HRAS was detectable in the mutant cell lines, they did not exhibit a proliferative advantage or an enhanced response to agonist-evoked pERK signaling. These in vivo and in vitro results suggest that mouse spermatogonial stem cells are functionally resistant to a heterozygous HrasG12V mutation in the endogenous locus and that mechanisms could exist to prevent such harmful mutations from being expanded and transmitted to the next generation.


Subject(s)
Adult Germline Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Spermatogonia/metabolism , Adult Germline Stem Cells/physiology , Animals , Gain of Function Mutation/genetics , Germ-Line Mutation/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mutation/genetics , Paternal Age , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Selection, Genetic/genetics , Signal Transduction/genetics , Spermatogonia/physiology , Spermatozoa/metabolism , Testis/metabolism
5.
Endocr Relat Cancer ; 26(4): 411-423, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30689542

ABSTRACT

Loss of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) expression by CpG promoter hypermethylation is associated with metastasis in gastroenteropancreatic neuroendocrine tumors; however, the mechanism of how UCHL1 loss contributes to metastatic potential remains unclear. In this study, we first confirmed that loss of UCHL1 expression on immunohistochemistry was significantly associated with metastatic tumors in a translational pancreatic neuroendocrine tumor (PNET) cohort, with a sensitivity and specificity of 78% and 89%, respectively. To study the mechanism driving this aggressive phenotype, BON and QGP-1 metastatic PNET cell lines, which do not produce UCHL1, were stably transfected to re-express UCHL1. In vitro assays, RNA-sequencing, and reverse-phase protein array (RPPA) analyses were performed comparing empty-vector negative controls and UCHL1-expressing cell lines. UCHL1 re-expression is associated with lower anchorage-independent colony growth in BON cells, lower colony formation in QGP cells, and a higher percentage of cells in the G0/G1 cell-cycle phase in BON and QGP cells. On RPPA proteomic analysis, there was an upregulation of cell-cycle regulatory proteins CHK2 (1.2 fold change, p=0.004) and P21 (1.2 fold change, p=0.023) in BON cells expressing UCHL1; western blot confirmed upregulation of phosphorylated CHK2 and P21. There were no transcriptomic differences detected on RNA-Sequencing between empty-vector negative controls and UCHL1-expressing cell lines. In conclusion, UCHL1 loss correlates with metastatic potential in PNETs and its re-expression induces a less aggressive phenotype in vitro, in part by inducing cell-cycle arrest through post-translational regulation of phosphorylated CHK2. UCHL1 re-expression should be considered as a functional biomarker in detecting PNETs capable of metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Middle Aged , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Phenotype , Ubiquitin Thiolesterase/genetics
6.
Nat Commun ; 9(1): 2090, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844387

ABSTRACT

Evidence of male-to-female sexual transmission of Zika virus (ZIKV) and viral RNA in semen and sperm months after infection supports a potential role for testicular cells in ZIKV propagation. Here, we demonstrate that germ cells (GCs) are most susceptible to ZIKV. We found that only GCs infected by ZIKV, but not those infected by dengue virus and yellow fever virus, produce high levels of infectious virus. This observation coincides with decreased expression of interferon-stimulated gene Ifi44l in ZIKV-infected GCs, and overexpression of Ifi44l results in reduced ZIKV production. Using primary human testicular tissue, we demonstrate that human GCs are also permissive for ZIKV infection and production. Finally, we identified berberine chloride as a potent inhibitor of ZIKV infection in both murine and human testes. Together, these studies identify a potential cellular source for propagation of ZIKV in testes and a candidate drug for preventing sexual transmission of ZIKV.


Subject(s)
Antiviral Agents/pharmacology , Berberine/pharmacology , RNA, Viral/analysis , Sexually Transmitted Diseases, Viral/prevention & control , Spermatozoa/virology , Testis/virology , Virus Replication/drug effects , Zika Virus Infection/transmission , Zika Virus/growth & development , Animals , Antigens/biosynthesis , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Cytoskeletal Proteins/biosynthesis , Dengue Virus/growth & development , Humans , Interferon Type I/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Viral/isolation & purification , Receptor, Interferon alpha-beta/genetics , Sexually Transmitted Diseases, Viral/virology , Testis/cytology , Vero Cells , Virus Replication/physiology , Yellow fever virus/growth & development , Zika Virus/isolation & purification , Zika Virus Infection/virology
7.
Stem Cells Dev ; 25(20): 1483-1494, 2016 10.
Article in English | MEDLINE | ID: mdl-27596369

ABSTRACT

Spermatogonial stem cells (SSCs) propagate mammalian spermatogenesis throughout male reproductive life by continuously self-renewing and differentiating, ultimately, into sperm. SSCs can be cultured for long periods and restore spermatogenesis upon transplantation back into the native microenvironment in vivo. Conventionally, SSC research has been focused mainly on male infertility and, to a lesser extent, on cell reprogramming. With the advent of genome-wide sequencing technology, however, human studies have uncovered a wide range of pathogenic alleles that arise in the male germline. A subset of de novo point mutations (DNMs) was shown to originate in SSCs and cause congenital disorders in children. This review describes both monogenic diseases (e.g., Apert syndrome) and complex disorders that are either known or suspected to be driven by mutations in SSCs. We propose that SSC culture is a suitable model for studying the origin and mechanisms of these diseases. Lastly, we discuss strategies for future clinical implementation of SSC-based technology, from detecting mutation burden by sperm screening to gene correction in vitro.

8.
Stem Cell Reports ; 6(5): 772-783, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27132888

ABSTRACT

Human male germ cell tumors (GCTs) are derived from primordial germ cells (PGCs). The master pluripotency regulator and neuroectodermal lineage effector transcription factor SOX2 is repressed in PGCs and the seminoma (SEM) subset of GCTs. The mechanism of SOX2 repression and its significance to GC and GCT development currently are not understood. Here, we show that SOX2 repression in SEM-derived TCam-2 cells is mediated by the Polycomb repressive complex (PcG) and the repressive H3K27me3 chromatin mark that are enriched at its promoter. Furthermore, SOX2 repression in TCam-2 cells can be abrogated by recruitment of the constitutively expressed H3K27 demethylase UTX to the SOX2 promoter through retinoid signaling, leading to expression of neuronal and other lineage genes. SOX17 has been shown to initiate human PGC specification, with its target PRDM1 suppressing mesendodermal genes. Our results are consistent with a role for SOX2 repression in normal germline development by suppressing neuroectodermal genes.


Subject(s)
Neoplasms, Germ Cell and Embryonal/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , SOXB1 Transcription Factors/genetics , SOXF Transcription Factors/genetics , Seminoma/genetics , Testicular Neoplasms/genetics , Cell Lineage/genetics , Chromatin/genetics , Gene Expression Regulation, Neoplastic , Germ Cells/pathology , Histone Demethylases/genetics , Humans , Male , Neoplasms, Germ Cell and Embryonal/pathology , Nuclear Proteins/genetics , Polycomb-Group Proteins/genetics , Promoter Regions, Genetic , Seminoma/pathology , Testicular Neoplasms/pathology
9.
Nat Commun ; 7: 11275, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27117588

ABSTRACT

Spermatogonial stem and progenitor cells (SSCs) generate adult male gametes. During in vitro expansion, these unipotent murine cells spontaneously convert to multipotent adult spermatogonial-derived stem cells (MASCs). Here we investigate this conversion process through integrative transcriptomic and epigenomic analyses. We find in SSCs that promoters essential to maintenance and differentiation of embryonic stem cells (ESCs) are enriched with histone H3-lysine4 and -lysine 27 trimethylations. These bivalent modifications are maintained at most somatic promoters after conversion, bestowing MASCs an ESC-like promoter chromatin. At enhancers, the core pluripotency circuitry is activated partially in SSCs and completely in MASCs, concomitant with loss of germ cell-specific gene expression and initiation of embryonic-like programs. Furthermore, SSCs in vitro maintain the epigenomic characteristics of germ cells in vivo. Our observations suggest that SSCs encode innate plasticity through the epigenome and that both conversion of promoter chromatin states and activation of cell type-specific enhancers are prominent features of reprogramming.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , Embryonic Stem Cells/metabolism , Epigenomics/methods , Multipotent Stem Cells/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Histones/metabolism , Lysine/metabolism , Male , Methylation , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Spermatogenesis/genetics , Spermatogonia/cytology
10.
Schizophr Res ; 168(1-2): 421-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215504

ABSTRACT

Multiple lines of evidence corroborate impaired signaling pathways as relevant to the underpinnings of schizophrenia. There has been an interest in neurotrophins, since they are crucial mediators of neurodevelopment and in synaptic connectivity in the adult brain. Neurotrophins and their receptors demonstrate aberrant expression patterns in cortical areas for schizophrenia cases in comparison to control subjects. There is little known about the contribution of neurotrophin genes in psychiatric disorders. To begin to address this issue, we conducted high-coverage targeted exome capture in a subset of neurotrophin genes in 48 comprehensively characterized cases with schizophrenia-related psychosis. We herein report rare missense polymorphisms and novel missense mutations in neurotrophin receptor signaling pathway genes. Furthermore, we observed that several genes have a higher propensity to harbor missense coding variants than others. Based on this initial analysis we suggest that rare variants and missense mutations in neurotrophin genes might represent genetic contributions involved across psychiatric disorders.


Subject(s)
Nerve Growth Factors/genetics , Polymorphism, Genetic/genetics , Schizophrenia/pathology , Signal Transduction/genetics , Adolescent , Adult , Analysis of Variance , Depression/etiology , Female , Humans , Intelligence/genetics , Male , Middle Aged , Models, Molecular , Nerve Growth Factors/metabolism , Psychiatric Status Rating Scales , Schizophrenia/complications , Young Adult
11.
Schizophr Res ; 166(1-3): 119-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26091878

ABSTRACT

Schizophrenia is a debilitating syndrome with high heritability. Genomic studies reveal more than a hundred genetic variants, largely nonspecific and of small effect size, and not accounting for its high heritability. De novo mutations are one mechanism whereby disease related alleles may be introduced into the population, although these have not been leveraged to explore the disease in general samples. This paper describes a framework to find high impact genes for schizophrenia. This study consists of two different datasets. First, whole exome sequencing was conducted to identify disruptive de novo mutations in 14 complete parent-offspring trios with sporadic schizophrenia from Jerusalem, which identified 5 sporadic cases with de novo gene mutations in 5 different genes (PTPRG, TGM5, SLC39A13, BTK, CDKN3). Next, targeted exome capture of these genes was conducted in 48 well-characterized, unrelated, ethnically diverse schizophrenia cases, recruited and characterized by the same research team in New York (NY sample), which demonstrated extremely rare and potentially damaging variants in three of the five genes (MAF<0.01) in 12/48 cases (25%); including PTPRG (5 cases), SCL39A13 (4 cases) and TGM5 (4 cases), a higher number than usually identified by whole exome sequencing. Cases differed in cognition and illness features based on which mutation-enriched gene they carried. Functional de novo mutations in protein-interaction domains in sporadic schizophrenia can illuminate risk genes that increase the propensity to develop schizophrenia across ethnicities.


Subject(s)
Mutation , Schizophrenia/genetics , Adult , Datasets as Topic , Female , Genetic Predisposition to Disease , Humans , Israel , Male , New York , Parents , Psychotic Disorders/ethnology , Psychotic Disorders/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Schizophrenia/ethnology , Sequence Analysis, DNA
12.
Stem Cell Reports ; 3(2): 219-26, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25254335

ABSTRACT

Pathogenic de novo mutations increase with fathers' age and could be amplified through competition between genetically distinct subpopulations of spermatogonial stem cells (SSCs). Here, we tested the fitness of SSCs bearing wild-type human FGFR2 or an Apert syndrome mutant, FGFR2 (S252W), to provide experimental evidence for SSC competition. The S252W allele conferred enhanced FGFR2-mediated signaling, particularly at very low concentrations of ligand, and also subtle changes in gene expression. Mutant SSCs exhibited improved competitiveness in vitro and increased stem cell activity in vivo upon transplantation. The fitness advantage in vitro only occurred in low concentrations of fibroblast growth factor (FGF), was independent of FGF-driven proliferation, and was accompanied by increased response to glial cell line-derived neurotrophic factor (GDNF). Our studies provide experimental evidence of enhanced stem cell fitness in SSCs bearing a paternal age-associated mutation. Our model will be useful for interrogating other candidate mutations in the future to reveal mechanisms of disease risk.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2/genetics , Stem Cells/cytology , Alleles , Animals , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Paternal Age , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Spermatogonia/cytology , Stem Cell Transplantation , Testis/metabolism
13.
Ann Surg Oncol ; 21 Suppl 4: S672-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24854489

ABSTRACT

BACKGROUND: Well-differentiated gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare tumors with varying metastatic potential. The underlying molecular basis for metastasis by GEP-NETs remains undefined. METHODS: Quantitative PCR and immunohistochemistry (IHC) staining for ubiquitin carboxyl-terminal esterase L1 (UCHL1) gene and protein expression was performed on a group of localized and metastatic well-differentiated GEP-NET samples acquired from a prospectively maintained tissue bank. The ability of extent of UCHL1 IHC staining to differentiate localized and metastatic tumors was compared with Ki-67 index. RESULTS: Among 46 total samples, UCHL1 expression at both the gene and protein level was significantly greater among localized GEP-NETs compared with metastatic tumors and metastases (p < 0.001). Hypermethylation of the UCHL1 promoter was commonly observed among metastatic primary tumors and metastases (those with the lowest UCHL1 expression) but not among localized tumors (p < 0.001). Poor staining (<50 %) for UCHL1 was observed in 27 % of localized tumors compared with 87 % of metastatic tumors (p = 0.001). The presence of <50 % staining for UCHL1 was 88 % sensitive and 73 % specific for identifying metastatic disease. In contrast, there was no association between Ki-67 index and metastatic disease. In multivariable analysis, only UCHL1 staining <50 % [odds ratio (OR) 24.5, p = 0.035] and vascular invasion (OR 38.4, p = 0.03) were independent risk factors for metastatic disease at the time of initial surgery. CONCLUSIONS: Loss of UCHL1 expression by CpG promoter hypermethylation is associated with metastatic GEP-NETs. Extent of UCHL1 staining should be explored as a potentially clinically useful adjunct to Ki-67 index in evaluating GEP-NETs for aggressive features.


Subject(s)
Carcinoid Tumor/genetics , Carcinoid Tumor/secondary , CpG Islands/genetics , Digestive System Neoplasms/genetics , Digestive System Neoplasms/pathology , Ubiquitin Thiolesterase/genetics , Adult , Aged , Blood Vessels/pathology , Carcinoid Tumor/chemistry , Digestive System Neoplasms/chemistry , Female , Gene Silencing , Humans , Ki-67 Antigen/analysis , Male , Methylation , Middle Aged , Mitotic Index , Neoplasm Invasiveness , Neoplasm Metastasis , Promoter Regions, Genetic , Risk Factors , Sensitivity and Specificity , Ubiquitin Thiolesterase/analysis
14.
J Vis Exp ; (72): e50017, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23462452

ABSTRACT

Spermatogonial stem and progenitor cells (SSCs) of the testis represent a classic example of adult mammalian stem cells and preserve fertility for nearly the lifetime of the animal. While the precise mechanisms that govern self-renewal and differentiation in vivo are challenging to study, various systems have been developed previously to propagate murine SSCs in vitro using a combination of specialized culture media and feeder cells(1-3). Most in vitro forays into the biology of SSCs have derived cell lines from neonates, possibly due to the difficulty in obtaining adult cell lines(4). However, the testis continues to mature up until ~5 weeks of age in most mouse strains. In the early post-natal period, dramatic changes occur in the architecture of the testis and in the biology of both somatic and spermatogenic cells, including alterations in expression levels of numerous stem cell-related genes. Therefore, neonatally-derived SSC lines may not fully recapitulate the biology of adult SSCs that persist after the adult testis has reached a steady state. Several factors have hindered the production of adult SSC lines historically. First, the proportion of functional stem cells may decrease during adulthood, either due to intrinsic or extrinsic factors(5,6). Furthermore, as with other adult stem cells, it has been difficult to enrich SSCs sufficiently from total adult testicular cells without using a combination of immunoselection or other sorting strategies(7). Commonly employed strategies include the use of cryptorchid mice as a source of donor cells due to a higher ratio of stem cells to other cell types(8). Based on the hypothesis that removal of somatic cells from the initial culture disrupts interactions with the stem cell niche that are essential for SSC survival, we previously developed methods to derive adult lines that do not require immunoselection or cryptorchid donors but rather employ serial enrichment of SSCs in culture, referred to hereafter as SESC(2,3). The method described below entails a simple procedure for deriving adult SSC lines by dissociating adult donor seminiferous tubules, followed by plating of cells on feeders comprised of a testicular stromal cell line (JK1)(3). Through serial passaging, strongly adherent, contaminating non-germ cells are depleted from the culture with concomitant enrichment of SSCs. Cultures produced in this manner contain a mixture of spermatogonia at different stages of differentiation, which contain SSCs, based on long-term self renewal capability. The crux of the SESC method is that it enables SSCs to make the difficult transition from self-renewal in vivo to long-term self-renewal in vitro in a radically different microenvironment, produces long-term SSC lines, free of contaminating somatic cells, and thereby enables subsequent experimental manipulation of SSCs.


Subject(s)
Cytological Techniques/methods , Spermatogonia/cytology , Stem Cells/cytology , Animals , Cell Line , Cells, Cultured , Male , Mice
15.
Biomed Res Int ; 2013: 384734, 2013.
Article in English | MEDLINE | ID: mdl-23484114

ABSTRACT

Adult spermatogonial stem cells (SSCs) represent a distinctive source of stem cells in mammals for several reasons. First, by giving rise to spermatogenesis, SSCs are responsible for the propagation of a father's genetic material. As such, autologous SSCs have been considered for treatment of infertility and other purposes, including correction of inherited disorders. Second, adult spermatogonia can spontaneously produce embryonic-like stem cells in vitro, which could be used as an alternative for therapeutic, diagnostic, or drug discovery strategies for humans. Therefore, an increasing urgency is driving efforts to understand the biology of SSCs and improve techniques to manipulate them in vitro as a prerequisite to achieve the aforementioned goals. The characterization of adult SSCs also requires reproducible methods to isolate and maintain them in long-term culture. Herein, we describe recent major advances and challenges in propagation of adult SSCs from mice and humans during the past few years, including the use of unique cell surface markers and defined cultured conditions.


Subject(s)
Adult Stem Cells/cytology , Cell Culture Techniques/methods , Infertility, Male/therapy , Spermatogonia/cytology , Stem Cell Transplantation , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Animals , Humans , Male , Mice , Spermatogonia/metabolism , Transplantation, Autologous
16.
PLoS One ; 8(12): e85143, 2013.
Article in English | MEDLINE | ID: mdl-24386459

ABSTRACT

Hematogenous metastasis accounts for the majority of cancer-related deaths, yet the mechanism remains unclear. Circulating tumor cells (CTCs) in blood may employ different pathways to cross blood endothelial barrier and establish a metastatic niche. Several studies provide evidence that prostate cancer (PCa) cell tethering and rolling on microvascular endothelium via E-selectin/E-selectin ligand interactions under shear flow theoretically promote extravasation and contribute to the development of metastases. However, it is unknown if CTCs from PCa patients interact with E-selectin expressed on endothelium, initiating a route for tumor metastases. Here we report that CTCs derived from PCa patients showed interactions with E-selectin and E-selectin expressing endothelial cells. To examine E-selectin-mediated interactions of PCa cell lines and CTCs derived from metastatic PCa patients, we used fluorescently-labeled anti-prostate specific membrane antigen (PSMA) monoclonal antibody J591-488 which is internalized following cell-surface binding. We employed a microscale flow device consisting of E-selectin-coated microtubes and human umbilical vein endothelial cells (HUVECs) on parallel-plate flow chamber simulating vascular endothelium. We observed that J591-488 did not significantly alter the rolling behavior in PCa cells at shear stresses below 3 dyn/cm(2). CTCs obtained from 31 PCa patient samples showed that CTCs tether and stably interact with E-selectin and E-selectin expressing HUVECs at physiological shear stress. Interestingly, samples collected during disease progression demonstrated significantly more CTC/E-selectin interactions than samples during times of therapeutic response (p=0.016). Analysis of the expression of sialyl Lewis X (sLe(x)) in patient samples showed that a small subset comprising 1.9-18.8% of CTCs possess high sLe(x) expression. Furthermore, E-selectin-mediated interactions between prostate CTCs and HUVECs were diminished in the presence of anti-E-selectin neutralizing antibody. CTC-Endothelial interactions provide a novel insight into potential adhesive mechanisms of prostate CTCs as a means to initiate metastasis.


Subject(s)
E-Selectin/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neoplasm Proteins/metabolism , Neoplastic Cells, Circulating/metabolism , Prostatic Neoplasms/metabolism , Stress, Physiological , Blood Flow Velocity , Cell Line , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/pathology
17.
Cell Stem Cell ; 9(4): 317-29, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21982232

ABSTRACT

The transcription factor Sox2 maintains the pluripotency of early embryonic cells and regulates the formation of several epithelia during fetal development. Whether Sox2 continues to play a role in adult tissues remains largely unknown. We show here that Sox2 marks adult cells in several epithelial tissues where its expression has not previously been characterized, including the stomach, cervix, anus, testes, lens, and multiple glands. Genetic lineage tracing and transplantation experiments demonstrate that Sox2-expressing cells continuously give rise to mature cell types within these tissues, documenting their self-renewal and differentiation potentials. Consistent with these findings, ablation of Sox2(+) cells in mice results in a disruption of epithelial tissue homeostasis and lethality. Developmental fate mapping reveals that Sox2(+) adult stem cells originate from fetal Sox2(+) tissue progenitors. Thus, our results identify Sox2 expression in numerous adult endodermal and ectodermal stem cell compartments, which are critical for normal tissue regeneration and survival.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Regeneration , SOXB1 Transcription Factors/metabolism , Adult Stem Cells/drug effects , Animals , Cell Compartmentation , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Ganciclovir/pharmacology , Green Fluorescent Proteins/metabolism , Homeostasis/drug effects , Infertility, Male/pathology , Male , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Recombinant Fusion Proteins/metabolism , Regeneration/drug effects , Spermatogenesis/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology , Stomach/cytology , Survival Analysis , Testis/drug effects , Testis/pathology
19.
Cell ; 142(3): 468-79, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20691905

ABSTRACT

Hyperactivity of mTORC1, a key mediator of cell growth, leads to stem cell depletion, although the underlying mechanisms are poorly defined. Using spermatogonial progenitor cells (SPCs) as a model system, we show that mTORC1 impairs stem cell maintenance by a negative feedback from mTORC1 to receptors required to transduce niche-derived signals. We find that SPCs lacking Plzf, a transcription factor essential for SPC maintenance, have enhanced mTORC1 activity. Aberrant mTORC1 activation in Plzf(-/-) SPCs inhibits their response to GDNF, a growth factor critical for SPC self-renewal, via negative feedback at the level of the GDNF receptor. Plzf opposes mTORC1 activity by inducing expression of the mTORC1 inhibitor Redd1. Thus, we identify the mTORC1-Plzf functional interaction as a critical rheostat for maintenance of the spermatogonial pool and propose a model whereby negative feedback from mTORC1 to the GDNF receptor balances SPC growth with self-renewal.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Spermatogonia/cytology , Stem Cells/cytology , Transcription Factors/metabolism , Animals , Feedback, Physiological , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Kruppel-Like Transcription Factors/genetics , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Promyelocytic Leukemia Zinc Finger Protein , Proteins , Signal Transduction , Spermatogonia/metabolism , Stem Cells/metabolism , TOR Serine-Threonine Kinases , Testis/cytology
20.
Cell Stem Cell ; 6(3): 251-64, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20207228

ABSTRACT

Bone marrow endothelial cells (ECs) are essential for reconstitution of hematopoiesis, but their role in self-renewal of long-term hematopoietic stem cells (LT-HSCs) is unknown. We have developed angiogenic models to demonstrate that EC-derived angiocrine growth factors support in vitro self-renewal and in vivo repopulation of authentic LT-HSCs. In serum/cytokine-free cocultures, ECs, through direct cellular contact, stimulated incremental expansion of repopulating CD34(-)Flt3(-)cKit(+)Lineage(-)Sca1(+) LT-HSCs, which retained their self-renewal ability, as determined by single-cell and serial transplantation assays. Angiocrine expression of Notch ligands by ECs promoted proliferation and prevented exhaustion of LT-HSCs derived from wild-type, but not Notch1/Notch2-deficient, mice. In transgenic notch-reporter (TNR.Gfp) mice, regenerating TNR.Gfp(+) LT-HSCs were detected in cellular contact with sinusoidal ECs. Interference with angiocrine, but not perfusion, function of SECs impaired repopulation of TNR.Gfp(+) LT-HSCs. ECs establish an instructive vascular niche for clinical-scale expansion of LT-HSCs and a cellular platform to identify stem cell-active trophogens.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Signal Transduction , Animals , Cell Communication , Cell Lineage , Cell Proliferation , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Ligands , Mice , Mice, Knockout , Receptor, Notch1/deficiency , Receptor, Notch1/metabolism , Receptor, Notch2/deficiency , Receptor, Notch2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...