ID: 22: DIFFERENTIAL ACTIONS OF ESTROGEN RECEPTOR α AND β VIA NON-GENOMIC SIGNALING IN HUMAN PROSTATE STEM-PROGENITOR CELLS

2016 ◽  
Vol 64 (4) ◽  
pp. 933-933
Author(s):  
S Majumdar ◽  
JC Rinaldi ◽  
T Gauntner ◽  
L Xie ◽  
W Hu ◽  
...  

Genomic signaling via estrogen receptors (ER) has been widely studied and implicated as the main ER signaling pathway in prostate development and carcinogenesis. Non-genomic ER signaling has also been reported in prostate epithelium although down-stream cascades have not been clarified. Our lab has recently identified ERs in human prostate epithelial stem/progenitor cells and shown that that 17β-estradiol (E2) can stimulate stem cell symmetric self-renewal and progenitor cell proliferation. In this study we interrogate non-genomic membrane initiated ER signaling in this prostate stem/progenitor cell population. Human prostate stem-progenitor cells were enriched from primary prostate epithelial cell cultures (PrEC) of young, disease-free donors using a 3D prostasphere (PS) model as previously described. Cells were labeled using ERα or ERβ antibodies along with prostate stem cell markers CD49f and TROP2 followed by triple channel FACS to quantify ERα+/ERβ+ cell numbers. To explore ERα, the benign human prostate stem cell line WPE with extremely low levels of ERα and ERβ, was stably transfected with a lentiviral-ERα expression vector. The human prostate cancer stem-like cell line HuSLC (ERβ++, ERα−) was utilized to interrogate ERb actions. Cells were exposed to 10 nM estradiol (E2) over a 15 to 60 minute time course +/− ICI 182,870 (ICI), an ERα/β antagonist. FACS analysis of day 7 PS cells labeled for ERα or ERβ revealed 66% of day 7 PS cells as ERα+ and 40% as ERβ+. Among ERα or ERβ positive PS cells, 4% were Trop2+/CD49fhigh (stem-like cells) and 10–12% were Trop2+/CD49fmedium (early stage progenitor cells). PS exposed to 10 nM E2 showed sequential phosphorylation of Src, Erk1/2, p38, Akt and NFκB (p65) over 60 minutes. Phosphorylation of up-and downstream targets (EGFR, Jnk, GSK 3α/β, p70 S6 kinase, PRAS40, MSK1/2) was also seen using a phospho-kinase array. Furthermore, phosphorylation of ERα at S167 was noted over 60 min of E2 exposure enabling enhancement of genomic ERα transactivational activity in a feed-forward manner. ICI attenuated Akt and Erk1/2 phosphorylation, confirming membrane bound ERs are involved in downstream signaling. E2 treatment of HuSLCs showed phosphorylation of Erk1/2 but not Akt, indicating that ERβ signals exclusively through the MAPK pathway in these cells. Conversely, E2 treatment of WPE-stem cells overexpressing ERα resulted in robust phosphorylation of Akt but lower levels of Erk1/2 phosphorylation suggesting that Akt activation may be more reliant on ERα signaling. To identify pathway specific roles, specific inhibitors were added to PS cultures. PS treated with LY294002 (Akt inhibitor) for 7 days attenuated the E2-mediated increase in PS number and size. Inhibition of the NFκB downstream of the Akt pathway by IKK VII (IKK inhibitor) blocked p65 phosphorylation, abrogated the E2-induced increase in stem cell symmetric self-renewal and blunted E2 stimulation of progenitor cell proliferation. Analysis of PS cyclin mRNA levels revealed a G1 arrest of progenitor cells upon IKK inhibition suggesting an essential role of NFκB in progenitor cell amplification. MAPK pathway inhibition with U0126(Erk1/2 inhibitor) resulted in an attenuation of the E2-mediated increase in PS number and size and an increase stem cell symmetric self-renewal suggesting that MAPK pathway activation promotes commitment to stem and progenitor cell expansion. Taken together, the present findings reveal that human prostate stem-progenitor cells express both ERα and ERβ which differentially activate different signaling cascades originating at the membrane. These signaling events may lead to unique downstream actions that influence prostate stem-progenitor cell proliferation as well as lineage commitment decisions.

Blood ◽  
1999 ◽  
Vol 93 (8) ◽  
pp. 2569-2577 ◽  
Author(s):  
Huei-Mei Huang ◽  
Jian-Chiuan Li ◽  
Yueh-Chun Hsieh ◽  
Hsin-Fang Yang-Yen ◽  
Jeffrey Jong-Young Yen

Abstract In vitro proliferation of hematopoietic stem cells requires costimulation by multiple regulatory factors whereas expansion of lineage-committed progenitor cells generated by stem cells usually requires only a single factor. The distinct requirement of factors for proliferation coincides with the differential temporal expression of the subunits of cytokine receptors during early stem cell differentiation. In this study, we explored the underlying mechanism of the requirement of costimulation in a hematopoietic progenitor cell line TF-1. We found that granulocyte-macrophage colony-stimulating factor (GM-CSF) optimally activated proliferation of TF-1 cells regardless of the presence or absence of stem cell factor (SCF). However, interleukin-5 (IL-5) alone sustained survival of TF-1 cells and required costimulation of SCF for optimal proliferation. The synergistic effect of SCF was partly due to its anti-apoptosis activity. Overexpression of the IL-5 receptor  subunit (IL5R) in TF-1 cells by genetic selection or retroviral infection also resumed optimal proliferation due to correction of the defect in apoptosis suppression. Exogenous expression of an oncogenic anti-apoptosis protein, Bcl-2, conferred on TF-1 cells an IL-5–dependent phenotype. In summary, our data suggested SCF costimulation is only necessary when the expression level of IL5R is low and apoptosis suppression is defective in the signal transduction of IL-5. Expression of Bcl-2 proteins released the growth restriction of the progenitor cells and may be implicated in leukemia formation.


Blood ◽  
1999 ◽  
Vol 93 (8) ◽  
pp. 2569-2577 ◽  
Author(s):  
Huei-Mei Huang ◽  
Jian-Chiuan Li ◽  
Yueh-Chun Hsieh ◽  
Hsin-Fang Yang-Yen ◽  
Jeffrey Jong-Young Yen

In vitro proliferation of hematopoietic stem cells requires costimulation by multiple regulatory factors whereas expansion of lineage-committed progenitor cells generated by stem cells usually requires only a single factor. The distinct requirement of factors for proliferation coincides with the differential temporal expression of the subunits of cytokine receptors during early stem cell differentiation. In this study, we explored the underlying mechanism of the requirement of costimulation in a hematopoietic progenitor cell line TF-1. We found that granulocyte-macrophage colony-stimulating factor (GM-CSF) optimally activated proliferation of TF-1 cells regardless of the presence or absence of stem cell factor (SCF). However, interleukin-5 (IL-5) alone sustained survival of TF-1 cells and required costimulation of SCF for optimal proliferation. The synergistic effect of SCF was partly due to its anti-apoptosis activity. Overexpression of the IL-5 receptor  subunit (IL5R) in TF-1 cells by genetic selection or retroviral infection also resumed optimal proliferation due to correction of the defect in apoptosis suppression. Exogenous expression of an oncogenic anti-apoptosis protein, Bcl-2, conferred on TF-1 cells an IL-5–dependent phenotype. In summary, our data suggested SCF costimulation is only necessary when the expression level of IL5R is low and apoptosis suppression is defective in the signal transduction of IL-5. Expression of Bcl-2 proteins released the growth restriction of the progenitor cells and may be implicated in leukemia formation.


2016 ◽  
Vol 64 (4) ◽  
pp. 928.1-928 ◽  
Author(s):  
D Hu ◽  
W Hu ◽  
S Majumdar ◽  
T Gauntner ◽  
Y Li ◽  
...  

Estrogens are implicated in prostate development and cancer, while stem cells are essential in tissue homeostasis and carcinogenesis. We have previously demonstrated that estradiol-17β (E2) treatment augments prostaspheres (PS) number and size, implicating them as direct estrogen targets. The present studies sought to elucidate specific roles for ERα and ERβ in prostate stem and progenitor cells.Prostate stem-progenitor cells were identified and isolated from normal primary prostate epithelial cells (PrEC) using long term BrdU retention in 3-D PS culture. FACS analyses (BrdU/ERα or ERβ) showed prostate stem and progenitor populations were both ERα+ and ERβ+. BrdU-retaining stem cells expressed high levels of ERβ and lower ERα as compared to non-label-retaining progenitor cells, suggesting ERβ dominance in the prostate stem cell. Estradiol increased BrdU-retaining cell numbers by enhancing stem cell self-renewal through symmetric division. While ERα siRNA blocked the E2-stimulated BrdU-retaining cells, ERβ knockdown augmented the E2-induced increase of BrdU-retaining cells. Together these findings suggest that ERα stimulates whereas ERβ suppresses stem cell self-renew. This conclusion is supported by separate studies on 2-D cultured PrEC with FACS stem-like cell side-population analysis using selective ER antagonists and siRNA. Although ERβ siRNA did not influence ERα mRNA levels, ERα siRNA doubled ERβ expression suggesting a suppressive role of ERα on ERβ action.In total, the present findings identify distinct localization patterns and roles for ERα and ERβ in human prostate stem-like and daughter progenitor cells with ERα driving self-renewal and ERβ braking division. We propose that a delicate balance between ERα and ERβ contributes to prostate stem cell niche homeostasis and that their dysregulation may contribute to prostate carcinogenesis and progression.


2020 ◽  
Author(s):  
Sayantanee Biswas ◽  
Michelle R. Emond ◽  
Kurtis Chenoweth ◽  
James D. Jontes

AbstractThe proliferation of neural progenitor cells provides the cellular substrate from which the nervous system is sculpted during development. The δ-protocadherin family of homophilic cell adhesion molecules is essential for the normal development of the nervous system and has been linked to an array of neurodevelopmental disorders. However, the biological functions of δ-protocadherins are not well-defined. Here, we show that the δ-protocadherins regulate proliferation in neural progenitor cells, as lesions in each of six, individual δ-protocadherin genes increase cell division in the developing hindbrain. Moreover, Wnt/β-catenin signaling is upregulated in δ-protocadherin mutants and inhibition of the canonical Wnt pathway occludes the observed proliferation increases. We show that the δ-protocadherins physically associate with the Wnt receptor Ryk, and that Ryk is required for the increased proliferation in protocadherin mutants. Thus, the δ-protocadherins act as novel regulators of Wnt/β-catenin signaling during neural development and could provide lineage-restricted local regulation of canonical Wnt signaling and cell proliferation.


2006 ◽  
Vol 290 (2) ◽  
pp. G189-G193 ◽  
Author(s):  
Neil D. Theise

This essay will address areas of liver stem/progenitor cell studies in which consensus has emerged and in which controversy still prevails over consensus, but it will also highlight important themes that inevitably should be a focus of liver stem/progenitor cell investigations in coming years. Thus concepts regarding cell plasticity, the existence of a physiological/anatomic stem cell niche, and whether intrahepatic liver stem/progenitor cells comprise true stem cells or progenitor cells (or both) will be approached in some detail.


2013 ◽  
Vol 113 (suppl_1) ◽  
Author(s):  
Elizebeth C Turner ◽  
Chien Ling Huang ◽  
Neya Sawhney ◽  
Kalai Govindarajan ◽  
Arun H S Kumar ◽  
...  

The aim of this study was to characterise a recently identified adult smooth muscle stem-like/progenitor cell (SMSPC) and exploit the ability to differentiate contractile smooth muscle cells (SMC) from SMSPCs facilitating tissue engineering of small to medium arteries, an approach which is hampered by inadequate sources of, and scale up methodologies for autologous SMCs. We initially identified a novel SMSPC in the bone marrow (BM) of rats that retained classic stem cell-like characteristics (clonogenicity, unlimited self-renewal, high telomerase activity, expression of stem cell markers) whilst expressing low levels of SMC-like transcripts. Spontaneous differentiation favoured a SMC phenotype where ~20-fold increases in calponin and SM-MHC protein expression were observed 10 days post-embryoid body formation, an effect augmented by TGFβ (P < 0.0001). Differentiation of SMSPCs from an undifferentiated state towards contractile SMCs was Myocd-dependent. Overexpression of Myocd in SMSPCs increased their percentage contraction in response to AngII (P < 0.05) and KCL (P < 0.05) and Ca2+ signalling to levels indistinguishable from rat SMCs. Knockdown of SMC-specific repressors resulted in Myocd-dependent SMC differentiation of SMSPCs. Taking advantage of the fact that SMSPCs possessed unlimited self-renewal capacity ex vivo and could be successfully reprogrammed into functional SMCs, we determined the vascular tissue engineering potential of SMSPCs and showed these cells integrate as viable tissue engineered vascular grafts in vivo. Specifically, after 1 month implantation, a Myocd-GFP+/Isl1+ smooth muscle layer was observed in the grafts along with a vWF+ luminal endothelial layer and multi-layered collagen and elastin fibre formation throughout the graft wall. We have recently isolated cells with similar SMSPC phenotype and differentiation potential from human peripheral blood. This study demonstrates that adult SMSPCs derived from rat BM and human blood can be reprogrammed to efficiently generate large quantities of mature contractile SMCs, displaying great utility as a cellular backbone for tissue engineered vascular grafts, making them an attractive source for vascular cell therapy and surgical applications.


2010 ◽  
Vol 17 (8) ◽  
pp. 1269-1273 ◽  
Author(s):  
R. Keith Reeves ◽  
Qing Wei ◽  
Patricia N. Fultz

ABSTRACT Fms-like tyrosine kinase 3 ligand (FLT3-L) is critical for the differentiation and self-renewal of CD34+ progenitor cells in primates and has been used therapeutically to mobilize progenitor and dendritic cells in vivo. However, little is known regarding the expansion of progenitor cells outside of peripheral blood, particularly in bone marrow (BM), where progenitor cells primarily reside. Evaluation of FLT3-L-mediated cell mobilization during lentivirus infections, where the numbers of CD34+ progenitor cells are reduced, is limited. We enumerated frequencies and absolute numbers of CD34+ progenitor cells in blood and BM of naive and SIV- or SHIV-infected macaques during and after the administration of FLT3-L. Flow cytometric analyses revealed that, while CD34+ cells increased in the circulation, no expansion was observed in BM. Furthermore, in the BM intracellular Ki67, a marker of cell proliferation, was downregulated in CD34+ progenitor cells but was upregulated significantly in the bulk cell population. Although the exact mechanism(s) remains unclear, these data suggest that CD34+ cell mobilization in blood was the result of cellular emigration from BM and not the proliferation of CD34+ cells already in the periphery. It is possible that the decreased progenitor cell proliferation observed in BM is evidence of a negative regulatory mechanism preventing hyperproliferation and development of neoplastic cells.


1993 ◽  
Vol 121 (6) ◽  
pp. 1397-1407 ◽  
Author(s):  
R D McKinnon ◽  
G Piras ◽  
J A Ida ◽  
M Dubois-Dalcq

Oligodendrocyte-type-2 astrocyte (O-2A) glial progenitor cells undergo a limited number of mitotic divisions in response to PDGF before differentiating into oligodendrocytes, the myelin-forming cell of the CNS. We examined the mechanism limiting O-2A proliferation, and demonstrate that these cells secrete an inhibitor of cell proliferation that can be neutralized with antibodies to TGF-beta. O-2A cells also secrete an inhibitory activity that cannot be neutralized with TGF-beta antibodies. O-2A progenitor cultures express TGF-beta 1 isoform and its transcript, while oligodendrocyte cultures express TGF-beta 1, beta-2, and beta-3 isoforms. Both recombinant TGF-beta 1 and O-2A conditioned medium inhibit the proliferation of O-2A progenitor cells cultured in the presence of PDGF, and this inhibition can be partially neutralized with polyclonal TGF-beta antibodies. Thus, TGF-beta produced by O-2A cells may limit PDGF-driven mitosis and promote oligodendrocyte development. TGF-beta is a less potent inhibitor of O-2A proliferation when these cells are cultured in the presence of bFGF, suggesting that bFGF interferes with TGF-beta signaling. Thus, the production of TGF-beta by cells in the O-2A lineage may account for the distinct effects of PDGF and bFGF on O-2A progenitor cell proliferation. Moreover, our results suggest that TGF-beta may be an important mediator of oligodendrocyte differentiation.


2016 ◽  
Vol 64 (4) ◽  
pp. 929.1-929 ◽  
Author(s):  
JD Rinaldi ◽  
W Hu ◽  
S Majundar ◽  
D Hu ◽  
GS Prins ◽  
...  

We previously demonstrated that estrogen regulates human prostate stem/progenitor cell amplification by directly targeting estrogen receptors (ERs); ERα stimulates whereas ERβ suppresses stem cell self-renewal. In addition to ERα and ERβ, we find that human prostate stem/progenitor cells express robust level of IGF-1R. Since ER actions can be modified by IGF-1R through ligand-independent ER phosphorylation, we herein sought to characterize potential cross-talk between estrogen and IGF-1 signaling pathways in regulating human prostate stem/progenitor cell amplification. Human prostate stem/progenitor cells were isolated from normal primary prostate epithelial cells (PrEC) using 3-D prostasphere (PS) culture. Similar to estradiol-17β (E2), 5 nM IGF-1 treatment increased the number of PS as well as long-term BrdU-retaining prostate stem cells. Conversely, knockdown of IGF-1R by siRNA decreased both parameters and consistently increased PS ERβ expression. Together these findings suggest that IGF-1R activation may drive prostate stem cell amplification through suppression of ERβ. Further studies revealed that E2 (10 nM) exposure induced IGF-1R phosphorylation while IGF-1R knockdown inhibited the non-genomic E2-induced pAkt and pERK confirming the cross-talk between these two signaling pathways. IGF-1R knockdown decreased PHLDA1, a known IGF-1 target gene, inhibited E2-induced ERα phosphorylation, suggesting a positive interaction between IGF-1R and ERα. In summary, the present results document robust crosstalk between estrogen and IGF-1 signaling which together regulate their downstream signal molecules including pAKT/pERK and PHLDA1. We propose that these pathways coordinately modulate prostate stem and progenitor cell numbers to effectively maintain glandular homeostasis. Supported by NIH/NCI award R01 CA172220; scholarship by FAPESP grant#2014/10965-6.


Sign in / Sign up

Export Citation Format

Share Document