16 results on '"Joseph Aldahl"'
Search Results
2. Aberrant androgen action in prostatic progenitor cells induces oncogenesis and tumor development through IGF1 and Wnt axes
- Author
-
Won Kyung Kim, Adam W. Olson, Jiaqi Mi, Jinhui Wang, Dong-Hoon Lee, Vien Le, Alex Hiroto, Joseph Aldahl, Christian H. Nenninger, Alyssa J. Buckley, Robert Cardiff, Sungyong You, and Zijie Sun
- Subjects
Science - Abstract
Activation of the androgen receptor (AR) through androgen binding is essential for prostate tumorigenesis. Here the authors show that AR activation in a subpopulation of prostatic progenitor cells can initiate prostatic intraepithelial neoplasia formation and promotes prostate cancer development through activation of IGF1 and Wnt/β-catenin signalling pathways.
- Published
- 2022
- Full Text
- View/download PDF
3. Loss of androgen signaling in mesenchymal sonic hedgehog responsive cells diminishes prostate development, growth, and regeneration.
- Author
-
Vien Le, Yongfeng He, Joseph Aldahl, Erika Hooker, Eun-Jeong Yu, Adam Olson, Won Kyung Kim, Dong-Hoon Lee, Monica Wong, Ruoyu Sheng, Jiaqi Mi, Joseph Geradts, Gerald R Cunha, and Zijie Sun
- Subjects
Genetics ,QH426-470 - Abstract
Prostate embryonic development, pubertal and adult growth, maintenance, and regeneration are regulated through androgen signaling-mediated mesenchymal-epithelial interactions. Specifically, the essential role of mesenchymal androgen signaling in the development of prostate epithelium has been observed for over 30 years. However, the identity of the mesenchymal cells responsible for this paracrine regulation and related mechanisms are still unknown. Here, we provide the first demonstration of an indispensable role of the androgen receptor (AR) in sonic hedgehog (SHH) responsive Gli1-expressing cells, in regulating prostate development, growth, and regeneration. Selective deletion of AR expression in Gli1-expressing cells during embryogenesis disrupts prostatic budding and impairs prostate development and formation. Tissue recombination assays showed that urogenital mesenchyme (UGM) containing AR-deficient mesenchymal Gli1-expressing cells combined with wildtype urogenital epithelium (UGE) failed to develop normal prostate tissue in the presence of androgens, revealing the decisive role of AR in mesenchymal SHH responsive cells in prostate development. Prepubescent deletion of AR expression in Gli1-expressing cells resulted in severe impairment of androgen-induced prostate growth and regeneration. RNA-sequencing analysis showed significant alterations in signaling pathways related to prostate development, stem cells, and organ morphogenesis in AR-deficient Gli1-expressing cells. Among these altered pathways, the transforming growth factor β1 (TGFβ1) pathway was up-regulated in AR-deficient Gli1-expressing cells. We further demonstrated the activation of TGFβ1 signaling in AR-deleted prostatic Gli1-expressing cells, which inhibits prostate epithelium growth through paracrine regulation. These data demonstrate a novel role of the AR in the Gli1-expressing cellular niche for regulating prostatic cell fate, morphogenesis, and renewal, and elucidate the mechanism by which mesenchymal androgen-signaling through SHH-responsive cells elicits the growth and regeneration of prostate epithelium.
- Published
- 2020
- Full Text
- View/download PDF
4. The comprehensive role of E-cadherin in maintaining prostatic epithelial integrity during oncogenic transformation and tumor progression.
- Author
-
Adam Olson, Vien Le, Joseph Aldahl, Eun-Jeong Yu, Erika Hooker, Yongfeng He, Dong-Hong Lee, Won Kyung Kim, Robert D Cardiff, Joseph Geradts, and Zijie Sun
- Subjects
Genetics ,QH426-470 - Abstract
E-cadherin complexes with the actin cytoskeleton via cytoplasmic catenins and maintains the functional characteristics and integrity of the epithelia in normal epithelial tissues. Lost expression of E-cadherin disrupts this complex resulting in loss of cell polarity, epithelial denudation and increased epithelial permeability in a variety of tissues. Decreased expression of E-cadherin has also been observed in invasive and metastatic human tumors. In this study, we investigated the effect of E-cadherin loss in prostatic epithelium using newly developed genetically engineered mouse models. Deletion of E-cadherin in prostatic luminal epithelial cells with modified probasin promoter driven Cre (PB-Cre4) induced the development of mouse prostatic intraepithelial neoplasia (PIN). An increase in levels of cytoplasmic and nuclear β-catenin appeared in E-cadherin deleted atypical cells within PIN lesions. Using various experimental approaches, we further demonstrated that the knockdown of E-cadherin expression elevated free cytoplasmic and nuclear β-catenin and enhanced androgen-induced transcription and cell growth. Intriguingly, pathological changes representing prostatic epithelial cell denudation and increased apoptosis accompanied the above PIN lesions. The essential role of E-cadherin in maintaining prostatic epithelial integrity and organization was further demonstrated using organoid culture approaches. To directly assess the role of loss of E-cadherin in prostate tumor progression, we generated a new mouse model with bigenic Cdh1 and Pten deletion in prostate epithelium. Early onset, aggressive tumor phenotypes presented in the compound mice. Strikingly, goblet cell metaplasia was observed, intermixed within prostatic tumor lesions of the compound mice. This study provides multiple lines of novel evidence demonstrating a comprehensive role of E-cadherin in maintaining epithelial integrity during the course of prostate oncogenic transformation, tumor initiation and progression.
- Published
- 2019
- Full Text
- View/download PDF
5. Deletion of the p16INK4a tumor suppressor and expression of the androgen receptor induce sarcomatoid carcinomas with signet ring cells in the mouse prostate.
- Author
-
Dong-Hong Lee, Eun-Jeong Yu, Joseph Aldahl, Julie Yang, Yongfeng He, Erika Hooker, Vien Le, Jiaqi Mi, Adam Olson, Huiqing Wu, Joseph Geradts, Guang Q Xiao, Mark L Gonzalgo, Robert D Cardiff, and Zijie Sun
- Subjects
Medicine ,Science - Abstract
The tumor suppressor p16Ink4a, encoded by the INK4a gene, is an inhibitor of cyclin D-dependent kinases 4 and 6, CDK4 and CDK6. This inhibition prevents the phosphorylation of the retinoblastoma protein (pRb), resulting in cellular senescence through inhibition of E2F-mediated transcription of S phase genes required for cell proliferation. The p16Ink4a plays an important role in tumor suppression, whereby its deletion, mutation, or epigenetic silencing is a frequently observed genetic alteration in prostate cancer. To assess its roles and related molecular mechanisms in prostate cancer initiation and progression, we generated a mouse model with conditional deletion of p16Ink4a in prostatic luminal epithelium. The mice underwent oncogenic transformation and developed prostatic intraepithelial neoplasia (PIN) from eight months of age, but failed to develop prostatic tumors. Given the prevalence of aberrant androgen signaling pathways in prostate cancer initiation and progression, we then generated R26hARL/wt:p16L/L: PB-Cre4 compound mice, in which conditional expression of the human AR transgene and deletion of p16Ink4a co-occur in prostatic luminal epithelial cells. While R26hARL/wt:PB-Cre4 mice showed no visible pathological changes, R26hARL/wt:p16L/L: PB-Cre4 compound mice displayed an early onset of high-grade PIN (HGPIN), prostatic carcinoma, and metastatic lesions. Strikingly, we observed tumors resembling human sarcomatoid carcinoma with intermixed focal regions of signet ring cell carcinoma (SRCC) in the prostates of the compound mice. Further characterization of these tumors showed they were of luminal epithelial cell origin, and featured characteristics of epithelial to mesenchymal transition (EMT) with enhanced proliferative and invasive capabilities. Our results not only implicate a biological role for AR expression and p16Ink4a deletion in the pathogenesis of prostatic SRCC, but also provide a new and unique genetically engineered mouse (GEM) model for investigating the molecular mechanisms for SRCC development.
- Published
- 2019
- Full Text
- View/download PDF
6. Androgen receptor with short polyglutamine tract preferably enhances Wnt/β-catenin-mediated prostatic tumorigenesis
- Author
-
Dong-Hoon Lee, Adam Olson, Jiaqi Mi, Joseph Geradts, Erika Hooker, Diane M. Robins, Yongfeng He, Vien Le, Won Kyung Kim, Joseph Aldahl, Zijie Sun, and Eun-Jeong Yu
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Carcinogenesis ,medicine.drug_class ,MYC ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,The androgen receptor ,Genetics ,medicine ,Animals ,Humans ,Wnt Signaling Pathway ,Molecular Biology ,beta Catenin ,Cell Proliferation ,Sequence Analysis, RNA ,Prostate Cancer ,Prostate ,Wnt signaling pathway ,Prostatic Neoplasms ,β-catenin ,Polyglutamine tract ,Androgen ,medicine.disease ,Wnt signaling ,Black or African American ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Disease Models, Animal ,030104 developmental biology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Catenin ,Cancer research ,Peptides ,Chromatin immunoprecipitation - Abstract
Polyglutamine (polyQ) tract polymorphism within the human androgen receptor (AR) shows population heterogeneity. African American men possess short polyQ tracts significantly more frequently than Caucasian American men. The length of polyQ tracts is inversely correlated with the risk of prostate cancer, age of onset, and aggressiveness at diagnosis. Aberrant activation of Wnt signaling also reveals frequently in advanced prostate cancer, and an enrichment of androgen and Wnt signaling activation has been observed in African American patients. Here, we assessed aberrant expression of AR bearing different polyQ tracts and stabilized β-catenin in prostate tumorigenesis using newly generated mouse models. We observed an early onset oncogenic transformation, accelerated tumor cell growth, and aggressive tumor phenotypes in the compound mice bearing short polyQ tract AR and stabilized β-catenin. RNA sequencing analysis showed a robust enrichment of Myc-regulated downstream genes in tumor samples bearing short polyQ AR versus those with longer polyQ tract AR. Upstream regulator analysis further identified Myc as the top candidate of transcriptional regulators in tumor cells from the above mouse samples with short polyQ tract AR and β-catenin. Chromatin immunoprecipitation analyses revealed increased recruitment of β-catenin and AR on the c-Myc gene regulatory locus in the tumor tissues expressing stabilized β-catenin and shorter polyQ tract AR. These data demonstrate a promotional role of aberrant activation of Wnt/β-catenin in combination with short polyQ AR expression in prostate tumorigenesis and suggest a potential mechanism underlying aggressive prostatic tumor development, which has been frequently observed in African American patients.
- Published
- 2020
7. Stromal androgen and hedgehog signaling regulates stem cell niches in pubertal prostate development
- Author
-
Joseph Aldahl, Alex Hiroto, Sungyong You, Gerald R. Cunha, Jinhui Wang, Vien Le, Minhyung Kim, Adam Olson, Xiwei Wu, Zijie Sun, Dong-Hoon Lee, and Won Kyung Kim
- Subjects
Male ,Stromal cell ,medicine.drug_class ,Cell ,Morphogenesis ,Biology ,Zinc Finger Protein GLI1 ,Paracrine signalling ,Mice ,Prostate ,medicine ,Animals ,Cell Lineage ,Hedgehog Proteins ,RNA-Seq ,Stem Cell Niche ,Molecular Biology ,Cells, Cultured ,Cell Differentiation ,Epithelial Cells ,Androgen ,Stem Cells and Regeneration ,Hedgehog signaling pathway ,Cell biology ,medicine.anatomical_structure ,Receptors, Androgen ,Androgens ,Stem cell ,Single-Cell Analysis ,Transcriptome ,Developmental Biology ,Signal Transduction - Abstract
Stromal androgen-receptor (AR) action is essential for prostate development, morphogenesis and regeneration. However, mechanisms underlying how stromal AR maintains the cell niche in support of pubertal prostatic epithelial growth are unknown. Here, using advanced mouse genetic tools, we demonstrate that selective deletion of stromal AR expression in prepubescent Shh-responsive Gli1-expressing cells significantly impedes pubertal prostate epithelial growth and development. Single-cell transcriptomic analyses showed that AR loss in these prepubescent Gli1-expressing cells dysregulates androgen signaling-initiated stromal-epithelial paracrine interactions, leading to growth retardation of pubertal prostate epithelia and significant development defects. Specifically, AR loss elevates Shh-signaling activation in both prostatic stromal and adjacent epithelial cells, directly inhibiting prostatic epithelial growth. Single-cell trajectory analyses further identified aberrant differentiation fates of prostatic epithelial cells directly altered by stromal AR deletion. In vivo recombination of AR-deficient stromal Gli1-lineage cells with wild-type prostatic epithelial cells failed to develop normal prostatic epithelia. These data demonstrate previously unidentified mechanisms underlying how stromal AR-signaling facilitates Shh-mediated cell niches in pubertal prostatic epithelial growth and development.
- Published
- 2021
8. Aberrant androgen action in prostatic progenitor cells induces oncogenesis and tumor development through IGF1 and Wnt axes
- Author
-
Won Kyung Kim, Adam W. Olson, Jiaqi Mi, Jinhui Wang, Dong-Hoon Lee, Vien Le, Alex Hiroto, Joseph Aldahl, Christian H. Nenninger, Alyssa J. Buckley, Robert Cardiff, Sungyong You, and Zijie Sun
- Subjects
Male ,Multidisciplinary ,Carcinogenesis ,Stem Cells ,Prostate ,General Physics and Astronomy ,Prostatic Neoplasms ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,Wnt Proteins ,Cell Transformation, Neoplastic ,Receptors, Androgen ,Androgens ,Humans ,Insulin-Like Growth Factor I ,Wnt Signaling Pathway ,beta Catenin - Abstract
Androgen/androgen receptor (AR) signaling pathways are essential for prostate tumorigenesis. However, the fundamental mechanisms underlying the AR functioning as a tumor promoter in inducing prostatic oncogenesis still remain elusive. Here, we demonstrate that a subpopulation of prostatic Osr1 (odd skipped-related 1)-lineage cells functions as tumor progenitors in prostate tumorigenesis. Single cell transcriptomic analyses reveal that aberrant AR activation in these cells elevates insulin-like growth factor 1 (IGF1) signaling pathways and initiates oncogenic transformation. Elevating IGF1 signaling further cumulates Wnt/β-catenin pathways in transformed cells to promote prostate tumor development. Correlations between altered androgen, IGF1, and Wnt/β-catenin signaling are also identified in human prostate cancer samples, uncovering a dynamic regulatory loop initiated by the AR through prostate cancer development. Co-inhibition of androgen and Wnt-signaling pathways significantly represses the growth of AR-positive tumor cells in both ex-vivo and in-vivo, implicating co-targeting therapeutic strategies for these pathways to treat advanced prostate cancer.
- Published
- 2021
9. Loss of the tumor suppressor, Tp53, enhances the androgen receptor-mediated oncogenic transformation and tumor development in the mouse prostate
- Author
-
Junhee Yoon, Yongfeng He, Won Kyung Kim, Vien Le, Dong-Hoon Lee, Erika Hooker, Joseph Aldahl, Robert D. Cardiff, Joseph Geradts, Eun-Jeong Yu, Sungyong You, Zijie Sun, Huiqing Wu, Julie S Yang, and Daniel T. Johnson
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Transgene ,SOX2 ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,The androgen receptor ,Genetics ,medicine ,p53 tumor suppressor ,Animals ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,Regulation of gene expression ,Prostate Cancer ,SOXB1 Transcription Factors ,Prostatic Neoplasms ,medicine.disease ,3. Good health ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Androgen receptor ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Cancer research ,Tumor Suppressor Protein p53 ,Transcriptome ,Carcinogenesis ,knockout mice ,Gene Deletion ,Signal Transduction - Abstract
Recent genome analysis of human prostate cancers demonstrated that both AR gene amplification and TP53 mutation are among the most frequently observed alterations in advanced prostate cancer. However, the biological role of these dual genetic alterations in prostate tumorigenesis is largely unknown. In addition, there are no biologically relevant models that can be used to assess the molecular mechanisms for these genetic abnormalities. Here, we report a novel mouse model, in which elevated transgenic AR expression and Trp53 deletion occur simultaneously in mouse prostatic epithelium to mimic human prostate cancer cells. These compound mice developed an earlier onset of high-grade prostatic intraepithelial neoplasia and accelerated prostate tumors in comparison with mice harboring only the AR transgene. Histological analysis showed prostatic sarcomatoid and basaloid carcinomas with massive squamous differentiation in the above compound mice. RNA-sequencing analyses identified a robust enrichment of the signature genes for human prostatic basal cell carcinomas in the above prostate tumors. Master regulator analysis revealed SOX2 as a transcriptional regulator in prostatic basal cell tumors. Elevated expression of SOX2 and its downstream target genes were detected in prostatic tumors of the compound mice. Chromatin immunoprecipitation analyses implicate a coregulatory role of AR and SOX2 in the expression of prostatic basal cell signature genes. Our data demonstrate a critical role of SOX2 in prostate tumorigenesis and provide mechanistic insight into prostate tumor aggressiveness and progression mediated by aberrant AR and p53 signaling pathways.
- Published
- 2019
10. A pivotal role of androgen signaling in Notch-responsive cells in prostate development, maturation, and regeneration
- Author
-
Charles Murtaugh, Zijie Sun, Erika Hooker, Joseph Aldahl, Won Kyung Kim, Eun-Jeong Yu, Monica T. Y. Wong, Adam Olson, Gerald R. Cunha, Vien Le, Yongfeng He, and Dong-Hoon Lee
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.drug_class ,Population ,Notch signaling pathway ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Prostate ,medicine ,Animals ,Regeneration ,HES1 ,education ,Molecular Biology ,education.field_of_study ,Receptors, Notch ,Regeneration (biology) ,Cell Biology ,Androgen ,Embryonic stem cell ,Cell biology ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Androgen ,Models, Animal ,Androgens ,Transcription Factor HES-1 ,030217 neurology & neurosurgery ,Signal Transduction ,Developmental Biology - Abstract
Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence also indicates a regulatory role of Notch signaling in prostate development, differentiation, and growth. However, the collaborative regulatory mechanisms of androgen and Notch signaling during prostate development, growth, and regeneration are largely unknown. Hairy and Enhancer of Split 1 (Hes1) is a transcriptional regulator of Notch signaling pathways, and its expression is responsive to Notch signaling. Hes1-expressing cells have been shown to possess the regenerative capability to repopulate a variety of adult tissues. In this study, we developed new mouse models to directly assess the role of the androgen receptor in prostatic Hes1-expressing cells. Selective deletion of AR expression in embryonic Hes1-expressing cells impeded early prostate development both in vivo and in tissue xenograft experiments. Prepubescent deletion of AR expression in Hes1-expressing cells resulted in prostate glands containing abnormalities in cell morphology and gland architecture. A population of castration-resistant Hes1-expressing cells was revealed in the adult prostate, with the ability to repopulate prostate epithelium following androgen supplementation. Deletion of AR in Hes1-expressing cells diminishes their regenerative ability. These lines of evidence demonstrate a critical role for the AR in Notch-responsive cells during the course of prostate development, morphogenesis, and regeneration, and implicate a mechanism underlying interaction between the androgen and Notch signaling pathways in the mouse prostate.
- Published
- 2019
11. Androgen action in cell fate and communication during prostate development at single-cell resolution
- Author
-
Alex Hiroto, Jiaqi Mi, Adam Olson, Xiwei Wu, Vien Le, Jinhui Wang, Hong Zeng, Dong-Hoon Lee, Joseph Aldahl, Won Kyung Kim, and Zijie Sun
- Subjects
Male ,Stromal cell ,medicine.drug_class ,Cell Communication ,Biology ,Mesoderm ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Cell Lineage ,Genes, Developmental ,RNA-Seq ,Molecular Biology ,030304 developmental biology ,Prostatic bud formation ,0303 health sciences ,Mesenchymal stem cell ,Wnt signaling pathway ,Prostate ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Epithelial Cells ,Androgen ,Hedgehog signaling pathway ,Cell biology ,Androgen receptor ,Receptors, Androgen ,Androgens ,Signal transduction ,Single-Cell Analysis ,Stromal Cells ,030217 neurology & neurosurgery ,Gene Deletion ,Developmental Biology ,Signal Transduction ,Research Article - Abstract
Androgens/androgen receptor (AR)-mediated signaling pathways are essential for prostate development, morphogenesis and regeneration. Specifically, stromal AR signaling has been shown to be essential for prostatic initiation. However, the molecular mechanisms underlying AR-initiated mesenchymal-epithelial interactions in prostate development remain unclear. Here, using a newly generated mouse model, we have directly addressed the fate and role of genetically marked AR-expressing cells during embryonic prostate development. Androgen signaling-initiated signaling pathways were identified in mesenchymal niche populations at single-cell transcriptomic resolution. The dynamic cell-signaling networks regulated by stromal AR were additionally characterized in relation to prostatic epithelial bud formation. Pseudotime analyses further revealed the differentiation trajectory and fate of AR-expressing cells in both prostatic mesenchymal and epithelial cell populations. Specifically, the cellular properties of Zeb1-expressing progenitors were assessed. Selective deletion of AR signaling in a subpopulation of mesenchymal rather than epithelial cells dysregulated the expression of the master regulators and significantly impaired prostatic bud formation. These data provide novel, high-resolution evidence demonstrating the important role of mesenchymal androgen signaling in the cellular niche controlling prostate early development by initiating dynamic mesenchyme-epithelia cell interactions.
- Published
- 2020
12. Loss of androgen signaling in mesenchymal sonic hedgehog responsive cells diminishes prostate development, growth, and regeneration
- Author
-
Erika Hooker, Jiaqi Mi, Monica T. Y. Wong, Won Kyung Kim, Yongfeng He, Eun-Jeong Yu, Adam Olson, Zijie Sun, Joseph Aldahl, Dong-Hoon Lee, Vien Le, Ruoyu Sheng, Gerald R. Cunha, Joseph Geradts, and Barsh, Gregory S
- Subjects
Male ,Cancer Research ,Physiology ,Developmental Signaling ,QH426-470 ,Regenerative Medicine ,Biochemistry ,Epithelium ,Androgen ,Mice ,Endocrinology ,0302 clinical medicine ,Cell Signaling ,Prostate ,Transforming Growth Factor beta ,Receptors ,Medicine and Health Sciences ,Morphogenesis ,2.1 Biological and endogenous factors ,Aetiology ,Sonic hedgehog ,Cells, Cultured ,Genetics (clinical) ,Cancer ,Pediatric ,0303 health sciences ,Cultured ,biology ,integumentary system ,Prostate Cancer ,Signaling cascades ,Animal Models ,Cell biology ,medicine.anatomical_structure ,Experimental Organism Systems ,Receptors, Androgen ,Paracrine Signaling ,Androgens ,Stem Cell Research - Nonembryonic - Non-Human ,Anatomy ,Stem cell ,Research Article ,Biotechnology ,Signal Transduction ,Urologic Diseases ,1.1 Normal biological development and functioning ,Mesenchyme ,Cells ,Mouse Models ,Research and Analysis Methods ,Zinc Finger Protein GLI1 ,03 medical and health sciences ,Paracrine signalling ,Exocrine Glands ,Model Organisms ,Underpinning research ,medicine ,Genetics ,Animals ,Regeneration ,Hedgehog Proteins ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Endocrine Physiology ,Regeneration (biology) ,Mesenchymal stem cell ,Biology and Life Sciences ,Epithelial Cells ,Mesenchymal Stem Cells ,Cell Biology ,Stem Cell Research ,Hormones ,Androgen receptor ,Biological Tissue ,TGF-beta signaling cascade ,Animal Studies ,biology.protein ,Prostate Gland ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Prostate embryonic development, pubertal and adult growth, maintenance, and regeneration are regulated through androgen signaling-mediated mesenchymal-epithelial interactions. Specifically, the essential role of mesenchymal androgen signaling in the development of prostate epithelium has been observed for over 30 years. However, the identity of the mesenchymal cells responsible for this paracrine regulation and related mechanisms are still unknown. Here, we provide the first demonstration of an indispensable role of the androgen receptor (AR) in sonic hedgehog (SHH) responsive Gli1-expressing cells, in regulating prostate development, growth, and regeneration. Selective deletion of AR expression in Gli1-expressing cells during embryogenesis disrupts prostatic budding and impairs prostate development and formation. Tissue recombination assays showed that urogenital mesenchyme (UGM) containing AR-deficient mesenchymal Gli1-expressing cells combined with wildtype urogenital epithelium (UGE) failed to develop normal prostate tissue in the presence of androgens, revealing the decisive role of AR in mesenchymal SHH responsive cells in prostate development. Prepubescent deletion of AR expression in Gli1-expressing cells resulted in severe impairment of androgen-induced prostate growth and regeneration. RNA-sequencing analysis showed significant alterations in signaling pathways related to prostate development, stem cells, and organ morphogenesis in AR-deficient Gli1-expressing cells. Among these altered pathways, the transforming growth factor β1 (TGFβ1) pathway was up-regulated in AR-deficient Gli1-expressing cells. We further demonstrated the activation of TGFβ1 signaling in AR-deleted prostatic Gli1-expressing cells, which inhibits prostate epithelium growth through paracrine regulation. These data demonstrate a novel role of the AR in the Gli1-expressing cellular niche for regulating prostatic cell fate, morphogenesis, and renewal, and elucidate the mechanism by which mesenchymal androgen-signaling through SHH-responsive cells elicits the growth and regeneration of prostate epithelium., Author summary Prostate formation, growth, and regeneration, as well as tumorigenesis, depend on androgens and androgen receptor (AR)-mediated signaling pathways. Tissue recombination assays done more than 30 years ago demonstrated a decisive role for stromal androgen signaling in prostatic epithelium development. However, in the intervening time, the identity of the mesenchymal cells in the urogenital sinus mesenchyme that convey androgen signaling and control prostate epithelium development, morphogenesis, and regeneration has not been determined. In this study, using mouse genetic tools, we demonstrate for the first time that selective deletion of AR in mesenchymal Gli1-expressing cells abolishes early development of prostate tissue and normal prostate formation, and diminishes prostate pubertal growth and regeneration. In addition, using tissue recombination assays, we directly determined an essential requirement for AR expression in mesenchymal Gli1-expressing cells during prostate epithelium development. Our results not only resolve a 30-year-old scientific puzzle by identifying the mesenchymal cell properties of androgen-responsive cells that elicit development of the embryonic prostate epithelium, but also explore a new regulatory mechanism for androgen and Shh signaling-mediated cellular niches in regulating prostatic cell fate, growth, and renewal through paracrine regulation. Given the importance of sex hormone and hedgehog signaling pathways in human development and tumorigenesis, this study extends beyond the field of prostate biology, raising new questions underlying sex hormone and SHH signaling in development and tumorigenesis.
- Published
- 2020
13. Aberrant activation of hepatocyte growth factor/MET signaling promotes β-catenin–mediated prostatic tumorigenesis
- Author
-
Zijie Sun, Joseph Aldahl, Erika Hooker, Eun-Jeong Yu, Dong-Hoon Lee, Joseph Geradts, Vien Le, Adam Olson, Won Kyung Kim, Ariana Pineda, Jiaqi Mi, and Yongfeng He
- Subjects
0301 basic medicine ,Male ,Carcinogenesis ,Mice, SCID ,medicine.disease_cause ,Biochemistry ,Proto-Oncogene Mas ,Receptor tyrosine kinase ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Prostate ,medicine ,Animals ,Humans ,Molecular Biology ,beta Catenin ,030102 biochemistry & molecular biology ,biology ,Hepatocyte Growth Factor ,Wnt signaling pathway ,Prostatic Neoplasms ,Molecular Bases of Disease ,Cell Biology ,Proto-Oncogene Proteins c-met ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Catenin ,Cancer research ,biology.protein ,Hepatocyte growth factor ,medicine.drug ,Signal Transduction - Abstract
Co-occurrence of aberrant hepatocyte growth factor (HGF)/MET proto-oncogene receptor tyrosine kinase (MET) and Wnt/β-catenin signaling pathways has been observed in advanced and metastatic prostate cancers. This co-occurrence positively correlates with prostate cancer progression and castration-resistant prostate cancer development. However, the biological consequences of these abnormalities in these disease processes remain largely unknown. Here, we investigated the aberrant activation of HGF/MET and Wnt/β-catenin cascades in prostate tumorigenesis by using a newly generated mouse model in which both murine Met transgene and stabilized β-catenin are conditionally co-expressed in prostatic epithelial cells. These compound mice displayed accelerated prostate tumor formation and invasion compared with their littermates that expressed only stabilized β-catenin. RNA-Seq and quantitative RT-PCR analyses revealed increased expression of genes associated with tumor cell proliferation, progression, and metastasis. Moreover, Wnt signaling pathways were robustly enriched in prostate tumor samples from the compound mice. ChIP-qPCR experiments revealed increased β-catenin recruitment within the regulatory regions of the Myc gene in tumor cells of the compound mice. Interestingly, the occupancy of MET on the Myc promoter also appeared in the compound mouse tumor samples, implicating a novel role of MET in β-catenin–mediated transcription. Results from implanting prostate graft tissues derived from the compound mice and controls into HGF-transgenic mice further uncovered that HGF induces prostatic oncogenic transformation and cell growth. These results indicate a role of HGF/MET in β-catenin–mediated prostate cancer cell growth and progression and implicate a molecular mechanism whereby nuclear MET promotes aberrant Wnt/β-catenin signaling–mediated prostate tumorigenesis.
- Published
- 2019
14. Activation of hepatocyte growth factor/MET signaling initiates oncogenic transformation and enhances tumor aggressiveness in the murine prostate
- Author
-
Zijie Sun, Daniel T. Johnson, Dong Hoon Lee, Jiaqi Mi, Yongfeng He, Mark L. Gonzalgo, Eun Jeong Yu, Huiqing Wu, Erika Hooker, Vien Le, Hong Zeng, Joseph Aldahl, and Steven Balog
- Subjects
0301 basic medicine ,Male ,Tumor suppressor gene ,Mice, Transgenic ,medicine.disease_cause ,Biochemistry ,Proto-Oncogene Mas ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,medicine ,PTEN ,Animals ,Epithelial–mesenchymal transition ,Molecular Biology ,biology ,Hepatocyte Growth Factor ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Molecular Bases of Disease ,Cell Biology ,Proto-Oncogene Proteins c-met ,medicine.disease ,030104 developmental biology ,Cell Transformation, Neoplastic ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Hepatocyte growth factor ,Carcinogenesis ,medicine.drug ,Signal Transduction - Abstract
Emerging evidence has shown that the hepatocyte growth factor (HGF) and its receptor, MET proto-oncogene, receptor tyrosine kinase (MET), promote cell proliferation, motility, morphogenesis, and angiogenesis. Whereas up-regulation of MET expression has been observed in aggressive and metastatic prostate cancer, a clear understanding of MET function in prostate tumorigenesis remains elusive. Here, we developed a conditional Met transgenic mouse strain, H11(Met/+):PB-Cre4, to mimic human prostate cancer cells with increased MET expression in the prostatic luminal epithelium. We found that these mice develop prostatic intraepithelial neoplasia after HGF administration. To further assess the biological role of MET in prostate cancer progression, we bred H11(Met/+)/Pten(LoxP/LoxP):PBCre4 compound mice, in which transgenic Met expression and deletion of the tumor suppressor gene Pten occurred simultaneously only in prostatic epithelial cells. These compound mice exhibited accelerated prostate tumor formation and invasion as well as increased metastasis compared with Pten(LoxP/LoxP):PB-Cre4 mice. Moreover, prostatic sarcomatoid carcinomas and lesions resembling the epithelial-to-mesenchymal transition developed in tumor lesions of the compound mice. RNA-Seq and qRT-PCR analyses revealed a robust enrichment of known tumor progression and metastasis-promoting genes in samples isolated from H11(Met/+)/Pten(LoxP/LoxP):PB-Cre4 compound mice compared with those from Pten(LoxP/LoxP):PB-Cre4 littermate controls. HGF-induced cell proliferation and migration also increased in mouse embryonic fibroblasts (MEFs) from animals with both Met transgene expression and Pten deletion compared with Pten-null MEFs. The results from these newly developed mouse models indicate a role for MET in hastening tumorigenesis and metastasis when combined with the loss of tumor suppressors.
- Published
- 2018
15. The comprehensive role of E-cadherin in maintaining prostatic epithelial integrity during oncogenic transformation and tumor progression
- Author
-
Won Kyung Kim, Joseph Aldahl, Robert D. Cardiff, Joseph Geradts, Erika Hooker, Yongfeng He, Adam Olson, Eun Jeong Yu, Zijie Sun, Dong Hong Lee, Vien Le, and Barsh, Gregory S
- Subjects
Male ,Aging ,Cancer Research ,Carcinogenesis ,Apoptosis ,Tumor initiation ,QH426-470 ,Cell Transformation ,Epithelium ,Transgenic ,CDH1 ,Mice ,Prostate cancer ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,2.1 Biological and endogenous factors ,RNA, Small Interfering ,Organ Cultures ,Aetiology ,beta Catenin ,Genetics (clinical) ,Cancer ,Staining ,Prostatic Intraepithelial Neoplasia ,0303 health sciences ,Tumor ,Cell Death ,Prostate Cancer ,Prostate Diseases ,Prostate ,Cell Staining ,Animal Models ,Cadherins ,CD ,Organoids ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Experimental Organism Systems ,Oncology ,Cell Processes ,Disease Progression ,Biological Cultures ,Anatomy ,Cellular Types ,Research Article ,Urologic Diseases ,Urology ,Primary Cell Culture ,Mice, Transgenic ,Mouse Models ,Biology ,Research and Analysis Methods ,Small Interfering ,Cell Line ,03 medical and health sciences ,Exocrine Glands ,Model Organisms ,Antigens, CD ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Antigens ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Cell Proliferation ,030304 developmental biology ,Neoplastic ,Goblet cell ,Animal ,Cadherin ,PTEN Phosphohydrolase ,Biology and Life Sciences ,Cancers and Neoplasms ,Prostatic Neoplasms ,Epithelial Cells ,Cell Biology ,medicine.disease ,Actin cytoskeleton ,Disease Models, Animal ,Genitourinary Tract Tumors ,Biological Tissue ,HEK293 Cells ,Specimen Preparation and Treatment ,Tumor progression ,Catenin ,Disease Models ,Animal Studies ,Cancer research ,biology.protein ,RNA ,Prostate Gland ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
E-cadherin complexes with the actin cytoskeleton via cytoplasmic catenins and maintains the functional characteristics and integrity of the epithelia in normal epithelial tissues. Lost expression of E-cadherin disrupts this complex resulting in loss of cell polarity, epithelial denudation and increased epithelial permeability in a variety of tissues. Decreased expression of E-cadherin has also been observed in invasive and metastatic human tumors. In this study, we investigated the effect of E-cadherin loss in prostatic epithelium using newly developed genetically engineered mouse models. Deletion of E-cadherin in prostatic luminal epithelial cells with modified probasin promoter driven Cre (PB-Cre4) induced the development of mouse prostatic intraepithelial neoplasia (PIN). An increase in levels of cytoplasmic and nuclear β-catenin appeared in E-cadherin deleted atypical cells within PIN lesions. Using various experimental approaches, we further demonstrated that the knockdown of E-cadherin expression elevated free cytoplasmic and nuclear β-catenin and enhanced androgen-induced transcription and cell growth. Intriguingly, pathological changes representing prostatic epithelial cell denudation and increased apoptosis accompanied the above PIN lesions. The essential role of E-cadherin in maintaining prostatic epithelial integrity and organization was further demonstrated using organoid culture approaches. To directly assess the role of loss of E-cadherin in prostate tumor progression, we generated a new mouse model with bigenic Cdh1 and Pten deletion in prostate epithelium. Early onset, aggressive tumor phenotypes presented in the compound mice. Strikingly, goblet cell metaplasia was observed, intermixed within prostatic tumor lesions of the compound mice. This study provides multiple lines of novel evidence demonstrating a comprehensive role of E-cadherin in maintaining epithelial integrity during the course of prostate oncogenic transformation, tumor initiation and progression., Author summary The biological significance of E-cadherin in maintaining prostatic epithelial integrity and related molecular mechanisms are still unclear. In this study, using mouse genetic tools, we directly address this important and unresolved question. Conditional deletion of E-cadherin in mouse prostatic epithelia resulted in prostatic intraepithelial neoplasia (PIN) development but no prostatic tumor formation. Both in vivo and in vitro data showed that loss of E-cadherin modulates the cellular localization of β-catenin, elevates its cytoplasmic and nuclear levels, and enhances its activity in transcription and cell proliferation. Intriguingly, in addition to PIN lesions, increased epithelial denudation and cell apoptosis also appeared within PIN lesions. This implicates that although lost E-cadherin is sufficient to introduce oncogenic transformation in prostatic epithelia, it also induces cell apoptosis and disrupts epithelial structure, preventing atypical PIN cells from progressing to tumor cells. Simultaneous deletion of Pten, a tumor suppressor, and E-cadherin in prostatic epithelia resulted in early onset, invasive prostatic tumors with admixture of goblet cells. These results demonstrate a critical role of E-cadherin in promoting prostatic tumor transdifferentiation and progression. This study further elucidates the dynamic role of E-cadherin in maintaining prostatic epithelial integrity during tumor initiation and progression.
- Published
- 2019
16. Deletion of the p16INK4a tumor suppressor and expression of the androgen receptor induce sarcomatoid carcinomas with signet ring cells in the mouse prostate
- Author
-
Joseph Geradts, Yongfeng He, Zijie Sun, Dong Hong Lee, Guang Q. Xiao, Mark L. Gonzalgo, Huiqing Wu, Joseph Aldahl, Adam Olson, Julie Yang, Robert D. Cardiff, Vien Le, Erika Hooker, Eun Jeong Yu, Jiaqi Mi, and Saleem, MOHAMMAD
- Subjects
Male ,0301 basic medicine ,Carcinogenesis ,medicine.disease_cause ,Transgenic ,Androgen ,Mice ,Prostate cancer ,0302 clinical medicine ,Adenocarcinomas ,Signet ring cell carcinoma ,Receptors ,Medicine and Health Sciences ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Staining ,Prostatic Intraepithelial Neoplasia ,Multidisciplinary ,Signet ring cell ,Prostate Cancer ,Prostate Diseases ,Retinoblastoma protein ,Cell Staining ,Animal Models ,Signet Ring Cell ,3. Good health ,Experimental Organism Systems ,Oncology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Medicine ,Anatomy ,Research Article ,Urologic Diseases ,Epithelial-Mesenchymal Transition ,General Science & Technology ,Urology ,Science ,Mice, Transgenic ,Mouse Models ,Biology ,Research and Analysis Methods ,Carcinomas ,03 medical and health sciences ,Model Organisms ,Exocrine Glands ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Sarcomatoid carcinoma ,Immunohistochemistry Techniques ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Proliferation ,Carcinoma ,Cancers and Neoplasms ,Biology and Life Sciences ,Prostatic Neoplasms ,medicine.disease ,Histochemistry and Cytochemistry Techniques ,Genitourinary Tract Tumors ,030104 developmental biology ,Specimen Preparation and Treatment ,Animal Studies ,Immunologic Techniques ,biology.protein ,Cancer research ,Prostate Gland ,Cyclin-dependent kinase 6 ,Carcinoma, Signet Ring Cell ,Gene Deletion - Abstract
The tumor suppressor p16Ink4a, encoded by the INK4a gene, is an inhibitor of cyclin D-dependent kinases 4 and 6, CDK4 and CDK6. This inhibition prevents the phosphorylation of the retinoblastoma protein (pRb), resulting in cellular senescence through inhibition of E2F-mediated transcription of S phase genes required for cell proliferation. The p16Ink4a plays an important role in tumor suppression, whereby its deletion, mutation, or epigenetic silencing is a frequently observed genetic alteration in prostate cancer. To assess its roles and related molecular mechanisms in prostate cancer initiation and progression, we generated a mouse model with conditional deletion of p16Ink4a in prostatic luminal epithelium. The mice underwent oncogenic transformation and developed prostatic intraepithelial neoplasia (PIN) from eight months of age, but failed to develop prostatic tumors. Given the prevalence of aberrant androgen signaling pathways in prostate cancer initiation and progression, we then generated R26hARL/wt:p16L/L: PB-Cre4 compound mice, in which conditional expression of the human AR transgene and deletion of p16Ink4a co-occur in prostatic luminal epithelial cells. While R26hARL/wt:PB-Cre4 mice showed no visible pathological changes, R26hARL/wt:p16L/L: PB-Cre4 compound mice displayed an early onset of high-grade PIN (HGPIN), prostatic carcinoma, and metastatic lesions. Strikingly, we observed tumors resembling human sarcomatoid carcinoma with intermixed focal regions of signet ring cell carcinoma (SRCC) in the prostates of the compound mice. Further characterization of these tumors showed they were of luminal epithelial cell origin, and featured characteristics of epithelial to mesenchymal transition (EMT) with enhanced proliferative and invasive capabilities. Our results not only implicate a biological role for AR expression and p16Ink4a deletion in the pathogenesis of prostatic SRCC, but also provide a new and unique genetically engineered mouse (GEM) model for investigating the molecular mechanisms for SRCC development.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.