1
|
Liu Y, Lomeli I, Kron SJ. Therapy-Induced Cellular Senescence: Potentiating Tumor Elimination or Driving Cancer Resistance and Recurrence? Cells 2024; 13:1281. [PMID: 39120312 PMCID: PMC11312217 DOI: 10.3390/cells13151281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Cellular senescence has been increasingly recognized as a hallmark of cancer, reflecting its association with aging and inflammation, its role as a response to deregulated proliferation and oncogenic stress, and its induction by cancer therapies. While therapy-induced senescence (TIS) has been linked to resistance, recurrence, metastasis, and normal tissue toxicity, TIS also has the potential to enhance therapy response and stimulate anti-tumor immunity. In this review, we examine the Jekyll and Hyde nature of senescent cells (SnCs), focusing on how their persistence while expressing the senescence-associated secretory phenotype (SASP) modulates the tumor microenvironment through autocrine and paracrine mechanisms. Through the SASP, SnCs can mediate both resistance and response to cancer therapies. To fulfill the unmet potential of cancer immunotherapy, we consider how SnCs may influence tumor inflammation and serve as an antigen source to potentiate anti-tumor immune response. This new perspective suggests treatment approaches based on TIS to enhance immune checkpoint blockade. Finally, we describe strategies for mitigating the detrimental effects of senescence, such as modulating the SASP or targeting SnC persistence, which may enhance the overall benefits of cancer treatment.
Collapse
Affiliation(s)
| | | | - Stephen J. Kron
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
2
|
Du Z, Chen X, Zhu P, Lv Q, Yong J, Gu J. Knocking down SOX2 overcomes the resistance of prostate cancer to castration via notch signaling. Mol Biol Rep 2023; 50:9007-9017. [PMID: 37716921 DOI: 10.1007/s11033-023-08757-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/16/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is a terminal type of advanced cancer resistant to androgen deprivation therapy (ADT). Due to the poor therapeutic response of CRPC, novel treatment strategies are urgently required. This study aimed to clarify the regulatory roles of the SOX2/Notch axis in CRPC. METHODS For the evaluation of the SOX2, Notch, and Hey1 expression in the prostate cancer (PCa) and CRPC tissues, we conducted immunohistochemistry (IHC) analyses. RT-PCR, Western blotting, and immunofluorescence were performed to evaluate SOX2 and Notch expression in enzalutamide-resistant LNCaP cells (Enza-R). CCK-8, Transwell, Wound healing, and Western blotting assays were used to assess the viability, invasion, migration, cell cycle, and drug-resistant in Enza-R cells. RESULTS Compared to the PCa tissues, CRPC tissues exhibited significantly elevated SOX2, Notch1, and Hey1 expression. SOX2-positive patients were more likely to develop bone metastases than SOX2-negative ones. Significant activation of the signaling associated with SOX2 and Notch was detected in Enza-R cells. The suppression of SOX2 clearly inactivated the Notch signaling and inhibited malignant behaviors, including proliferation, invasion, migration, and drug resistance in Enza-R cells. Theγsecretase inhibitor, GSI-IX, abrogated the enzalutamide resistance by inhibiting Notch signaling in vitro in vitro. Also, GSI-IX alone had a significant anti-tumor effect in Enza-R cells. CONCLUSION We demonstrated that SOX2/Notch signaling was responsible for Enzalutamide resistance in CRPC. Targeting SOX2/Notch signaling might represent a new choice for the treatment and therapy of CRPC.
Collapse
Affiliation(s)
- Zhongbo Du
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China.
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Xiaobin Chen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Pingyu Zhu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qi Lv
- Department of Operation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jun Yong
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Junqing Gu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
3
|
Xie H, Guo L, Ma Q, Zhang W, Yang Z, Wang Z, Peng S, Wang K, Wen S, Shang Z, Niu Y. YAP is required for prostate development, regeneration, and prostate stem cell function. Cell Death Discov 2023; 9:339. [PMID: 37689711 PMCID: PMC10492789 DOI: 10.1038/s41420-023-01637-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Prostate development and regeneration depend on prostate stem cell function, the delicate balance of stem cell self-renewal and differentiation. However, mechanisms modulating prostate stem cell function remain poorly identified. Here, we explored the roles of Yes-associated protein 1 (YAP) in prostate stem cells, prostate development and regeneration. Using YAPfl/fl, CD133-CreER mice, we found that stem cell-specific YAP-deficient mice had compromised branching morphogenesis and epithelial differentiation, resulting in damaged prostate development. YAP inhibition also significantly affected the regeneration process of mice prostate, leading to impaired regenerated prostate. Furthermore, YAP ablation in prostate stem cells significantly reduced its self-renewal activity in vitro, and attenuated prostate regeneration of prostate grafts in vivo. Further analysis revealed a decrease in Notch and Hedgehog pathways expression in YAP inhibition cells, and treatment with exogenous Shh partially restored the self-renewal ability of prostate sphere cells. Taken together, our results revealed the roles of YAP in prostate stem cell function and prostate development and regeneration through regulation of the Notch and Hedgehog signaling pathways.
Collapse
Affiliation(s)
- Hui Xie
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Linpei Guo
- Gene and Immunotherapy Center, The Second Hospital of Shandong University, 250033, Jinan, Shandong, China
| | - Qianwang Ma
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Wenyi Zhang
- Department of Radiology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhao Yang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhun Wang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Shuanghe Peng
- Department of Pathology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Keruo Wang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Simeng Wen
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China.
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China.
| |
Collapse
|
4
|
Zhou Z, Jia D, Kwon O, Li S, Sun H, Roudier MP, Lin DW, True L, Morrissey C, Creighton CJ, Lee JK, Xin L. Androgen-regulated stromal complement component 7 (C7) suppresses prostate cancer growth. Oncogene 2023; 42:2428-2438. [PMID: 37400528 PMCID: PMC10802183 DOI: 10.1038/s41388-023-02759-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023]
Abstract
The complement system is a major component of the innate immune system that works through the cytolytic effect of the membrane attack complex (MAC). Complement component 7 (C7) is essential for MAC assembly and its precisely regulated expression level is crucial for the cytolytic activity of MAC. We show that C7 is specifically expressed by the stromal cells in both mouse and human prostates. The expression level of C7 inversely correlates with clinical outcomes in prostate cancer. C7 is positively regulated by androgen signaling in the mouse prostate stromal cells. The androgen receptor directly transcriptionally regulates the mouse and human C7. Increasing C7 expression in the C57Bl/6 syngeneic RM-1 and Pten-Kras allografts suppresses tumor growth in vivo. Conversely, C7 haploinsufficiency promotes tumor growth in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Interestingly, replenishing C7 in androgen-sensitive Pten-Kras tumors during androgen depletion only slightly enhances cellular apoptosis, highlighting the diverse mechanisms employed by tumors to counteract complement activity. Collectively, our research indicates that augmenting complement activity could be a promising therapeutic approach to impede the development of castration resistance in prostate cancer.
Collapse
Affiliation(s)
- Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Deyong Jia
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Ohjoon Kwon
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Shan Li
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Huiyun Sun
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Daniel W Lin
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Lawrence True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - John K Lee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Martinez MAQ, Mullarkey AA, Yee C, Zhao CZ, Zhang W, Shen K, Matus DQ. Reevaluating the relationship between EGL-43 (EVI1) and LIN-12 (Notch) during C. elegans anchor cell invasion. Biol Open 2022; 11:bio059668. [PMID: 36445013 PMCID: PMC9751802 DOI: 10.1242/bio.059668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022] Open
Abstract
Development of the Caenorhabditis elegans reproductive tract is orchestrated by the anchor cell (AC). This occurs in part through a cell invasion event that connects the uterine and vulval tissues. Several key transcription factors regulate AC invasion, such as EGL-43, HLH-2, and NHR-67. Specifically, these transcription factors function together to maintain the post-mitotic state of the AC, a requirement for AC invasion. Recently, a mechanistic connection has been made between loss of EGL-43 and AC cell-cycle entry. The current model states that EGL-43 represses LIN-12 (Notch) expression to prevent AC proliferation, suggesting that Notch signaling has mitogenic effects in the invasive AC. To reexamine the relationship between EGL-43 and LIN-12, we first designed and implemented a heterologous co-expression system called AIDHB that combines the auxin-inducible degron (AID) system of plants with a live cell-cycle sensor based on human DNA helicase B (DHB). After validating AIDHB using AID-tagged GFP, we sought to test it by using AID-tagged alleles of egl-43 and lin-12. Auxin-induced degradation of either EGL-43 or LIN-12 resulted in the expected AC phenotypes. Lastly, we seized the opportunity to pair AIDHB with RNAi to co-deplete LIN-12 and EGL-43, respectively, which revealed that LIN-12 is not required for AC proliferation following loss of EGL-43.
Collapse
Affiliation(s)
- Michael A. Q. Martinez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Angelina A. Mullarkey
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Callista Yee
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Chris Z. Zhao
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Kang Shen
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - David Q. Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
6
|
Jia D, Zhou Z, Kwon OJ, Zhang L, Wei X, Zhang Y, Yi M, Roudier MP, Regier MC, Dumpit R, Nelson PS, Headley M, True L, Lin DW, Morrissey C, Creighton CJ, Xin L. Stromal FOXF2 suppresses prostate cancer progression and metastasis by enhancing antitumor immunity. Nat Commun 2022; 13:6828. [PMID: 36369237 PMCID: PMC9652358 DOI: 10.1038/s41467-022-34665-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) mediate an immunosuppressive effect, but the underlying mechanism remains incompletely defined. Here we show that increasing prostatic stromal Foxf2 suppresses the growth and progression of both syngeneic and autochthonous mouse prostate cancer models in an immunocompetent context. Mechanistically, Foxf2 moderately attenuates the CAF phenotype and transcriptionally downregulates Cxcl5, which diminish the immunosuppressive myeloid cells and enhance T cell cytotoxicity. Increasing prostatic stromal Foxf2 sensitizes prostate cancer to the immune checkpoint blockade therapies. Augmenting lung stromal Foxf2 also mediates an immunosuppressive milieu and inhibits lung colonization of prostate cancer. FOXF2 is expressed higher in the stroma of human transition zone (TZ) than peripheral zone (PZ) prostate. The stromal FOXF2 expression level in primary prostate cancers inversely correlates with the Gleason grade. Our study establishes Foxf2 as a stromal transcription factor modulating the tumor immune microenvironment and potentially explains why cancers are relatively rare and indolent in the TZ prostate.
Collapse
Affiliation(s)
- Deyong Jia
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Xing Wei
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Mingyang Yi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Mary C Regier
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Ruth Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark Headley
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Daniel W Lin
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Ma M, He W, Zhao K, Xue L, Xia S, Zhang B. Targeting aldehyde dehydrogenase for prostate cancer therapies. Front Oncol 2022; 12:1006340. [PMID: 36300093 PMCID: PMC9589344 DOI: 10.3389/fonc.2022.1006340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men in the United States. About 10 – 20% of PCa progress to castration-resistant PCa (CRPC), which is accompanied by metastasis and therapeutic resistance. Aldehyde dehydrogenase (ALDH) is famous as a marker of cancer stem-like cells in different cancer types, including PCa. Generally, ALDHs catalyze aldehyde oxidation into less toxic carboxylic acids and give cancers a survival advantage by reducing oxidative stress caused by aldehyde accumulation. In PCa, the expression of ALDHs is associated with a higher tumor stage and more lymph node metastasis. Functionally, increased ALDH activity makes PCa cells gain more capabilities in self-renewal and metastasis and reduces the sensitivity to castration and radiotherapy. Therefore, it is promising to target ALDH or ALDHhigh cells to eradicate PCa. However, challenges remain in moving the ALDH inhibitors to PCa therapy, potentially due to the toxicity of pan-ALDH inhibitors, the redundancy of ALDH isoforms, and the lack of explicit understanding of the metabolic signaling transduction details. For targeting PCa stem-like cells (PCSCs), different regulators have been revealed in ALDHhigh cells to control cell proliferation and tumorigenicity. ALDH rewires essential signaling transduction in PCa cells. It has been shown that ALDHs produce retinoic acid (RA), bind with androgen, and modulate diverse signaling. This review summarizes and discusses the pathways directly modulated by ALDHs, the crucial regulators that control the activities of ALDHhigh PCSCs, and the recent progress of ALDH targeted therapies in PCa. These efforts will provide insight into improving ALDH-targeted treatment.
Collapse
Affiliation(s)
| | | | | | | | - Siyuan Xia
- *Correspondence: Siyuan Xia, ; Baotong Zhang,
| | | |
Collapse
|
8
|
SOX8 Knockdown Overcomes Enzalutamide Resistance in Castration-Resistant Prostate Cancer by Inhibiting the Notch Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9235837. [PMID: 36246971 PMCID: PMC9560839 DOI: 10.1155/2022/9235837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is still challenging to treat. Dissatisfaction with androgen signal-targeted therapy forces people to look for other treatment strategies. Therefore, this study is aimed at exploring the role of SOX8/Notch signaling in CRPC. The upregulation of SOX8, Notch4, and Hes5 indicated a poor progression-free survival (PFS) in CRPC patients. The expression of these proteins was also upregulated in enzalutamide-resistant LNCaP cells (Enza-R). Moreover, knocking down SOX8 inhibited malignant biological behaviors and decreased the activation of Notch signaling in Enza-R cells. Importantly, knocking down SOX8 obviously reversed the enzalutamide resistance in Enza-R cells, while RO0429097 (a γ secretase inhibitor inactivates Notch signaling) exerted similar effects. At last, we found that both SOX8 knockdown and/or RO0429097 suppressed tumor growth and bone metastasis in vivo. Altogether, our study indicated that the SOX8/Notch signaling is involved in CRPC and that these enzymes are possible targets to develop novel treatment for CRPC.
Collapse
|
9
|
Wei X, Zhang L, Zhang Y, Cooper C, Brewer C, Tsai CF, Wang YT, Glaz M, Wessells HB, Que J, Titus MA, Cirulli V, Glaser A, Liu T, Reder NP, Creighton CJ, Xin L. Ablating Lgr5-expressing prostatic stromal cells activates the ERK-mediated mechanosensory signaling and disrupts prostate tissue homeostasis. Cell Rep 2022; 40:111313. [PMID: 36070687 PMCID: PMC9491244 DOI: 10.1016/j.celrep.2022.111313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/12/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023] Open
Abstract
Functional implication of stromal heterogeneity in the prostate remains incompletely understood. Using lineage tracing and light-sheet imaging, we show that some fibroblast cells at the mouse proximal prostatic ducts and prostatic urethra highly express Lgr5. Genetic ablation of these anatomically restricted stromal cells, but not nonselective ablation of prostatic stromal cells, rapidly induces prostate epithelial turnover and dedifferentiation that are reversed following spontaneous restoration of the Lgr5+ stromal cells. RNA sequencing (RNA-seq) analysis indicates that ablating the Lgr5+ stromal cells activates a mechanosensory response. Ablating the Lgr5+ stromal cells impairs the control of prostatic ductal outlet, increases prostate tissue stiffness, and activates the mitogen-activated protein kinase (MAPK). Suppressing MAPK overrides the elevated epithelial proliferation. In summary, the Lgr5+ stromal cells regulate prostate tissue homeostasis and maintain its functional integrity in a long-distance manner. Our study implies that the cells at organ junctions most likely control organ homeostasis by sustaining a balanced mechanoforce.
Collapse
Affiliation(s)
- Xing Wei
- Department of Urology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Li Zhang
- Department of Urology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cody Cooper
- Alpenglow Biosciences, Inc., Seattle, WA 98103, USA
| | - Chris Brewer
- Alpenglow Biosciences, Inc., Seattle, WA 98103, USA
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Yi-Ting Wang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Micah Glaz
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98109, USA
| | - Hunter B Wessells
- Department of Urology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Mark A Titus
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston TX 77030, USA
| | - Vincenzino Cirulli
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Adam Glaser
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | | | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Xin
- Department of Urology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston TX 77030, USA.
| |
Collapse
|
10
|
Mugisha S, Di X, Disoma C, Jiang H, Zhang S. Fringe family genes and their modulation of Notch signaling in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188746. [PMID: 35660646 DOI: 10.1016/j.bbcan.2022.188746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
Fringes are glycosyltransferases that transfer N-acetylglucosamine to the O-linked fucose of Notch receptors. They regulate the Notch signaling activity that drives tumor formation and progression, resulting in poor prognosis. However, the specific tumor-promoting role of Fringes differs depending on the type of cancer. Although a particular Fringe member could act as a tumor suppressor in one cancer type, it may act as an oncogene in another. This review discusses the tumorigenic role of the Fringe family (lunatic fringe, manic fringe, and radical fringe) in modulating Notch signaling in various cancers. Although the crucial functions of Fringes continue to emerge as more mechanistic studies are being pursued, further translational research is needed to explore their roles and therapeutic benefits in various malignancies.
Collapse
Affiliation(s)
- Samson Mugisha
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Xiaotang Di
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Cyrollah Disoma
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Hao Jiang
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China.
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
11
|
Liu Y, Wang J, Horton C, Yu C, Knudsen B, Stefanson J, Hu K, Stefanson O, Green J, Guo C, Xie Q, Wang ZA. Stromal AR inhibits prostate tumor progression by restraining secretory luminal epithelial cells. Cell Rep 2022; 39:110848. [PMID: 35613593 PMCID: PMC9175887 DOI: 10.1016/j.celrep.2022.110848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/03/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Androgen receptor (AR) is expressed in both the prostate epithelium and the prostate stroma and plays diverse roles in prostate physiology. Although low expression of stromal AR is clinically associated with advanced cancer stage and worse outcome, whether stromal AR inhibits or promotes prostate cancer progression remains controversial. Here, we specifically delete AR in smooth muscle cells of the adult mouse prostate under two tumorigenic conditions, namely, the Hi-Myc genetic model and the T + E2 hormonal carcinogenesis model. Histology analyses show that stromal AR deletion exacerbates tumor progression phenotypes in both models. Furthermore, single-cell analyses of the tumor samples reveal that secretory luminal cells are the cell population particularly affected by stromal AR deletion, as they transition to a cellular state of potentiated PI3K-mTORC1 activities. Our results suggest that stromal AR normally inhibits prostate cancer progression by restraining secretory luminal cells and imply possible unintended negative effects of androgen deprivation therapy.
Collapse
Affiliation(s)
- Yueli Liu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jiawen Wang
- Sequencing Center, National Institute of Biological Sciences, Beijing 102206, China
| | - Corrigan Horton
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Chuan Yu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Beatrice Knudsen
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua Stefanson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ofir Stefanson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jonathan Green
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Charlene Guo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Qing Xie
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
12
|
Wei X, Roudier MP, Kwon OJ, Lee JD, Kong K, Dumpit R, True L, Morrissey C, Lin DW, Nelson PS, Xin L. Paracrine Wnt signaling is necessary for prostate epithelial proliferation. Prostate 2022; 82:517-530. [PMID: 35014711 PMCID: PMC8866211 DOI: 10.1002/pros.24298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 11/07/2022]
Abstract
INTRODUCTION The Wnt proteins play key roles in the development, homeostasis, and disease progression of many organs including the prostate. However, the spatiotemporal expression patterns of Wnt proteins in prostate cell lineages at different developmental stages and in prostate cancer remain inadequately characterized. METHODS We isolated the epithelial and stromal cells in the developing and mature mouse prostate by flow cytometry and determined the expression levels of Wnt ligands. We used Visium spatial gene expression analysis to determine the spatial distribution of Wnt ligands in the mouse prostatic glands. Using laser-capture microscopy in combination with gene expression analysis, we also determined the expression patterns of Wnt signaling components in stromal and cancer cells in advanced human prostate cancer specimens. To investigate how the stroma-derived Wnt ligands affect prostate development and homeostasis, we used a Col1a2-CreERT2 mouse model to disrupt the Wnt transporter Wntless specifically in prostate stromal cells. RESULTS We showed that the prostate stromal cells are a major source of several Wnt ligands. Visium spatial gene expression analysis revealed a distinct spatial distribution of Wnt ligands in the prostatic glands. We also showed that Wnt signaling components are highly expressed in the stromal compartment of primary and advanced human prostate cancer. Blocking stromal Wnt secretion attenuated prostate epithelial proliferation and regeneration but did not affect cell survival and lineage maintenance. DISCUSSION Our study demonstrates a critical role of stroma-derived Wnt ligands in prostate development and homeostasis.
Collapse
Affiliation(s)
- Xing Wei
- Department of Urology, University of Washington, Seattle, WA, USA 98109
| | | | - Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA, USA 98109
| | - Justin Daho Lee
- Molecular Engineering Ph.D. Program, University of Washington, Seattle, WA, USA 98109
- Department of Bioengineering, University of Washington, Seattle, WA, USA 98109
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA 98109
| | - Kevin Kong
- Department of Biology, University of Washington, Seattle, WA, USA 98109
| | - Ruth Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA 98109
| | - Lawrence True
- Department of Urology, University of Washington, Seattle, WA, USA 98109
- Department of Pathology, University of Washington, Seattle, WA, USA 98109
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA 98109
| | - Daniel W. Lin
- Department of Urology, University of Washington, Seattle, WA, USA 98109
| | - Peter S. Nelson
- Department of Urology, University of Washington, Seattle, WA, USA 98109
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA 98109
- Department of Pathology, University of Washington, Seattle, WA, USA 98109
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, USA 98109
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA 98109
| |
Collapse
|
13
|
Kwon OJ, Zhang B, Jia D, Zhang L, Wei X, Zhou Z, Liu D, Huynh KT, Zhang K, Zhang Y, Labhart P, Sboner A, Barbieri C, Haffner MC, Creighton CJ, Xin L. Elevated expression of the colony-stimulating factor 1 (CSF1) induces prostatic intraepithelial neoplasia dependent of epithelial-Gp130. Oncogene 2022; 41:1309-1323. [PMID: 34999736 PMCID: PMC8882147 DOI: 10.1038/s41388-021-02169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/24/2022]
Abstract
Macrophages are increased in human benign prostatic hyperplasia and prostate cancer. We generate a Pb-Csf1 mouse model with prostate-specific overexpression of macrophage colony-stimulating factor (M-Csf/Csf1). Csf1 overexpression promotes immune cell infiltration into the prostate, modulates the macrophage polarity in a lobe-specific manner, and induces senescence and low-grade prostatic intraepithelial neoplasia (PIN). The Pb-Csf1 prostate luminal cells exhibit increased stem cell features and undergo an epithelial-to-mesenchymal transition. Human prostate cancer patients with high CSF-1 expression display similar transcriptional alterations with the Pb-Csf1 model. P53 knockout alleviates senescence but fails to progress PIN lesions. Ablating epithelial Gp130 but not Il1r1 substantially blocks PIN lesion formation. The androgen receptor (AR) is downregulated in Pb-Csf1 mice. ChIP-Seq analysis reveals altered AR binding in 2482 genes although there is no significant widespread change in global AR transcriptional activity. Collectively, our study demonstrates that increased macrophage infiltration causes PIN formation but fails to transform prostate cells.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Deyong Jia
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Xing Wei
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Deli Liu
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Khoi Trung Huynh
- Department of Biology, University of Washington, Seattle, WA, 98109, USA
| | - Kai Zhang
- Department of Urology, University of Washington, Seattle, WA, 98109, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Andrea Sboner
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Chris Barbieri
- Sandra and Edward Meyer Cancer Center and Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Michael C Haffner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, 98109, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
14
|
Liu S, Hsu EC, Shen M, Aslan M, Stoyanova T. Metastasis Model to Test the Role of Notch Signaling in Prostate Cancer. Methods Mol Biol 2022; 2472:221-233. [PMID: 35674904 DOI: 10.1007/978-1-0716-2201-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Distant metastasis is the main cause of death in prostate cancer patients. Notch signaling plays an important role in driving prostate cancer aggressiveness and metastasis. In this chapter, we describe a protocol to measure prostate cancer metastatic colonization, incidences of metastasis, accurately quantify the burden of metastasis, and test the role of NOTCH1 receptor on prostate cancer metastatic colonization and homing to distant sites. The metastasis model presented here is established by intracardiac injection of control human prostate cancer cells and NOTCH1 downregulated cells. The cells are engineered to express both red fluorescent protein (RFP) and luciferase. In this model, whole body bioluminescence imaging, high-resolution, and quantitative fluorescence imaging are utilized for quantitative assessment of metastatic colonization and metastasis burden. Further, histopathology analyses of diverse metastatic organs are performed. This model is a powerful and versatile tool to investigate the mechanisms underlying the function of NOTCH receptors in metastatic colonization in prostate cancer.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Michelle Shen
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Interruption of Klf5 acetylation in basal progenitor cells promotes luminal commitment by activating Notch signaling. J Genet Genomics 2021; 49:579-582. [PMID: 34952235 DOI: 10.1016/j.jgg.2021.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
|
16
|
He J, Zhang X, Xia X, Han M, Li F, Li C, Li Y, Gao D. Organoid technology for tissue engineering. J Mol Cell Biol 2021; 12:569-579. [PMID: 32249317 PMCID: PMC7683016 DOI: 10.1093/jmcb/mjaa012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/11/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
For centuries, attempts have been continuously made to artificially reconstitute counterparts of in vivo organs from their tissues or cells. Only in the recent decade has organoid technology as a whole technological field systematically emerged and been shown to play important roles in tissue engineering. Based on their self-organizing capacities, stem cells of versatile organs, both harvested and induced, can form 3D structures that are structurally and functionally similar to their in vivo counterparts. These organoid models provide a powerful platform for elucidating the development mechanisms, modeling diseases, and screening drug candidates. In this review, we will summarize the advances of this technology for generating various organoids of tissues from the three germ layers and discuss their drawbacks and prospects for tissue engineering.
Collapse
Affiliation(s)
- Juan He
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoyu Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyi Xia
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming Han
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunfeng Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
17
|
Rai S, Alsaidan OA, Yang H, Cai H, Wang L. Heparan sulfate inhibits transforming growth factor β signaling and functions in cis and in trans to regulate prostate stem/progenitor cell activities. Glycobiology 2021; 30:381-395. [PMID: 31829419 DOI: 10.1093/glycob/cwz103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate stem/progenitor cells (PrSCs) are responsible for adult prostate tissue homeostasis and regeneration. However, the related regulatory mechanisms are not completely understood. In this study, we examined the role of heparan sulfate (HS) in PrSC self-renewal and prostate regeneration. Using an in vitro prostate sphere formation assay, we found that deletion of the glycosyltransferase exostosin 1 (Ext1) abolished HS expression in PrSCs and disrupted their ability to self-renew. In associated studies, we observed that HS loss inhibited p63 and CK5 expression, reduced the number of p63+- or CK5+-expressing stem/progenitor cells, elevated CK8+ expression and the number of differentiated CK8+ luminal cells and arrested the spheroid cells in the G1/G0 phase of cell cycle. Mechanistically, HS expressed by PrSCs (in cis) or by neighboring cells (in trans) could maintain sphere formation. Furthermore, HS deficiency upregulated transforming growth factor β (TGFβ) signaling and inhibiting TGFβ signaling partially restored the sphere-formation activity of the HS-deficient PrSCs. In an in vivo prostate regeneration assay, simultaneous loss of HS in both epithelial cell and stromal cell compartments attenuated prostate tissue regeneration, whereas the retention of HS expression in either of the two cellular compartments was sufficient to sustain prostate tissue regeneration. We conclude that HS preserves self-renewal of adult PrSCs by inhibiting TGFβ signaling and functions both in cis and in trans to maintain prostate homeostasis and to support prostate regeneration.
Collapse
Affiliation(s)
- Sumit Rai
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Omar Awad Alsaidan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| | - Hua Yang
- Department of Molecular Pharmacology and Physiology, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Houjian Cai
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| | - Lianchun Wang
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA.,Department of Molecular Pharmacology and Physiology, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| |
Collapse
|
18
|
Xia L, Yang F, Wu X, Li S, Kan C, Zheng H, Wang S. SHP2 inhibition enhances the anticancer effect of Osimertinib in EGFR T790M mutant lung adenocarcinoma by blocking CXCL8 loop mediated stemness. Cancer Cell Int 2021; 21:337. [PMID: 34217295 PMCID: PMC8254369 DOI: 10.1186/s12935-021-02056-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/27/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Additional epidermal growth factor receptor (EGFR) mutations confer the drug resistance to generations of EGFR targeted tyrosine kinase inhibitor (EGFR-TKI), posing a major challenge to developing effective treatment of lung adenocarcinoma (LUAD). The strategy of combining EGFR-TKI with other synergistic or sensitizing therapeutic agents are considered a promising approach in the era of precision medicine. Moreover, the role and mechanism of SHP2, which is involved in cell proliferation, cytokine production, stemness maintenance and drug resistance, has not been carefully explored in lung adenocarcinoma (LUAD). METHODS To evaluate the impact of SHP2 on the efficacy of EGFR T790M mutant LUAD cells to Osimertinib, SHP2 inhibition was tested in Osimertinib treated LUAD cells. Cell proliferation and stemness were tested in SHP2 modified LUAD cells. RNA sequencing was performed to explore the mechanism of SHP2 promoted stemness. RESULTS This study demonstrated that high SHP2 expression level correlates with poor outcome of LUAD patients, and SHP2 expression is enriched in Osimertinib resistant LUAD cells. SHP2 inhibition suppressed the cell proliferation and damaged the stemness of EGFR T790M mutant LUAD. SHP2 facilitates the secretion of CXCL8 cytokine from the EGFR T790M mutant LUAD cells, through a CXCL8-CXCR1/2 positive feedback loop that promotes stemness and tumorigenesis. Our results further show that SHP2 mediates CXCL8-CXCR1/2 feedback loop through ERK-AKT-NFκB and GSK3β-β-Catenin signaling in EGFR T790M mutant LUAD cells. CONCLUSIONS Our data revealed that SHP2 inhibition enhances the anti-cancer effect of Osimertinib in EGFR T790M mutant LUAD by blocking CXCL8-CXCR1/2 loop mediated stemness, which may help provide an alternative therapeutic option to enhance the clinical efficacy of osimertinib in EGFR T790M mutant LUAD patients.
Collapse
Affiliation(s)
- Leiming Xia
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
- Department of Hematology, The Third affiliated hospital of Anhui Medical University, Hefei, China
- Department of Hematology, The fourth affiliated hospital of Anhui Medical University, Hefei, China
| | - Fan Yang
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
| | - Xiao Wu
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
| | - Suzhi Li
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
| | - Chen Kan
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
| | - Hong Zheng
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China
| | - Siying Wang
- Basic College of Medicine, Anhui Medical University, 81 Meishan road, Hefei, Anhui, China.
- Laboratory Center for Medical Science Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
19
|
Huang S, Yu F, Cheng Y, Li Y, Chen Y, Tang J, Bei Y, Tang Q, Zhao Y, Huang Y, Xiang Q. Transforming Growth Factor-β3/Recombinant Human-like Collagen/Chitosan Freeze-Dried Sponge Primed With Human Periodontal Ligament Stem Cells Promotes Bone Regeneration in Calvarial Defect Rats. Front Pharmacol 2021; 12:678322. [PMID: 33967817 PMCID: PMC8103166 DOI: 10.3389/fphar.2021.678322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with a skull defect are at risk of developing cerebrospinal fluid leakage and ascending bacterial meningitis at >10% per year. However, treatment with stem cells has brought great hope to large-area cranial defects. Having found that transforming growth factor (TGF)-β3 can promote the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs), we designed a hybrid TGF-β3/recombinant human-like collagen recombinant human collagen/chitosan (CS) freeze-dried sponge (TRFS) loading hPDLSCs (TRFS-h) to repair skull defects in rats. CFS with 2% CS was selected based on the swelling degree, water absorption, and moisture retention. The CS freeze-dried sponge (CFS) formed a porous three-dimensional structure, as observed by scanning electron microscopy. In addition, cytotoxicity experiments and calcein-AM/PI staining showed that TRFS had a good cellular compatibility and could be degraded completely at 90 days in the implantation site. Furthermore, bone healing was evaluated using micro-computed tomography in rat skull defect models. The bone volume and bone volume fraction were higher in TRFS loaded with hPDLSCs (TRFS-h) group than in the controls (p < 0.01, vs. CFS or TRFS alone). The immunohistochemical results indicated that the expression of Runx2, BMP-2, and collagen-1 (COL Ⅰ) in cells surrounding bone defects in the experimental group was higher than those in the other groups (p < 0.01, vs. CFS or TRFS alone). Taken together, hPDLSCs could proliferate and undergo osteogenic differentiation in TRFS (p < 0.05), and TRFS-h accelerated bone repair in calvarial defect rats. Our research revealed that hPDLSCs could function as seeded cells for skull injury, and their osteogenic differentiation could be accelerated by TGF-β3. This represents an effective therapeutic strategy for restoring traumatic defects of the skull.
Collapse
Affiliation(s)
- Shiyi Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Fenglin Yu
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Yating Cheng
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Yangfan Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Yini Chen
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Jianzhong Tang
- Biopharmaceutical R and D Center of Jinan University, Guangzhou, China
| | - Yu Bei
- Biopharmaceutical R and D Center of Jinan University, Guangzhou, China
| | - Qingxia Tang
- Department of Stomatology, Jinan University Medical College, Guangzhou, China
| | - Yueping Zhao
- Department of Stomatology, Jinan University Medical College, Guangzhou, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,Biopharmaceutical R and D Center of Jinan University, Guangzhou, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,Biopharmaceutical R and D Center of Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Du Z, Li L, Sun W, Zhu P, Cheng S, Yang X, Luo C, Yu X, Wu X. Systematic Evaluation for the Influences of the SOX17/Notch Receptor Family Members on Reversing Enzalutamide Resistance in Castration-Resistant Prostate Cancer Cells. Front Oncol 2021; 11:607291. [PMID: 33791203 PMCID: PMC8006330 DOI: 10.3389/fonc.2021.607291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
The treatment of castration-resistant prostate cancer (CRPC) remains challenging due to the failure of androgen deprivation therapy (ADT); hence the search for other molecular therapeutic targets besides androgen receptor signaling is ongoing. This study systematically investigated the expression of SOX17 and Notch receptors in CRPC tissues and cells in vitro, showing that consistent clinical CRPC, SOX17/Notch1, and Notch4 were responsible for enzalutamide resistance in CRPC cells. The γ secretase inhibitors, BMS-708163, GSI-IX, PF-3084014, and RO4929097 abrogated the enzalutamide resistance by inhibiting Notch1 or/and Notch4 in vitro, with GSI-IX and RO4929097 being more effective than BMS-708163 and PF-3084014 in reliving bone metastasis in vivo. In conclusion, the Notch1 and Notch4 inhibitors GSI-IX and RO4929097 are promising therapeutic agents for the treatment of CRPC.
Collapse
Affiliation(s)
- Zhongbo Du
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China.,Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Urology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Luo Li
- Center for Immunology Research, Chongqing Medical University, Chongqing, China
| | - Wei Sun
- Department of Urology, Fuling Center Hospital of Chongqing, Chongqing, China
| | - Pingyu Zhu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China.,Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shulin Cheng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China.,Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuesong Yang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Chunli Luo
- Key Laboratory of Diagnostics Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xiaodong Yu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China.,Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaohou Wu
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Kwon OJ, Choi JM, Zhang L, Jia D, Li Z, Zhang Y, Jung SY, Creighton CJ, Xin L. The Sca-1 + and Sca-1 - mouse prostatic luminal cell lineages are independently sustained. Stem Cells 2020; 38:1479-1491. [PMID: 32627901 DOI: 10.1002/stem.3253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
The phenotypic and functional heterogeneity of the mouse prostate epithelial cell lineages remains incompletely characterized. We show that the Sca-1+ luminal cells at the mouse proximal prostate express Sox2. These cells are replicative quiescent, castration resistant, and do not possess secretory function. We use the Probasin-CreERT2 and Sox2-CreERT2 models in concert with a fluorescent reporter line to label the Sca-1- and Sca-1+ luminal cells, respectively. By a lineage tracing approach, we show that the two luminal cell populations are independently sustained. Sox2 is dispensable for the maintenance of the Sca-1+ luminal cells but is essential for their facultative bipotent differentiation capacity. The Sca-1+ luminal cells share molecular features with the human TACSTD2+ luminal cells. This study corroborates the heterogeneity of the mouse prostate luminal cell lineage and shows that the adult mouse prostate luminal cell lineage is maintained by distinct cellular entities rather than a single progenitor population.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Jong Min Choi
- Department of Chemistry and Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Deyong Jia
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Zhouyihan Li
- Department of Chemistry and Biochemistry, University of Washington, Seattle, Washington, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Sung Yun Jung
- Department of Chemistry and Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, Washington, USA.,Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
22
|
Otani Y, Yoo JY, Chao S, Liu J, Jaime-Ramirez AC, Lee TJ, Hurwitz B, Yan Y, Dai H, Glorioso JC, Caligiuri MA, Yu J, Kaur B. Oncolytic HSV-Infected Glioma Cells Activate NOTCH in Adjacent Tumor Cells Sensitizing Tumors to Gamma Secretase Inhibition. Clin Cancer Res 2020; 26:2381-2392. [PMID: 32139403 PMCID: PMC7325527 DOI: 10.1158/1078-0432.ccr-19-3420] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/20/2019] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE To examine the effect of oncolytic herpes simplex virus (oHSV) on NOTCH signaling in central nervous system tumors. EXPERIMENTAL DESIGN Bioluminescence imaging, reverse phase protein array proteomics, fluorescence microscopy, reporter assays, and molecular biology approaches were used to evaluate NOTCH signaling. Orthotopic glioma-mouse models were utilized to evaluate effects in vivo. RESULTS We have identified that herpes simplex virus-1 (HSV-1; oncolytic and wild-type)-infected glioma cells induce NOTCH signaling, from inside of infected cells into adjacent tumor cells (inside out signaling). This was canonical NOTCH signaling, which resulted in activation of RBPJ-dependent transcriptional activity that could be rescued with dnMAML. High-throughput screening of HSV-1-encoded cDNA and miRNA libraries further uncovered that HSV-1 miR-H16 induced NOTCH signaling. We further identified that factor inhibiting HIF-1 (FIH-1) is a direct target of miR-H16, and that FIH-1 downregulation by virus encoded miR-H16 induces NOTCH activity. FIH-1 binding to Mib1 has been reported, but this is the first report that shows FIH-1 sequester Mib1 to suppress NOTCH activation. We observed that FIH-1 degradation induced NOTCH ligand ubiquitination and NOTCH activity. REMBRANDT and The Cancer Genome Atlas data analysis also uncovered a significant negative regulation between FIH-1 and NOTCH. Furthermore, combination of oHSV with NOTCH-blocking gamma secretase inhibitor (GSI) had a therapeutic advantage in two different intracranial glioma models treated with oncolytic HSV, without affecting safety profile of the virus in vivo. CONCLUSIONS To our knowledge this is the first report to identify impact of HSV-1 on NOTCH signaling and highlights the significance of combining oHSV and GSI for glioblastoma therapy.
Collapse
Affiliation(s)
- Yoshihiro Otani
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ji Young Yoo
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Samantha Chao
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
- Rice University, Houston, Texas
| | - Joseph Liu
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Alena Cristina Jaime-Ramirez
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Tae Jin Lee
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Brian Hurwitz
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York
| | - Yuanqing Yan
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Hongsheng Dai
- City of Hope National Medical Center, Duarte, California
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Jianhua Yu
- City of Hope National Medical Center, Duarte, California
| | - Balveen Kaur
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
23
|
Klf5 acetylation regulates luminal differentiation of basal progenitors in prostate development and regeneration. Nat Commun 2020; 11:997. [PMID: 32081850 PMCID: PMC7035357 DOI: 10.1038/s41467-020-14737-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/20/2019] [Indexed: 12/28/2022] Open
Abstract
Prostate development depends on balanced cell proliferation and differentiation, and acetylated KLF5 is known to alter epithelial proliferation. It remains elusive whether post-translational modifications of transcription factors can differentially determine adult stem/progenitor cell fate. Here we report that, in human and mouse prostates, Klf5 is expressed in both basal and luminal cells, with basal cells preferentially expressing acetylated Klf5. Functionally, Klf5 is indispensable for maintaining basal progenitors, their luminal differentiation, and the proliferation of their basal and luminal progenies. Acetylated Klf5 is also essential for basal progenitors' maintenance and proper luminal differentiation, as deacetylation of Klf5 causes excess basal-to-luminal differentiation; attenuates androgen-mediated organoid organization; and retards postnatal prostate development. In basal progenitor-derived luminal cells, Klf5 deacetylation increases their proliferation and attenuates their survival and regeneration following castration and subsequent androgen restoration. Mechanistically, Klf5 deacetylation activates Notch signaling. Klf5 and its acetylation thus contribute to postnatal prostate development and regeneration by controlling basal progenitor cell fate.
Collapse
|
24
|
Xiong X, Schober M, Tassone E, Khodadadi-Jamayran A, Sastre-Perona A, Zhou H, Tsirigos A, Shen S, Chang M, Melamed J, Ossowski L, Wilson EL. KLF4, A Gene Regulating Prostate Stem Cell Homeostasis, Is a Barrier to Malignant Progression and Predictor of Good Prognosis in Prostate Cancer. Cell Rep 2019; 25:3006-3020.e7. [PMID: 30540935 PMCID: PMC6405286 DOI: 10.1016/j.celrep.2018.11.065] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 12/18/2022] Open
Abstract
There is a considerable need to identify those individuals with prostate cancer who have indolent disease. We propose that
genes that control adult stem cell homeostasis in organs with slow turnover, such as the prostate, control cancer fate. One such
gene, KLF4, overexpressed in murine prostate stem cells, regulates their homeostasis, blocks malignant transformation, and
controls the self-renewal of tumor-initiating cells. KLF4 loss induces the molecular features of aggressive cancer and converts
PIN lesions to invasive sarcomatoid carcinomas; its re-expression in vivo reverses this process. Bioinformatic
analysis links these changes to human cancer. KLF4 and its downstream targets make up a gene signature that identifies indolent
tumors and predicts recurrence-free survival. This approach may improve prognosis and identify therapeutic targets for advanced
cancer. Available criteria for segregating prostate cancer patients into those requiring therapeutic intervention and those who can
be followed are inadequate. Xiong et al. show that KLF4 and its downstream targets make up a gene signature that identifies
indolent tumors. This approach may improve prognosis and identify therapeutic targets for advanced cancer.
Collapse
Affiliation(s)
- Xiaozhong Xiong
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| | - Markus Schober
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA; Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Evelyne Tassone
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Ana Sastre-Perona
- Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Hua Zhou
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Steven Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Miao Chang
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jonathan Melamed
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Liliana Ossowski
- Department of Medicine, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Elaine L Wilson
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA; Department of Urology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
25
|
Gordeeva O. TGFβ Family Signaling Pathways in Pluripotent and Teratocarcinoma Stem Cells' Fate Decisions: Balancing Between Self-Renewal, Differentiation, and Cancer. Cells 2019; 8:cells8121500. [PMID: 31771212 PMCID: PMC6953027 DOI: 10.3390/cells8121500] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
The transforming growth factor-β (TGFβ) family factors induce pleiotropic effects and are involved in the regulation of most normal and pathological cellular processes. The activity of different branches of the TGFβ family signaling pathways and their interplay with other signaling pathways govern the fine regulation of the self-renewal, differentiation onset and specialization of pluripotent stem cells in various cell derivatives. TGFβ family signaling pathways play a pivotal role in balancing basic cellular processes in pluripotent stem cells and their derivatives, although disturbances in their genome integrity induce the rearrangements of signaling pathways and lead to functional impairments and malignant transformation into cancer stem cells. Therefore, the identification of critical nodes and targets in the regulatory cascades of TGFβ family factors and other signaling pathways, and analysis of the rearrangements of the signal regulatory network during stem cell state transitions and interconversions, are key issues for understanding the fundamental mechanisms of both stem cell biology and cancer initiation and progression, as well as for clinical applications. This review summarizes recent advances in our understanding of TGFβ family functions in naїve and primed pluripotent stem cells and discusses how these pathways are involved in perturbations in the signaling network of malignant teratocarcinoma stem cells with impaired differentiation potential.
Collapse
Affiliation(s)
- Olga Gordeeva
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334 Moscow, Russia
| |
Collapse
|
26
|
Horton C, Liu Y, Yu C, Xie Q, Wang ZA. Luminal-contact-inhibition of epithelial basal stem cell multipotency in prostate organogenesis and homeostasis. Biol Open 2019; 8:bio.045724. [PMID: 31540905 PMCID: PMC6826291 DOI: 10.1242/bio.045724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prostate epithelial basal cells are highly plastic in their luminal differentiation capability. Basal stem cells actively produce luminal cells during organogenesis, but become restricted in the adult prostate unless receiving oncogenic or inflammatory stimuli. Given that the number of luminal cells increases relative to basal cells through development and that equilibrium is reached in the adulthood, we hypothesize that a negative-feedback mechanism exists to inhibit basal-to-luminal differentiation. We provide evidence supporting this hypothesis by comparing murine prostatic growth in a tissue reconstitution assay with cell recombinants of different basal-to-luminal ratios. Additionally, in organoid culture, hybrid organoids derived from adjacent basal and luminal cells showed reduced basal stem cell activities, suggesting contact inhibition. Importantly, removal of adult luminal cells in vivo via either an inducible Cre/loxP-Dre/rox dual-lineage-tracing system or orthotopic trypsin injection led to robust reactivation of basal stem cell activities, which acts independent of androgen. These data illustrate the prostate organ as a distinctive paradigm where cell contact from differentiated daughter cells restricts adult stem cell multipotency to maintain the steady-state epithelial architecture.
Collapse
Affiliation(s)
- Corrigan Horton
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Yueli Liu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Chuan Yu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Qing Xie
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
27
|
Jiao M, Qi M, Zhang F, Hu J, Feng T, Zhao M, Li X, Liu H, Teng W, Zhang J, Liu Z, Zhang L, Wu Z, Han B. CUL4B regulates cancer stem-like traits of prostate cancer cells by targeting BMI1 via miR200b/c. Prostate 2019; 79:1294-1303. [PMID: 31111526 DOI: 10.1002/pros.23835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cancer stem-like traits contribute to prostate cancer (PCa) progression and metastasis. Cullin 4B (CUL4B) is a member of the ubiquitin E3 ligase family and overexpressed in several solid malignancies including PCa. CUL4B has been suggested to be an oncogene through epigenetic repression of tumor suppressors. However, the link between CUL4B expression and cancer stem-like phenotype remains unclear. METHODS Western blot analysis, sphere formation, and colony formation assays were used to examine the effect of CUL4B on cancer stem-like traits in PCa cells. Mechanically, bioinformatic analysis was utilized to evaluate whether BMI1 was a target of CUL4B. Moreover, real-time polymerase chain reaction, chromatin immunoprecipitation, and luciferase reporter assays were performed to identify microRNAs regulated by CUL4B. Finally, Western blot assay was used to validate the regulation of CUL4B, miR200b, and miR200c (miR200b/c) on the stem-like characteristics of PCa cells. RESULTS CUL4B promotes PCa pluripotency-associated markers expression, sphere formation, and anchorage-independent growth ability in vitro. Mechanically, CUL4B upregulates BMI1 expression via epigenetically repressing miR200b/c expression. In addition, miR200b/c could partially reverse CUL4B-induced BMI1 and pluripotency-associated marker expression. CONCLUSIONS Our study revealed that CUL4B regulates cancer stem-like traits of prostate cancer cells by targeting BMI1 via miR200b/c, which might give novel insight into how CUL4B promotes PCa progression through regulating cancer stem-like traits.
Collapse
Affiliation(s)
- Meng Jiao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Second Hospital of Shandong University, Jinan, China
| | - Mei Qi
- Department of Pathology, Shandong University Qilu Hospital, Jinan, China
| | - Facai Zhang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Mingfeng Zhao
- Department of Pathology, Binzhou Medical University, Binzhou, China
| | - Xinjun Li
- Department of Pathology, Binzhou People's Hospital, Binzhou, China
| | - Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wei Teng
- Education Quality Management Office, Institute of Continuing Education, Shandong University, Jinan, China
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhiyan Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Shandong University Qilu Hospital, Jinan, China
| | - Lili Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhen Wu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Shandong University Qilu Hospital, Jinan, China
| |
Collapse
|
28
|
Genz B, Coleman MA, Irvine KM, Kutasovic JR, Miranda M, Gratte FD, Tirnitz-Parker JEE, Olynyk JK, Calvopina DA, Weis A, Cloonan N, Robinson H, Hill MM, Al-Ejeh F, Ramm GA. Overexpression of miRNA-25-3p inhibits Notch1 signaling and TGF-β-induced collagen expression in hepatic stellate cells. Sci Rep 2019; 9:8541. [PMID: 31189969 PMCID: PMC6561916 DOI: 10.1038/s41598-019-44865-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
During chronic liver injury hepatic stellate cells (HSCs), the principal source of extracellular matrix in the fibrotic liver, transdifferentiate into pro-fibrotic myofibroblast-like cells - a process potentially regulated by microRNAs (miRNAs). Recently, we found serum miRNA-25-3p (miR-25) levels were upregulated in children with Cystic Fibrosis (CF) without liver disease, compared to children with CF-associated liver disease and healthy individuals. Here we examine the role of miR-25 in HSC biology. MiR-25 was detected in the human HSC cell line LX-2 and in primary murine HSCs, and increased with culture-induced activation. Transient overexpression of miR-25 inhibited TGF-β and its type 1 receptor (TGFBR1) mRNA expression, TGF-β-induced Smad2 phosphorylation and subsequent collagen1α1 induction in LX-2 cells. Pull-down experiments with biotinylated miR-25 revealed Notch signaling (co-)activators ADAM-17 and FKBP14 as miR-25 targets in HSCs. NanoString analysis confirmed miR-25 regulation of Notch- and Wnt-signaling pathways. Expression of Notch signaling pathway components and endogenous Notch1 signaling was downregulated in miR-25 overexpressing LX-2 cells, as were components of Wnt signaling such as Wnt5a. We propose that miR-25 acts as a negative feedback anti-fibrotic control during HSC activation by reducing the reactivity of HSCs to TGF-β-induced collagen expression and modulating the cross-talk between Notch, Wnt and TGF-β signaling.
Collapse
Affiliation(s)
- Berit Genz
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Mater Research, Translational Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Miranda A Coleman
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research, Translational Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Jamie R Kutasovic
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mariska Miranda
- Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Francis D Gratte
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
| | - Janina E E Tirnitz-Parker
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - John K Olynyk
- Department of Gastroenterology & Hepatology, Fiona Stanley Fremantle Hospital Group, Murdoch, Western Australia, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Diego A Calvopina
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anna Weis
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nicole Cloonan
- Genomic Biology Lab, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Harley Robinson
- Precision & Systems Biomedicine, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle M Hill
- Precision & Systems Biomedicine, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Fares Al-Ejeh
- Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia. .,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
29
|
Rice MA, Hsu EC, Aslan M, Ghoochani A, Su A, Stoyanova T. Loss of Notch1 Activity Inhibits Prostate Cancer Growth and Metastasis and Sensitizes Prostate Cancer Cells to Antiandrogen Therapies. Mol Cancer Ther 2019; 18:1230-1242. [PMID: 31028097 DOI: 10.1158/1535-7163.mct-18-0804] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/29/2018] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Prostate cancer remains among the leading causes of cancer-related deaths in men. Patients with aggressive disease typically undergo hormone deprivation therapy. Although treatment is initially very successful, these men commonly progress to lethal, castration-resistant prostate cancer (CRPC) in 2 to 3 years. Standard therapies for CRPC include second-generation antiandrogens, which prolong patient lifespan by only several months. It is imperative to advance our understanding of the mechanisms leading to resistance to identify new therapies for aggressive prostate cancer. This study identifies Notch1 as a therapeutic target in prostate cancer. Loss of NOTCH1 in aggressive prostate cancer cells decreases proliferation, invasion, and tumorsphere formation. Therapeutic inhibition of Notch1 activity with gamma secretase inhibitors RO4929097 or DAPT in prostate cancer cells further results in decreased proliferative abilities. Loss of NOTCH1 and treatment of immunocompromised mice bearing prostate cancer xenografts with RO4929097 display significantly impaired tumor growth. Loss of NOTCH1 additionally decreased metastatic potential of prostate cancer cells in invasion assays in vitro as well as in vivo experiments. Moreover, treatment with gamma secretase inhibitors or NOTCH1 gene deletion synergized with antiandrogen therapies, enzalutamide or abiraterone, to decrease the growth of prostate cancer cells. Combination of gamma secretase inhibitors with abiraterone significantly inhibited cell migration and invasion, while combination with enzalutamide reversed enzalutamide-induced migration and invasion. These collective findings suggest loss of NOTCH1 delays growth of CRPC and inhibits metastasis, and inhibition of Notch1 activation in conjunction with second-generation antiandrogen therapies could delay growth and progression of prostate cancer.
Collapse
Affiliation(s)
- Meghan A Rice
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California
| | - Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California
| | - Ali Ghoochani
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California
| | - Austin Su
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, California.
| |
Collapse
|
30
|
Wei X, Zhang L, Zhou Z, Kwon OJ, Zhang Y, Nguyen H, Dumpit R, True L, Nelson P, Dong B, Xue W, Birchmeier W, Taketo MM, Xu F, Creighton CJ, Ittmann MM, Xin L. Spatially Restricted Stromal Wnt Signaling Restrains Prostate Epithelial Progenitor Growth through Direct and Indirect Mechanisms. Cell Stem Cell 2019; 24:753-768.e6. [PMID: 30982770 DOI: 10.1016/j.stem.2019.03.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/11/2018] [Accepted: 03/10/2019] [Indexed: 12/31/2022]
Abstract
Cell-autonomous Wnt signaling has well-characterized functions in controlling stem cell activity, including in the prostate. While niche cells secrete Wnt ligands, the effects of Wnt signaling in niche cells per se are less understood. Here, we show that stromal cells in the proximal prostatic duct near the urethra, a mouse prostate stem cell niche, not only produce multiple Wnt ligands but also exhibit strong Wnt/β-catenin activity. The non-canonical Wnt ligand Wnt5a, secreted by proximal stromal cells, directly inhibits proliefration of prostate epithelial stem or progenitor cells whereas stromal cell-autonomous canonical Wnt/β-catenin signaling indirectly suppresses prostate stem or progenitor activity via the transforming growth factor β (TGFβ) pathway. Collectively, these pathways restrain the proliferative potential of epithelial cells in the proximal prostatic ducts. Human prostate likewise exhibits spatially restricted distribution of stromal Wnt/β-catenin activity, suggesting a conserved mechanism for tissue patterning. Thus, this study shows how distinct stromal signaling mechanisms within the prostate cooperate to regulate tissue homeostasis.
Collapse
Affiliation(s)
- Xing Wei
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA; Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hoang Nguyen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center of Stem Cell and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruth Dumpit
- Human Biology Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence True
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Peter Nelson
- Human Biology Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Baijun Dong
- Department of Urology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xue
- Department of Urology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Walter Birchmeier
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Feng Xu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael M Ittmann
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX 77030, USA
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Urology, University of Washington, Seattle, WA 98109, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
31
|
Hao Y, Bjerke GA, Pietrzak K, Melhuish TA, Han Y, Turner SD, Frierson HF, Wotton D. TGFβ signaling limits lineage plasticity in prostate cancer. PLoS Genet 2018; 14:e1007409. [PMID: 29782499 PMCID: PMC5983872 DOI: 10.1371/journal.pgen.1007409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/01/2018] [Accepted: 05/10/2018] [Indexed: 01/08/2023] Open
Abstract
Although treatment options for localized prostate cancer (CaP) are initially effective, the five-year survival for metastatic CaP is below 30%. Mutation or deletion of the PTEN tumor suppressor is a frequent event in metastatic CaP, and inactivation of the transforming growth factor (TGF) ß signaling pathway is associated with more advanced disease. We previously demonstrated that mouse models of CaP based on inactivation of Pten and the TGFß type II receptor (Tgfbr2) rapidly become invasive and metastatic. Here we show that mouse prostate tumors lacking Pten and Tgfbr2 have higher expression of stem cell markers and genes indicative of basal epithelial cells, and that basal cell proliferation is increased compared to Pten mutants. To better model the primarily luminal phenotype of human CaP we mutated Pten and Tgfbr2 specifically in luminal cells, and found that these tumors also progress to invasive and metastatic cancer. Accompanying the transition to invasive cancer we observed de-differentiation of luminal tumor cells to an intermediate cell type with both basal and luminal markers, as well as differentiation to basal cells. Proliferation rates in these de-differentiated cells were lower than in either basal or luminal cells. However, de-differentiated cells account for the majority of cells in micro-metastases consistent with a preferential contribution to metastasis. We suggest that active TGFß signaling limits lineage plasticity in prostate luminal cells, and that de-differentiation of luminal tumor cells can drive progression to metastatic disease.
Collapse
Affiliation(s)
- Yi Hao
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
| | - Glen A. Bjerke
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
| | - Karolina Pietrzak
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
- Department of Cytobiochemistry, University of Lodz, Lodz, Poland
| | - Tiffany A. Melhuish
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
| | - Yu Han
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
| | - Stephen D. Turner
- Department of Public Health Sciences, University of Virginia, Charlottesville, United States of America
| | - Henry F. Frierson
- Department of Pathology, University of Virginia, Charlottesville, United States of America
| | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, United States of America
| |
Collapse
|
32
|
Dual tumor suppressing and promoting function of Notch1 signaling in human prostate cancer. Oncotarget 2018; 7:48011-48026. [PMID: 27384993 PMCID: PMC5216996 DOI: 10.18632/oncotarget.10333] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/12/2016] [Indexed: 12/22/2022] Open
Abstract
Adenocarcinomas of the prostate arise as multifocal heterogeneous lesions as the likely result of genetic and epigenetic alterations and deranged cell-cell communication. Notch signaling is an important form of intercellular communication with a role in growth/differentiation control and tumorigenesis. Contrasting reports exist in the literature on the role of this pathway in prostate cancer (PCa) development. We show here that i) compared to normal prostate tissue, Notch1 expression is significantly reduced in a substantial fraction of human PCas while it is unaffected or even increased in others; ii) acute Notch activation both inhibits and induces process networks associated with prostatic neoplasms; iii) down-modulation of Notch1 expression and activity in immortalized normal prostate epithelial cells increases their proliferation potential, while increased Notch1 activity in PCa cells suppresses growth and tumorigenicity through a Smad3-dependent mechanism involving p21WAF1/CIP1; iv) prostate cancer cells resistant to Notch growth inhibitory effects retain Notch1-induced upregulation of pro-oncogenic genes, like EPAS1 and CXCL6, also overexpressed in human PCas with high Notch1 levels. Taken together, these results reconcile conflicting data on the role of Notch1 in prostate cancer.
Collapse
|
33
|
Xiao L, Feng Q, Zhang Z, Wang F, Lydon JP, Ittmann MM, Xin L, Mitsiades N, He B. The essential role of GATA transcription factors in adult murine prostate. Oncotarget 2018; 7:47891-47903. [PMID: 27374105 PMCID: PMC5216986 DOI: 10.18632/oncotarget.10294] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/17/2016] [Indexed: 01/20/2023] Open
Abstract
GATA transcription factors are essential in mammalian cell lineage determination and have a critical role in cancer development. In cultured prostate cancer cells, GATA2 coordinates with androgen receptor (AR) to regulate gene transcription. In the murine prostate, among six GATA members, GATA2 and GATA3 are expressed. Immunofluorescence staining revealed that both GATA factors predominantly localize in the nuclei of luminal epithelial cells. The pioneer factor FoxA1 is exclusively detected in the luminal cells, whereas AR is detected in both luminal and basal cells. Using genetic engineering, we generated prostate-specific GATA2 and GATA3 knockout (KO) mice. Ablation of single GATA gene had marginal effect on prostate morphology and AR target gene expression, likely due to their genetic compensation. Double KO mice exhibited PIN III to IV lesions, but decreased prostate to body weight ratio, altered AR target gene expression, and expansion of p63-positive basal cells. However, deletion of GATA2 and GATA3 did not reduce the mRNA or protein levels of AR or FoxA1, indicating that GATA factors are not required for AR or FoxA1 expression in adult prostate. Surprisingly, GATA2 and GATA3 exhibit minimal expression in the ventral prostatic (VP) lobe. In contrast, FoxA1 and AR expression levels in VP are at least as high as those in anterior prostatic (AP) and dorsal-lateral prostatic (DLP) lobes. Together, our results indicate that GATA2 and GATA3 are essential for adult murine prostate function and in vivo AR signaling, and the lack of the GATA factor expression in the VP suggests a fundamental difference between VP and other prostatic lobes.
Collapse
Affiliation(s)
- Lijuan Xiao
- Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Qin Feng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Zheng Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Fen Wang
- The Center for Cancer and Stem Cell Biology, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey Veterans Affairs Medical Center, US Department of Veterans Affairs, Houston, TX, USA
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nicholas Mitsiades
- Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bin He
- Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
34
|
TGF-β Family Signaling in Ductal Differentiation and Branching Morphogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031997. [PMID: 28289061 DOI: 10.1101/cshperspect.a031997] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelial cells contribute to the development of various vital organs by generating tubular and/or glandular architectures. The fully developed forms of ductal organs depend on processes of branching morphogenesis, whereby frequency, total number, and complexity of the branching tissue define the final architecture in the organ. Some ductal tissues, like the mammary gland during pregnancy and lactation, disintegrate and regenerate through periodic cycles. Differentiation of branched epithelia is driven by antagonistic actions of parallel growth factor systems that mediate epithelial-mesenchymal communication. Transforming growth factor-β (TGF-β) family members and their extracellular antagonists are prominently involved in both normal and disease-associated (e.g., malignant or fibrotic) ductal tissue patterning. Here, we discuss collective knowledge that permeates the roles of TGF-β family members in the control of the ductal tissues in the vertebrate body.
Collapse
|
35
|
Montano M, Bushman W. Morphoregulatory pathways in prostate ductal development. Dev Dyn 2018; 246:89-99. [PMID: 27884054 DOI: 10.1002/dvdy.24478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/22/2023] Open
Abstract
The mouse prostate is a male sex-accessory gland comprised of a branched ductal network arranged into three separate bilateral lobes: the anterior, dorsolateral, and ventral lobes. Prostate ductal development is the primary morphogenetic event in prostate development and requires a complex regulation of spatiotemporal factors. This review provides an overview of prostate development and the major genetic regulators and signaling pathways involved. To identify new areas for further study, we briefly highlight the likely important, but relatively understudied, role of the extracellular matrix (ECM). Finally, we point out the potential importance of the ECM in influencing the behavior and prognosis of prostate cancer. Developmental Dynamics 246:89-99, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Monica Montano
- University of Wisconsin Madison, Department of Urology, Madison, Wisconsin.,University of Wisconsin Madison, Cellular and Molecular Pathology, Madison, Wisconsin.,University of Wisconsin Madison, Carbone Cancer Center, Clinical Sciences Center, Madison, Wisconsin
| | - Wade Bushman
- University of Wisconsin Madison, Department of Urology, Madison, Wisconsin.,University of Wisconsin Madison, Carbone Cancer Center, Clinical Sciences Center, Madison, Wisconsin
| |
Collapse
|
36
|
Histone 2B-GFP Label-Retaining Prostate Luminal Cells Possess Progenitor Cell Properties and Are Intrinsically Resistant to Castration. Stem Cell Reports 2017; 10:228-242. [PMID: 29276153 PMCID: PMC5768933 DOI: 10.1016/j.stemcr.2017.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/19/2022] Open
Abstract
The existence of slow-cycling luminal cells in the prostate has been suggested, but their identity and functional properties remain unknown. Using a bigenic mouse model to earmark, isolate, and characterize the quiescent stem-like cells, we identify a label-retaining cell (LRC) population in the luminal cell layer as luminal progenitors. Molecular and biological characterizations show that these luminal LRCs are significantly enriched in the mouse proximal prostate, exhibit relative dormancy, display bipotency in both in vitro and in vivo assays, and express a stem/progenitor gene signature with resemblance to aggressive prostate cancer. Importantly, these LRCs, compared with bulk luminal cells, maintain a lower level of androgen receptor (AR) expression and are less androgen dependent and also castration resistant in vivo. Finally, analysis of phenotypic markers reveals heterogeneity within the luminal progenitor cell pool. Our study establishes luminal LRCs as progenitors that may serve as a cellular origin for castration-resistant prostate cancer. A bigenic mouse model to study prostatic slow-cycling luminal epithelial cells Prostate label-retaining cells (LRCs) exhibit stem/progenitor cell activities Luminal LRCs are developmentally bipotent and display a progenitor gene signature Luminal LRCs resist castration and molecularly resemble aggressive prostate cancer
Collapse
|
37
|
Ito Y, Hoare M, Narita M. Spatial and Temporal Control of Senescence. Trends Cell Biol 2017; 27:820-832. [PMID: 28822679 DOI: 10.1016/j.tcb.2017.07.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 07/13/2017] [Accepted: 07/21/2017] [Indexed: 12/16/2022]
Abstract
Cellular senescence is an autonomous tumor suppressor mechanism leading to stable cell cycle arrest. Senescent cells are highly secretory, driving a range of different functions through the senescence-associated secretory phenotype (SASP). Recent findings have suggested that the composition of the SASP is dynamically and spatially regulated and that the changing composition of the SASP can determine the beneficial and detrimental aspects of the senescence program, tipping the balance to either an immunosuppressive/profibrotic environment or proinflammatory/fibrolytic state. Here, we discuss the current understanding of the temporal and spatial regulation of the SASP and the novel finding of NOTCH signaling as a regulator of SASP composition.
Collapse
Affiliation(s)
- Yoko Ito
- Cancer Research UK Cambridge Centre, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Matthew Hoare
- Cancer Research UK Cambridge Centre, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK; Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Masashi Narita
- Cancer Research UK Cambridge Centre, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK.
| |
Collapse
|
38
|
Ferrucci D, Biancardi MF, Nishan U, Rosa-Ribeiro R, Carvalho HF. Desquamation takes center stage at the origin of proliferative inflammatory atrophy, epithelial-mesenchymal transition, and stromal growth in benign prostate hyperplasia. Cell Biol Int 2017; 41:1265-1270. [PMID: 28877372 DOI: 10.1002/cbin.10867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/03/2017] [Indexed: 01/24/2023]
Abstract
In this commentary, we propose a relationship between desquamation, initially described as the collective detachment and deletion of epithelial cell in the prostate gland after castration, and proliferative inflammatory atrophy (PIA) and stromal growth in benign prostate hyperplasia (BPH). First, in response to diverse stimuli, including inflammatory mediators, epithelial cells desquamate and leave a large surface of the luminal side of the basement membrane (BM) exposed. Basal cells are activated into intermediate-type cells, which change morphology to cover and remodel the exposed BM (simple atrophy) to a new physiological demand (such as in the hypoandrogen environment, simulated by surgical and/or chemical castration) and/or to support re-epithelialization (under normal androgen levels). In the presence of inflammation (that might be the cause of desquamation), the intermediate-type cells proliferate and characterize PIA. Second, in other circumstances, desquamation is an early step of epithelial-to-mesenchymal transition (EMT), which contributes to stromal growth, as suggested by some experimental models of BPH. The proposed associations correlate unexplored cell behaviors and reveal the remarkable plasticity of the prostate epithelium that might be at the origin of prostate diseases.
Collapse
Affiliation(s)
- Danilo Ferrucci
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas SP, Brazil
| | - Manoel F Biancardi
- Department of Histology, Embryology, and Cell Biology, Institute of Biological Sciences, Federal University of Goiás, Goiania GO, Brazil
| | - Umar Nishan
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Rafaela Rosa-Ribeiro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas SP, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas SP, Brazil
| |
Collapse
|
39
|
Moad M, Hannezo E, Buczacki SJ, Wilson L, El-Sherif A, Sims D, Pickard R, Wright NA, Williamson SC, Turnbull DM, Taylor RW, Greaves L, Robson CN, Simons BD, Heer R. Multipotent Basal Stem Cells, Maintained in Localized Proximal Niches, Support Directed Long-Ranging Epithelial Flows in Human Prostates. Cell Rep 2017; 20:1609-1622. [PMID: 28813673 PMCID: PMC5565638 DOI: 10.1016/j.celrep.2017.07.061] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/24/2017] [Accepted: 07/21/2017] [Indexed: 12/15/2022] Open
Abstract
Sporadic mitochondrial DNA mutations serve as clonal marks providing access to the identity and lineage potential of stem cells within human tissues. By combining quantitative clonal mapping with 3D reconstruction of adult human prostates, we show that multipotent basal stem cells, confined to discrete niches in juxta-urethral ducts, generate bipotent basal progenitors in directed epithelial migration streams. Basal progenitors are then dispersed throughout the entire glandular network, dividing and differentiating to replenish the loss of apoptotic luminal cells. Rare lineage-restricted luminal stem cells, and their progeny, are confined to proximal ducts and provide only minor contribution to epithelial homeostasis. In situ cell capture from clonal maps identified delta homolog 1 (DLK1) enrichment of basal stem cells, which was validated in functional spheroid assays. This study establishes significant insights into niche organization and function of prostate stem and progenitor cells, with implications for disease.
Collapse
Affiliation(s)
- Mohammad Moad
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Edouard Hannezo
- Cavendish Laboratory, Department of Physics, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, UK; Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Simon J Buczacki
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Laura Wilson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Amira El-Sherif
- Department of Histopathology, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK; Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - David Sims
- Computational Genomics Analysis and Training (CGAT), MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Robert Pickard
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Nicholas A Wright
- Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Stuart C Williamson
- Clinical and Experimental Pharmacology Group, University of Manchester, Manchester M13 9PL, UK
| | - Doug M Turnbull
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Newcastle Centre for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Robert W Taylor
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Laura Greaves
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Newcastle Centre for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Craig N Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, UK; Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust/Medical Research Council Stem Cell Institute, Cambridge CB2 1QR, UK.
| | - Rakesh Heer
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK.
| |
Collapse
|
40
|
Frank SB, Berger PL, Ljungman M, Miranti CK. Human prostate luminal cell differentiation requires NOTCH3 induction by p38-MAPK and MYC. J Cell Sci 2017; 130:1952-1964. [PMID: 28446540 DOI: 10.1242/jcs.197152] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 04/17/2017] [Indexed: 12/14/2022] Open
Abstract
Many pathways dysregulated in prostate cancer are also involved in epithelial differentiation. To better understand prostate tumor initiation, we sought to investigate specific genes and mechanisms required for normal basal to luminal cell differentiation. Utilizing human prostate basal epithelial cells and an in vitro differentiation model, we tested the hypothesis that regulation of NOTCH3 by the p38 MAPK family (hereafter p38-MAPK), via MYC, is required for luminal differentiation. Inhibition (SB202190 and BIRB796) or knockdown of p38α (also known as MAPK14) and/or p38δ (also known as MAPK13) prevented proper differentiation. Additionally, treatment with a γ-secretase inhibitor (RO4929097) or knockdown of NOTCH1 and/or NOTCH3 greatly impaired differentiation and caused luminal cell death. Constitutive p38-MAPK activation through MKK6(CA) increased NOTCH3 (but not NOTCH1) mRNA and protein levels, which was diminished upon MYC inhibition (10058-F4 and JQ1) or knockdown. Furthermore, we validated two NOTCH3 enhancer elements through a combination of enhancer (e)RNA detection (BruUV-seq) and luciferase reporter assays. Finally, we found that the NOTCH3 mRNA half-life increased during differentiation or upon acute p38-MAPK activation. These results reveal a new connection between p38-MAPK, MYC and NOTCH signaling, demonstrate two mechanisms of NOTCH3 regulation and provide evidence for NOTCH3 involvement in prostate luminal cell differentiation.
Collapse
Affiliation(s)
- Sander B Frank
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA.,Genetics Program, Michigan State University, East Lansing, MI 48824, USA.,Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| | - Penny L Berger
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Mats Ljungman
- Translational Oncology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA .,Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| |
Collapse
|
41
|
Activation of Notch1 synergizes with multiple pathways in promoting castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2016; 113:E6457-E6466. [PMID: 27694579 DOI: 10.1073/pnas.1614529113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (CRPC) is the primary cause of prostate cancer-specific mortality. Defining new mechanisms that can predict recurrence and drive lethal CRPC is critical. Here, we demonstrate that localized high-risk prostate cancer and metastatic CRPC, but not benign prostate tissues or low/intermediate-risk prostate cancer, express high levels of nuclear Notch homolog 1, translocation-associated (Notch1) receptor intracellular domain. Chronic activation of Notch1 synergizes with multiple oncogenic pathways altered in early disease to promote the development of prostate adenocarcinoma. These tumors display features of epithelial-to-mesenchymal transition, a cellular state associated with increased tumor aggressiveness. Consistent with its activation in clinical CRPC, tumors driven by Notch1 intracellular domain in combination with multiple pathways altered in prostate cancer are metastatic and resistant to androgen deprivation. Our study provides functional evidence that the Notch1 signaling axis synergizes with alternative pathways in promoting metastatic CRPC and may represent a new therapeutic target for advanced prostate cancer.
Collapse
|
42
|
Wang L, Xie L, Tintani F, Xie H, Li C, Cui Z, Wan M, Zu X, Qi L, Cao X. Aberrant Transforming Growth Factor-β Activation Recruits Mesenchymal Stem Cells During Prostatic Hyperplasia. Stem Cells Transl Med 2016; 6:394-404. [PMID: 28191756 PMCID: PMC5442798 DOI: 10.5966/sctm.2015-0411] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/28/2016] [Indexed: 02/05/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is the overgrowth of prostate tissues with high prevalence in older men. BPH pathogenesis is not completely understood, but it is believed to be a result of de novo overgrowth of prostatic stroma. In this study, we show that aberrant activation of transforming growth factor‐β (TGF‐β) mobilizes mesenchymal/stromal stem cells (MSCs) in circulating blood, which are recruited for the prostatic stromal hyperplasia. Elevated levels of active TGF‐β were observed in both a phenylephrine‐induced prostatic hyperplasia mouse model and human BPH tissues. Nestin lineage tracing revealed that 39.6% ± 6.3% of fibroblasts and 73.3% ± 4.2% smooth muscle cells were derived from nestin+ cells in Nestin‐Cre, Rosa26‐YFPflox/+mice. Nestin+ MSCs were increased in the prostatic hyperplasia mice. Our parabiosis experiment demonstrate that nestin+ MSCs were mobilized and recruited to the prostatic stroma of wild‐type mice and gave rise to the fibroblasts. Moreover, injection of a TGF‐β neutralizing antibody (1D11) inhibits mobilization of MSCs, their recruitment to the prostatic stroma and hyperplasia. Importantly, knockout of TβRII in nestin+ cell lineage ameliorated stromal hyperplasia. Thus, elevated levels of TGF‐β‐induced mobilization and recruitment of MSCs to the reactive stroma resulting in overgrowth of prostate tissues in BPH and, thus, inhibition of TGF‐β activity could be a potential therapy for BPH. Stem Cells Translational Medicine2017;6:394–404
Collapse
Affiliation(s)
- Long Wang
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Liang Xie
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Francis Tintani
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hui Xie
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Changjun Li
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zhuang Cui
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mei Wan
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lin Qi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xu Cao
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Jagged1 upregulation in prostate epithelial cells promotes formation of reactive stroma in the Pten null mouse model for prostate cancer. Oncogene 2016; 36:618-627. [PMID: 27345403 PMCID: PMC5192002 DOI: 10.1038/onc.2016.232] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/15/2016] [Accepted: 05/30/2016] [Indexed: 12/13/2022]
Abstract
The role of Notch signaling in prostate cancer has not been defined definitively. Several large scale tissue microarray studies have revealed that the expression of some Notch signaling components including the Jagged1 ligand are upregulated in advanced human prostate cancer specimens. Jagged1 expressed by tumor cells may activate Notch signaling in both adjacent tumor cells and cells in tumor microenvironment. However, it remains undetermined whether increased Jagged1 expression reflects a cause for or a consequence of tumor progression in vivo. To address this question, we generated a novel R26-LSL-JAG1 mouse model that enables spatiotemporal Jagged1 expression. Prostate specific upregulation of Jagged1 neither interferes with prostate epithelial homeostasis nor significantly accelerates tumor initiation or progression in the prostate-specific Pten deletion mouse model for prostate cancer. However, Jagged1 upregulation results in increased inflammatory foci in tumors and incidence of intracystic adenocarcinoma. In addition, Jagged1 overexpression upregulates Tgfβ signaling in prostate stromal cells and promotes progression of a reactive stromal microenvironment in the Pten null prostate cancer model. Collectively, Jagged1 overexpression does not significantly accelerate prostate cancer initiation and progression in the context of loss-of-function of Pten, but alters tumor histopathology and microenvironment. Our study also highlights an understudied role of Notch signaling in regulating prostatic stromal homeostasis.
Collapse
|
44
|
Kwon OJ, Zhang L, Wang J, Su Q, Feng Q, Zhang XHF, Mani SA, Paulter R, Creighton CJ, Ittmann MM, Xin L. Notch promotes tumor metastasis in a prostate-specific Pten-null mouse model. J Clin Invest 2016; 126:2626-41. [PMID: 27294523 DOI: 10.1172/jci84637] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/22/2016] [Indexed: 12/28/2022] Open
Abstract
Although Notch signaling is deregulated in prostate cancer, the role of this pathway in disease development and progression is not fully understood. Here, we analyzed 2 human prostate cancer data sets and found that higher Notch signaling correlates with increased metastatic potential and worse disease survival rates. We used the Pten-null mouse prostate cancer model to investigate the function of Notch signaling in the initiation and progression of prostate cancer. Disruption of the transcription factor RBPJ in Pten-null mice revealed that endogenous canonical Notch signaling is not required for disease initiation and progression. However, augmentation of Notch activity in this model promoted both proliferation and apoptosis of prostate epithelial cells, which collectively reduced the primary tumor burden. The increase in cellular apoptosis was linked to DNA damage-induced p53 activation. Despite a reduced primary tumor burden, Notch activation in Pten-null mice promoted epithelial-mesenchymal transition and FOXC2-dependent tumor metastases but did not confer resistance to androgen deprivation. Notch activation also resulted in transformation of seminal vesicle epithelial cells in Pten-null mice. Our study highlights a multifaceted role for Notch signaling in distinct aspects of prostate cancer biology and supports Notch as a potential therapeutic target for metastatic prostate cancer.
Collapse
|
45
|
Welte T, Kim IS, Tian L, Gao X, Wang H, Li J, Holdman XB, Herschkowitz JI, Pond A, Xie G, Kurley S, Nguyen T, Liao L, Dobrolecki LE, Mo Q, Edwards DP, Huang S, Xin L, Xu J, Li Y, Lewis MT, Wang T, Westbrook TF, Rosen JM, Zhang XHF. Oncogenic mTOR signalling recruits myeloid-derived suppressor cells to promote tumour initiation. Nat Cell Biol 2016; 18:632-44. [PMID: 27183469 PMCID: PMC4884142 DOI: 10.1038/ncb3355] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) play critical roles in primary and metastatic cancer progression. MDSC regulation is widely variable even among patients harbouring the same type of malignancy, and the mechanisms governing such heterogeneity are largely unknown. Here, integrating human tumour genomics and syngeneic mammary tumour models, we demonstrate that mTOR signalling in cancer cells dictates a mammary tumour's ability to stimulate MDSC accumulation through regulating G-CSF. Inhibiting this pathway or its activators (for example, FGFR) impairs tumour progression, which is partially rescued by restoring MDSCs or G-CSF. Tumour-initiating cells (TICs) exhibit elevated G-CSF. MDSCs reciprocally increase TIC frequency through activating Notch in tumour cells, forming a feedforward loop. Analyses of primary breast cancers and patient-derived xenografts corroborate these mechanisms in patients. These findings establish a non-canonical oncogenic role of mTOR signalling in recruiting pro-tumorigenic MDSCs and show how defined cancer subsets may evolve to promote and depend on a distinct immune microenvironment.
Collapse
Affiliation(s)
- Thomas Welte
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Diana Helis Henry Medical Research Foundation, New Orlean, LA 70130, USA
| | - Ik Sun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lin Tian
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xia Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - June Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xue B. Holdman
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jason I. Herschkowitz
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Adam Pond
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | | | - Sarah Kurley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Tuan Nguyen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Qianxing Mo
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Dean P. Edwards
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Shixia Huang
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Li Xin
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jianming Xu
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Tian Wang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555
| | - Thomas F. Westbrook
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jeffrey M. Rosen
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| |
Collapse
|
46
|
Kwon OJ, Zhang B, Zhang L, Xin L. High fat diet promotes prostatic basal-to-luminal differentiation and accelerates initiation of prostate epithelial hyperplasia originated from basal cells. Stem Cell Res 2016; 16:682-91. [PMID: 27107344 DOI: 10.1016/j.scr.2016.04.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 01/03/2023] Open
Abstract
Recent lineage tracing studies showed that the prostate basal and luminal cells in adult mice are two independent lineages under the physiological condition, but basal cells are capable of generating luminal progenies during bacterial infection-induced prostatitis. Because acute bacterial infection in human prostate tissues is relatively rare, the disease relevance of the bacterial infection-induced basal-to-luminal differentiation is uncertain. Herein we employ a high fat diet-induced sterile prostate inflammation model to determine whether basal-to-luminal differentiation can be induced by inflammation irrespective of the underlying etiologies. A K14-CreER model and a fluorescent report line are utilized to specifically label basal cells with the green fluorescent protein. We show that high fat diet promotes immune cell infiltration into the prostate tissues and basal-to-luminal differentiation. Increased cell proliferation accompanies basal-to-luminal differentiation, suggesting a concurrent regulation of basal cell proliferation and differentiation. This study demonstrates that basal-to-luminal differentiation can be induced by different types of prostate inflammation evolved with distinct etiologies. Finally, high fat diet also accelerates initiation and progression of prostatic intraepithelial neoplasia that are originated from basal cells with loss-of-function of the tumor suppressor Pten. Because prostate cancer originated from basal cells tends to be invasive, our study also provides an alternative explanation for the association between obesity and aggressive prostate cancer.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States; Dan L. Duncan Cancer Center, Baylor College of Medicine, United States; Department of Pathology and Immunology.
| |
Collapse
|
47
|
Wei X, Orjalo AV, Xin L. CD133 does not enrich for the stem cell activity in vivo in adult mouse prostates. Stem Cell Res 2016; 16:597-606. [PMID: 27010655 DOI: 10.1016/j.scr.2016.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/12/2016] [Accepted: 03/10/2016] [Indexed: 11/30/2022] Open
Abstract
CD133 is widely used as a marker for stem/progenitor cells in many organ systems. Previous studies using in vitro stem cell assays have suggested that the CD133-expressing prostate basal cells may serve as the putative prostate stem cells. However, the precise localization of the CD133-expressing cells and their contributions to adult murine prostate homeostasis in vivo remain undetermined. We show that loss of function of CD133 does not impair murine prostate morphogenesis, homeostasis and regeneration, implying a dispensable role for CD133 in prostate stem cell function. Using a CD133-CreER(T2) model in conjunction with a fluorescent report line, we show that CD133 is not only expressed in a fraction of prostate basal cells, but also in some luminal cells and stromal cells. CD133(+) basal cells possess higher in vitro sphere-forming activities than CD133(-) basal cells. However, the in vivo lineage tracing study reveals that the two cell populations possess the same regenerative capacity and contribute equally to the maintenance of the basal cell lineage. Similarly, CD133(+) and CD133(-) luminal cells are functionally equivalent in maintaining the luminal cell lineage. Collectively, our study demonstrates that CD133 does not enrich for the stem cell activity in vivo in adult murine prostate. This study does not contradict previous reports showing CD133(+) cells as prostate stem cells in vitro. Instead, it highlights a substantial impact of biological contexts on cellular behaviors.
Collapse
Affiliation(s)
- Xing Wei
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, United States
| | - Arturo V Orjalo
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., United States
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States; Department of Pathology and Immunology, United States; Dan L. Duncan Cancer Center, Baylor College of Medicine, United States.
| |
Collapse
|
48
|
Stem cell and neurogenic gene-expression profiles link prostate basal cells to aggressive prostate cancer. Nat Commun 2016; 7:10798. [PMID: 26924072 PMCID: PMC4773505 DOI: 10.1038/ncomms10798] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/21/2016] [Indexed: 12/26/2022] Open
Abstract
The prostate gland mainly contains basal and luminal cells constructed as a pseudostratified epithelium. Annotation of prostate epithelial transcriptomes provides a foundation for discoveries that can impact disease understanding and treatment. Here we describe a genome-wide transcriptome analysis of human benign prostatic basal and luminal epithelial populations using deep RNA sequencing. Through molecular and biological characterizations, we show that the differential gene-expression profiles account for their distinct functional properties. Strikingly, basal cells preferentially express gene categories associated with stem cells, neurogenesis and ribosomal RNA (rRNA) biogenesis. Consistent with this profile, basal cells functionally exhibit intrinsic stem-like and neurogenic properties with enhanced rRNA transcription activity. Of clinical relevance, the basal cell gene-expression profile is enriched in advanced, anaplastic, castration-resistant and metastatic prostate cancers. Therefore, we link the cell-type-specific gene signatures to aggressive subtypes of prostate cancer and identify gene signatures associated with adverse clinical features. Gene-expression profiles can be used to predict the prognosis of cancer patients. Here, the authors describe gene expression profiles of human prostate epithelial lineages and show that basal cells have intrinsic stem and neurogenic properties, and molecularly resemble aggressive prostate cancer.
Collapse
|
49
|
Su Q, Xin L. Notch signaling in prostate cancer: refining a therapeutic opportunity. Histol Histopathol 2016; 31:149-57. [PMID: 26521657 PMCID: PMC4822406 DOI: 10.14670/hh-11-685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Notch is an evolutionarily conserved signaling pathway that plays a critical role in specifying cell fate and regulating tissue homeostasis and carcinogenesis. Studies using organ cultures and genetically engineered mouse models have demonstrated that Notch signaling regulates prostate development and homeostasis. However, the role of the Notch signaling pathway in prostate cancer remains inconclusive. Many published studies have documented consistent deregulation of major Notch signaling components in human prostate cancer cell lines, mouse models for prostate cancers, and human prostate cancer specimens at both the mRNA and the protein levels. However, functional studies in human cancer cells by modulation of Notch pathway elements suggest both tumor suppressive and oncogenic roles of Notch. These controversies may originate from our inadequate understanding of the regulation of Notch signaling under versatile genetic contexts, and reflect the multifaceted and pleiotropic roles of Notch in regulating different aspects of prostate cancer cell biology, such as proliferation, metastasis, and chemo-resistance. Future comprehensive studies using various mouse models for prostate cancer may help clarify the role of Notch signaling in prostate cancer and provide a solid basis for determining whether and how Notch should be employed as a therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Qingtai Su
- Department of Molecular and Cellular Biology, Baylor College of Medicine, and Graduate Program in Integrative Molecular and Biomedical Sciences, Houston, Texas, USA
| | - Li Xin
- Department of Molecular and Cellular Biology, Department of Pathology and Immunology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
50
|
Pedrosa AR, Graça JL, Carvalho S, Peleteiro MC, Duarte A, Trindade A. Notch signaling dynamics in the adult healthy prostate and in prostatic tumor development. Prostate 2016; 76:80-96. [PMID: 26419726 DOI: 10.1002/pros.23102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/16/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND The Notch signaling pathway has been implicated in prostate development, maintenance and tumorigenesis by its key role in cell-fate determination, differentiation and proliferation. Therefore, we proposed to analyze Notch family members transcription and expression, including ligands (Dll1, 3, 4 and Jagged1 and 2), receptors (Notch1-4) and effectors (Hes1, 2, 5 and Hey1, 2, L), in both normal and tumor bearing mouse prostates to better understand the dynamics of Notch signaling in prostate tumorigenesis. METHODS Wild type mice and transgenic adenocarcinoma of the mouse prostate model (TRAMP) mice were sacrificed at 18, 24 or 30 weeks of age and the prostates collected and processed for either whole prostate or prostate cell specific populations mRNA analysis and for protein expression analysis by immunohistochemistry and immunofluorescence. RESULTS We observed that Dll1 and Dll4 are expressed in the luminal compartment of the mouse healthy prostate, whereas Jagged2 expression is restricted to the basal and stromal compartment. Additionally, Notch2 and Notch4 are normally expressed in the prostate luminal compartment while Notch2 and Notch3 are also expressed in the stromal layer of the healthy prostate. As prostate tumor development takes place, there is up-regulation of Notch components. Particularly, the prostate tumor lesions have increased expression of Jagged1 and 2, of Notch3 and of Hey1. We have also detected the presence of activated Notch3 in prostatic tumors that co-express Jagged1 and ultimately the Hey1 effector. CONCLUSIONS Taken together our results point out the Notch axis Jagged1-2/Notch3/Hey1 to be important for prostate tumor development and worthy of additional functional studies and validation in human clinical disease.
Collapse
Affiliation(s)
- Ana-Rita Pedrosa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - José L Graça
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Sandra Carvalho
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Maria C Peleteiro
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|