1
|
Asemota S, Effah W, Young KL, Holt J, Cripe L, Ponnusamy S, Thiyagarajan T, Hwang DJ, He Y, Mcnamara K, Johnson D, Wang Y, Grimes B, Khosrosereshki Y, Hollingsworth TJ, Fleming MD, Pritchard FE, Hendrix A, Khan F, Fan M, Makowski L, Yin Z, Sasano H, Hayes DN, Pfeffer LM, Miller DD, Narayanan R. Identification of a targetable JAK-STAT enriched androgen receptor and androgen receptor splice variant positive triple-negative breast cancer subtype. Cell Rep 2023; 42:113461. [PMID: 37979170 PMCID: PMC10872270 DOI: 10.1016/j.celrep.2023.113461] [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: 06/29/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype with no targeted therapeutics. The luminal androgen receptor (LAR) subtype constitutes 15% of TNBC and is enriched for androgen receptor (AR) and AR target genes. Here, we show that a cohort of TNBC not only expresses AR at a much higher rate (∼80%) but also expresses AR splice variants (AR-SVs) (∼20%), further subclassifying LAR-TNBC. Higher AR and AR-SV expression and corresponding aggressive phenotypes are observed predominantly in specimens obtained from African American women. LAR TNBC specimens are enriched for interferon, Janus kinase (JAK)-signal activator and transducer (STAT), and androgen signaling pathways, which are exclusive to AR-expressing epithelial cancer cells. AR- and AR-SV-expressing TNBC cell proliferation and xenograft and patient-tumor explant growth are inhibited by AR N-terminal domain-binding selective AR degrader or by a JAK inhibitor. Biochemical analysis suggests that STAT1 is an AR coactivator. Collectively, our work identifies pharmacologically targetable TNBC subtypes and identifies growth-promoting interaction between AR and JAK-STAT signaling.
Collapse
Affiliation(s)
- Sarah Asemota
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Wendy Effah
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Kirsten L Young
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jeremiah Holt
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Linnea Cripe
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Suriyan Ponnusamy
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Yali He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Keely Mcnamara
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8577, Japan
| | - Daniel Johnson
- Molecular Bioinformatics Core, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Yinan Wang
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Brandy Grimes
- West Cancer Center and Research Institute, Memphis, TN 38138, USA
| | - Yekta Khosrosereshki
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - T J Hollingsworth
- Department of Ophthalmology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Martin D Fleming
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Frances E Pritchard
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Ashley Hendrix
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Farhan Khan
- Department of Pathology, Methodist Hospital, Memphis, TN 38104, USA
| | - Meiyun Fan
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Liza Makowski
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Zheng Yin
- Biomedical and Informatics Services Core, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8577, Japan
| | - D Neil Hayes
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Lawrence M Pfeffer
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Ramesh Narayanan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| |
Collapse
|
2
|
Connelly JA, Zhang X, Chen Y, Chao Y, Shi Y, Jacob TC, Wang QJ. Protein kinase D2 confers neuroprotection by promoting AKT and CREB activation in ischemic stroke. Neurobiol Dis 2023; 187:106305. [PMID: 37730136 PMCID: PMC10836334 DOI: 10.1016/j.nbd.2023.106305] [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: 06/18/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023] Open
Abstract
Ischemic stroke, constituting 80-90% of all strokes, is a leading cause of death and long-term disability in adults. There is an urgent need to discover new targets and therapies for this devastating condition. Protein kinase D (PKD), as a key target of diacylglycerol involved in ischemic responses, has not been well studied in ischemic stroke, particularly PKD2. In this study, we found that PKD2 expression and activity were significantly upregulated in the ipsilateral side of the brain after transient focal cerebral ischemia, which coincides with the upregulation of PKD2 in primary neurons in response to in vitro ischemia, implying a potential role of PKD2 in neuronal survival in ischemic stroke. Using kinase-dead PKD2 knock-in (PKD2-KI) mice, we examined whether loss of PKD2 activity affected stroke outcomes in mice subjected to 1 h of transient middle cerebral artery occlusion (tMCAO) and 24 h of reperfusion. Our data demonstrated that PKD2-KI mice exhibited larger infarction volumes and worsened neurological scores, indicative of increased brain injury, as compared to the wild-type (WT) mice, confirming a neuroprotective role of PKD2 in ischemia/reperfusion (I/R) injury. Mouse primary neurons obtained from PKD2-KI mice also exhibited increased cell death as compared to the WT neurons when subjected to in vitro ischemia. We have further identified AKT and CREB as two main signaling nodes through which PKD2 regulates neuronal survival during I/R injury. In summary, PKD2 confers neuroprotection in ischemic stroke by promoting AKT and CREB activation and targeted activation of PKD2 may benefit neuronal survival in ischemic stroke.
Collapse
Affiliation(s)
- Jaclyn A Connelly
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Xuejing Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yejie Shi
- Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
3
|
Shukla S, Riveros C, Al-Toubat M, Chardon-Robles J, Osumi T, Serrano S, Kase AM, Petit JL, Meurice N, Gleba J, Copland JA, Chauhan J, Fletcher S, Balaji KC. The Bivalent Bromodomain Inhibitor MT-1 Inhibits Prostate Cancer Growth. Cancers (Basel) 2023; 15:3851. [PMID: 37568667 PMCID: PMC10416835 DOI: 10.3390/cancers15153851] [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: 06/21/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Bromodomains (BD) are epigenetic readers of histone acetylation involved in chromatin remodeling and transcriptional regulation of several genes including protooncogene cellular myelocytomatosis (c-Myc). c-Myc is difficult to target directly by agents due to its disordered alpha helical protein structure and predominant nuclear localization. The epigenetic targeting of c-Myc by BD inhibitors is an attractive therapeutic strategy for prostate cancer (PC) associated with increased c-Myc upregulation with advancing disease. MT-1 is a bivalent BD inhibitor that is 100-fold more potent than the first-in-class BD inhibitor JQ1. MT-1 decreased cell viability and causes cell cycle arrest in G0/G1 phase in castration-sensitive and resistant PC cell lines in a dose-dependent fashion. The inhibition of c-Myc function by MT-1 was molecularly corroborated by the de-repression of Protein Kinase D1 (PrKD) and increased phosphorylation of PrKD substrate proteins: threonine 120, serine 11, and serine 216 amino acid residues in β-Catenin, snail, and cell division cycle 25c (CDC25c) proteins, respectively. The treatment of 3D cell cultures derived from three unique clinically annotated heavily pretreated patient-derived PC xenografts (PDX) mice models with increasing doses of MT-1 demonstrated the lowest IC50 in tumors with c-Myc amplification and clinically resistant to Docetaxel, Cabazitaxel, Abiraterone, and Enzalutamide. An intraperitoneal injection of either MT-1 or in combination with 3jc48-3, an inhibitor of obligate heterodimerization with MYC-associated protein X (MAX), in mice implanted with orthotopic PC PDX, decreased tumor growth. This is the first pre-clinical study demonstrating potential utility of MT-1 in the treatment of PC with c-Myc dysregulation.
Collapse
Affiliation(s)
- Sanjeev Shukla
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| | - Carlos Riveros
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| | - Mohammed Al-Toubat
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| | - Jonathan Chardon-Robles
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
- Department of Neurological Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Teruko Osumi
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| | - Samuel Serrano
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| | - Adam M. Kase
- Department of Hematology Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Joachim L. Petit
- Department of Hematology Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nathalie Meurice
- Department of Hematology Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Justyna Gleba
- Cancer Biology Department, Mayo Clinic, Jacksonville, FL 32224, USA
| | - John A. Copland
- Cancer Biology Department, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jay Chauhan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - K. C. Balaji
- Department of Urology, University of Florida Health, Jacksonville, FL 32209, USA
| |
Collapse
|
4
|
Roy A, Prasad S, Chen Y, Chao Y, Liu Y, Zhao J, Wang QJ. Protein Kinase D2 and D3 Promote Prostate Cancer Cell Bone Metastasis by Positively Regulating Runx2 in a MEK/ERK1/2-Dependent Manner. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:624-637. [PMID: 36740185 PMCID: PMC10155267 DOI: 10.1016/j.ajpath.2023.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Advanced-stage prostate tumors metastasize to the bone, often causing death. The protein kinase D (PKD) family has been implicated in prostate cancer development; however, its role in prostate cancer metastasis remains elusive. This study examined the contribution of PKD, particularly PKD2 and PKD3 (PKD2/3), to the metastatic potential of prostate cancer cells and the effect of PKD inhibition on prostate cancer bone metastasis in vivo. Depletion of PKD2/3 by siRNAs or inhibition by the PKD inhibitor CRT0066101 in AR-positive and AR-negative castration-resistant prostate cancer cells potently inhibited colony formation and cell migration. Depletion or inhibition of PKD2/3 significantly blocked tumor cell invasion and suppressed the expression of genes related to bone metastasis in the highly invasive PC3-ML cells. The reduced invasive activity resulting from PKD2/3 depletion was in part mediated by the transcription factor Runx2, as its silencing decreased PKD2/3-mediated metastatic gene expression through the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 signaling axis. Furthermore, inhibition of PKD by CRT0066101 potently decreased the frequency of bone micrometastases in a mouse model of bone metastasis based on intracardiac injection of PC3-ML cells. These results indicate that PKD2/3 plays an important role in the bone metastasis of prostate cancer cells, and its inhibition may be beneficial for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Sahdeo Prasad
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yu Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jinjun Zhao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
5
|
Potential role for protein kinase D inhibitors in prostate cancer. J Mol Med (Berl) 2023; 101:341-349. [PMID: 36843036 DOI: 10.1007/s00109-023-02298-4] [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: 06/23/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 02/28/2023]
Abstract
Protein kinase D (PrKD), a novel serine-threonine kinase, belongs to a family of calcium calmodulin kinases that consists of three isoforms: PrKD1, PrKD2, and PrKD3. The PrKD isoforms play a major role in pathologic processes such as cardiac hypertrophy and cancer progression. The charter member of the family, PrKD1, is the most extensively studied isoform. PrKD play a dual role as both a proto-oncogene and a tumor suppressor depending on the cellular context. The duplicity of PrKD can be highlighted in advanced prostate cancer (PCa) where expression of PrKD1 is suppressed whereas the expressions of PrKD2 and PrKD3 are upregulated to aid in cancer progression. As understanding of the PrKD signaling pathways has been better elucidated, interest has been garnered in the development of PrKD inhibitors. The broad-spectrum kinase inhibitor staurosporine acts as a potent PrKD inhibitor and is the most well-known; however, several other novel and more specific PrKD inhibitors have been developed over the last two decades. While there is tremendous potential for PrKD inhibitors to be used in a clinical setting, none has progressed beyond preclinical trials due to a variety of challenges. In this review, we focus on PrKD signaling in PCa and the potential role of PrKD inhibitors therein, and explore the possible clinical outcomes based on known function and expression of PrKD isoforms at different stages of PCa.
Collapse
|
6
|
Yao J, Jiang Y, Geng S, Sun L. Protein Kinase D1 Is Increased in Tumor Tissue, Correlates With Advanced Tumor Features and Worse Prognosis of Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2020; 19:1533033820934129. [PMID: 32985371 PMCID: PMC7534063 DOI: 10.1177/1533033820934129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE This study aimed to assess protein kinase D1 expression and its association with tumor characteristics as well as prognosis in patients with non-small cell lung cancer. METHODS Protein kinase D1 expression in tumor tissues and adjacent tissues from 172 patients with non-small cell lung cancer who underwent surgical resection were analyzed by immunohistochemical staining. Based on the total immunohistochemical score, protein kinase D1 expression was classified as protein kinase D1 high expression (further divided into protein kinase D1 high+++, protein kinase D1 high++, and protein kinase D1 high+ expressions) and protein kinase D1 low expression. Clinical characteristics of patients with non-small cell lung cancer were acquired from the database. Accumulating disease-free survival and overall survival were calculated based on patients' relapse/survival status. RESULTS Protein kinase D1 expression was increased in tumor tissues compared to adjacent tissues (P < .001). Tumor protein kinase D1 high expression correlated with poorer pathological differentiation (P = .041), increased tumor size (P = .003), the presence of lymph node metastasis (P = .001), and elevated tumor, nodes and metastases stage (P < .001). Besides, both accumulating disease-free survival and overall survival were decreased in patients with tumor protein kinase D1 high expression compared to patients with tumor protein kinase D1 low expression (P = .010 for disease-free survival and P = 0.005 for overall survival). Moreover, they were lowest in patients with tumor protein kinase D1 high+++ expression, followed by patients with tumor protein kinase D1 high++ expression, then patients with tumor protein kinase D1 high+ expression, and highest in patients with tumor protein kinase D1 low expression (P < .001 for disease-free survival and P = .001 for overall survival). Notably, higher tumor protein kinase D1 expression was an independent predictive factor for decreased disease-free survival (P = .001) and overall survival (P = .004). CONCLUSIONS Protein kinase D1 might be a potential marker to identify patients with non-small cell lung cancer with worse tumor features and prognosis.
Collapse
Affiliation(s)
- Jing Yao
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Jiang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Geng
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Sun
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Nyquist MD, Corella A, Mohamad O, Coleman I, Kaipainen A, Kuppers DA, Lucas JM, Paddison PJ, Plymate SR, Nelson PS, Mostaghel EA. Molecular determinants of response to high-dose androgen therapy in prostate cancer. JCI Insight 2019; 4:129715. [PMID: 31503550 DOI: 10.1172/jci.insight.129715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/04/2019] [Indexed: 12/16/2022] Open
Abstract
Clinical trials of high-dose androgen (HDA) therapy for prostate cancer (PC) have shown promising efficacy but are limited by lack of criteria to identify likely responders. To elucidate factors that govern the growth-repressive effects of HDAs, we applied an unbiased integrative approach using genetic screens and transcriptional profiling of PC cells with or without demonstrated phenotypic sensitivity to androgen-mediated growth repression. Through this comprehensive analysis, we identified genetic events and related signaling networks that determine the response to both HDA and androgen withdrawal. We applied these findings to develop a gene signature that may serve as an early indicator of treatment response and identify men with tumors that are amenable to HDA therapy.
Collapse
Affiliation(s)
| | | | | | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Arja Kaipainen
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Daniel A Kuppers
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jared M Lucas
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Stephen R Plymate
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| |
Collapse
|
8
|
Youssef I, Ricort JM. Deciphering the Role of Protein Kinase D1 (PKD1) in Cellular Proliferation. Mol Cancer Res 2019; 17:1961-1974. [PMID: 31311827 DOI: 10.1158/1541-7786.mcr-19-0125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/05/2019] [Accepted: 07/11/2019] [Indexed: 11/16/2022]
Abstract
Protein kinase D1 (PKD1) is a serine/threonine kinase that belongs to the calcium/calmodulin-dependent kinase family, and is involved in multiple mechanisms implicated in tumor progression such as cell motility, invasion, proliferation, protein transport, and apoptosis. While it is expressed in most tissues in the normal state, PKD1 expression may increase or decrease during tumorigenesis, and its role in proliferation is context-dependent and poorly understood. In this review, we present and discuss the current landscape of studies investigating the role of PKD1 in the proliferation of both cancerous and normal cells. Indeed, as a potential therapeutic target, deciphering whether PKD1 exerts a pro- or antiproliferative effect, and under what conditions, is of paramount importance.
Collapse
Affiliation(s)
- Ilige Youssef
- Centre National de la Recherche Scientifique, CNRS UMR_8113, Laboratoire de Biologie et Pharmacologie Appliquée, Cachan, France.,École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Cachan, France
| | - Jean-Marc Ricort
- Centre National de la Recherche Scientifique, CNRS UMR_8113, Laboratoire de Biologie et Pharmacologie Appliquée, Cachan, France. .,École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Cachan, France.,Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| |
Collapse
|
9
|
Antognelli C, Ferri I, Bellezza G, Siccu P, Love HD, Talesa VN, Sidoni A. Glyoxalase 2 drives tumorigenesis in human prostate cells in a mechanism involving androgen receptor and p53-p21 axis. Mol Carcinog 2017; 56:2112-2126. [PMID: 28470764 DOI: 10.1002/mc.22668] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/26/2017] [Accepted: 05/01/2017] [Indexed: 12/20/2022]
Abstract
Glyoxalase 2 (Glo2), a metabolic enzyme, is overexpressed in some human cancers which suggests this enzyme may play a role in human tumorigenesis. In prostate cancer (PCa), the role of Glo2 has been scarcely investigated and there are no studies addressing a causative involvement of this protein in this neoplasia. Here, we examined the immunohistochemical profile of Glo2 in human PCa and benign adjacent tissues and investigated Glo2 involvement in PCa development in human prostate cell lines. PCa and matched adjacent normal tissues were obtained from paraffin sections of primary PCa from 20 patients who had undergone radical prostatectomy. Histopathological diagnosis was confirmed for each sample. Glo2 expression analysis was performed by immunohistochemistry in prostate tissues, and by qRT-PCR and immunoblotting in prostate cell lines. The causative and mechanistic role of Glo2 in prostate tumorigenesis was demonstrated by Glo2 ectopic expression/silencing and employing specific activators/inhibitors. Our results showed that Glo2 was selectively expressed in PCa but not in the luminal compartment of the adjacent benign epithelium consistently in all the examined 20 cases. Glo2 expression in PCa was dependent on androgen receptor (AR) and was aimed at stimulating cell proliferation and eluding apoptosis through a mechanism involving the p53-p21 axis. Glo2 was intensely expressed in the basal cells of benign glands but was not involved in PCa genesis. Our results demonstrate for the first time that Glo2 drives prostate tumorigenesis and suggest that it may represent a novel adjuvant marker in the pathological diagnosis of early PCa.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paola Siccu
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Harold D Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vincenzo N Talesa
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Roy A, Ye J, Deng F, Wang QJ. Protein kinase D signaling in cancer: A friend or foe? Biochim Biophys Acta Rev Cancer 2017; 1868:283-294. [PMID: 28577984 DOI: 10.1016/j.bbcan.2017.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/18/2022]
Abstract
Protein kinase D is a family of evolutionarily conserved serine/threonine kinases that belongs to the Ca++/Calmodulin-dependent kinase superfamily. Signal transduction pathways mediated by PKD can be triggered by a variety of stimuli including G protein-coupled receptor agonists, growth factors, hormones, and cellular stresses. The regulatory mechanisms and physiological roles of PKD have been well documented including cell proliferation, survival, migration, angiogenesis, regulation of gene expression, and protein/membrane trafficking. However, its precise roles in disease progression, especially in cancer, remain elusive. A plethora of studies documented the cell- and tissue-specific expressions and functions of PKD in various cancer-associated biological processes, while the causes of the differential effects of PKD have not been thoroughly investigated. In this review, we have discussed the structural-functional properties, activation mechanisms, signaling pathways and physiological functions of PKD in the context of human cancer. Additionally, we have provided a comprehensive review of the reported tumor promoting or tumor suppressive functions of PKD in several major cancer types and discussed the discrepancies that have been raised on PKD as a major regulator of malignant transformation.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Jing Ye
- Department of Anesthesiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
11
|
Roy A, Wang QJ. Protein Kinase D: A Potential Therapeutic Target in Prostate Cancer. MOLECULAR AND CELLULAR PHARMACOLOGY 2017; 9:1-4. [PMID: 34765081 PMCID: PMC8580385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Protein kinase D (PKD) belongs to a family of serine/threonine kinases in the calcium/calmodulin-dependent kinase superfamily. It modulates a number of signal transduction pathways involved in regulation of cell proliferation, survival, migration, angiogenesis, regulation of gene expression, and protein/membrane trafficking, mediated by variety of stimuli such as growth factors, hormones, and cellular stresses. Although its role in cancer progression remains elusive, current literature supports a potential tumor promoting function of the selective PKD isoforms in prostate cancer, making them promising therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|