1
|
Han H, Liu H, Steiner R, Zhao Z, Jin ZG. Inhibition of protein kinase D and its substrate phosphatidylinositol-4 kinase III beta blocks common human coronavirus replication. Microbiol Spectr 2024; 12:e0150124. [PMID: 39540754 PMCID: PMC11619529 DOI: 10.1128/spectrum.01501-24] [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: 06/20/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the infection of a coronavirus, named as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronaviruses can be replicated in the infected host cells. Coronavirus replication involves various steps, including membrane fusion, peri-nuclear particle formation, and matrix vesicle transport to the cell membrane via the endoplasmic reticulum-Golgi-lysosome route. Recent studies have suggested that protein kinase D (PKD) plays a crucial role in regulation of vesicle formation and trafficking in the trans-Golgi network (TGN). Thus, we hypothesize that inhibiting PKD and its associated pathway could be an effective strategy to limit viral replication. Here, we report that molecular and pharmacological inhibition of PKD and its substrate phosphatidylinositol-4 kinase III beta (PI4KIIIβ) significantly diminishes the replication of common human coronaviruses. Specifically, we found that the PKD-silencing siRNA and the PKD inhibitor CRT0066101 have broad-spectrum antiviral activity against HCoV-OC43, HCoV-NL63, and HCoV-229E in cultured cells. Mechanistic studies revealed that the deactivation of PKD reduced the activation of PI4KIIIβ, thereby blocking the transport of viral particles in the host cells. Furthermore, the PI4KIIIβ inhibitor, BQR695, also exhibited antiviral activity against those coronaviruses. In conclusion, PKD and its substrate, PI4KIIIβ, may serve as novel antiviral targets for human coronaviruses and warrant further investigation. IMPORTANCE Human coronaviruses can lead to a range of clinical symptoms, from asymptomatic infection to severe illness and death, with a limited array of antiviral drugs available. Protein kinase D (PKD) is involved in various cellular processes, such as cell proliferation, apoptosis, and membrane fission of the Golgi apparatus. However, the specific role of PKD in the human coronavirus life cycle remains unclear. In this study, we found that PKD inhibitors effectively attenuated human coronavirus replication at the trans-Golgi network (TGN) stage in the viral life cycle. Furthermore, inhibiting PKD reduced PI4KIIIβ activation, thereby blocking viral replication in the host cells. Importantly, PI4KIIIβ inhibitors also blocked human coronavirus replication. Thus, PKD may represent a promising therapeutic target against both current circulating and future emerging coronaviruses.
Collapse
Affiliation(s)
- Huijuan Han
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zhijun Zhao
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zheng-Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
2
|
Retracted: PKC-delta and PKD activate MAPK signal pathway in mechano-transcription of colonic smooth muscle cells. Neurogastroenterol Motil 2024; 36:e14623. [PMID: 37278189 DOI: 10.1111/nmo.14623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 06/07/2023]
Abstract
Retraction: [PKC-delta and PKD activate MAPK signal pathway in mechano-transcription of colonic smooth muscle cells, Z. Yang, K. He, T. Wang, et al. Neurogastroenterology & Motility 2023; e14623 (https://onlinelibrary.wiley.com/doi/full/10.1111/nmo.14623)]. The above article, published online on June 6, 2023 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the authors, the Journal Editor in Chief, Maura Corsetti, and John Wiley & Sons Ltd. The retraction has been agreed due to unat[1]tributed overlap between this article and the abstract published in Gastroenterology: Li F, Sarna SK and Shi XP. Roles of PKCs and PKD in Mechanotranscription in Colonic Smooth Muscle Cells: Inhibition of Mechanotranscription as a Potential Treatment for Motility Dysfunction in Obstructive Disorders. In: 2012 Digestive Disease Week Abstract Supplement; May 19-22, San Diego, CA. Abstract 120 (https://www.gastrojournal.org/article/S0016-5085(12)60115-2/pdf).
Collapse
|
3
|
Lv C, Zhou L, Meng Y, Yuan H, Geng J. PKD knockdown mitigates Ang II-induced cardiac hypertrophy and ferroptosis via the JNK/P53 signaling pathway. Cell Signal 2024; 113:110974. [PMID: 37972803 DOI: 10.1016/j.cellsig.2023.110974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/18/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Cardiac hypertrophy is studied in relation to energy metabolism, autophagy, and ferroptosis, which are associated with cardiovascular adverse events and chronic heart failure. Protein kinase D (PKD) has been shown to play a degenerative role in cardiac hypertrophy. However, the role of ferroptosis in PKD-involved cardiac hypertrophy remains unclear. METHODS A cardiac hypertrophy model was induced by a subcutaneous injection of angiotensin II (Ang II) for 4 weeks. Adeno-associated virus serotype 9 (AAV9)-PKD or AAV9-Negative control were injected through the caudal vein 2 weeks prior to the injection of Ang II. The degree of cardiac hypertrophy was assessed using echocardiography and by observing cardiomyocyte morphology. Levels of ferroptosis and protein expression in the Jun N-terminal kinase (JNK)/P53 signaling pathway were measured both in vivo and in vitro. RESULTS The results indicated that PKD knockdown reduces Ang II-induced cardiac hypertrophy, enhances cardiac function and inhibits ferroptosis. The involvement of the JNK/P53 pathway in this process was further confirmed by in vivo and in vitro experiments. CONCLUSION In conclusion, our findings suggest that PKD knockdown mitigates Ang II-induced cardiac hypertrophy and ferroptosis via the JNK/P53 signaling pathway.
Collapse
Affiliation(s)
- Chanyuan Lv
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Shandong 250021, China.
| | - Liuyi Zhou
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Shandong 250021, China
| | - Yongkang Meng
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Shandong 250021, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Shandong 250021, China.
| | - Jing Geng
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Shandong 250021, China.
| |
Collapse
|
4
|
Obata Y, Kurokawa K, Tojima T, Natsume M, Shiina I, Takahashi T, Abe R, Nakano A, Nishida T. Golgi retention and oncogenic KIT signaling via PLCγ2-PKD2-PI4KIIIβ activation in gastrointestinal stromal tumor cells. Cell Rep 2023; 42:113035. [PMID: 37616163 DOI: 10.1016/j.celrep.2023.113035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/19/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023] Open
Abstract
Most gastrointestinal stromal tumors (GISTs) develop due to gain-of-function mutations in the tyrosine kinase gene, KIT. We recently showed that mutant KIT mislocalizes to the Golgi area and initiates uncontrolled signaling. However, the molecular mechanisms underlying its Golgi retention remain unknown. Here, we show that protein kinase D2 (PKD2) is activated by the mutant, which causes Golgi retention of KIT. In PKD2-inhibited cells, KIT migrates from the Golgi region to lysosomes and subsequently undergoes degradation. Importantly, delocalized KIT cannot trigger downstream activation. In the Golgi/trans-Golgi network (TGN), KIT activates the PKD2-phosphatidylinositol 4-kinase IIIβ (PKD2-PI4KIIIβ) pathway through phospholipase Cγ2 (PLCγ2) to generate a PI4P-rich membrane domain, where the AP1-GGA1 complex is aberrantly recruited. Disruption of any factors in this cascade results in the release of KIT from the Golgi/TGN. Our findings show the molecular mechanisms underlying KIT mislocalization and provide evidence for a strategy for inhibition of oncogenic signaling.
Collapse
Affiliation(s)
- Yuuki Obata
- Laboratory of Intracellular Traffic & Oncology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Takuro Tojima
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Miyuki Natsume
- Laboratory of Intracellular Traffic & Oncology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Applied Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Isamu Shiina
- Department of Applied Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryo Abe
- Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Toshirou Nishida
- National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
5
|
Venkat R, Verma E, Daimary UD, Kumar A, Girisa S, Dutta U, Ahn KS, Kunnumakkara AB. The Journey of Resveratrol from Vineyards to Clinics. Cancer Invest 2023; 41:183-220. [PMID: 35993769 DOI: 10.1080/07357907.2022.2115057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
With rising technological advancements, several factors influence the lifestyle of people and stimulate chronic inflammation that severely affects the human body. Chronic inflammation leads to a broad range of physical and pathophysiological distress. For many years, non-steroidal drugs and corticosteroids were most frequently used in treating inflammation and related ailments. However, long-term usage of these drugs aggravates the conditions of chronic diseases and is presented with morbid side effects, especially in old age. Hence, the quest for safe and less toxic anti-inflammatory compounds of high therapeutic potential with least adverse side effects has shifted researchers' attention to ancient medicinal system. Resveratrol (RSV) - 3,4,5' trihydroxystilbene is one such naturally available polyphenolic stilbene derivative obtained from various plant sources. For over 2000 years, these plants have been used in Asian medicinal system for curing inflammation-associated disorders. There is a wealth of in vitro, in vivo and clinical evidence that shows RSV could induce anti-aging health benefits including, anti-cancer, anti-inflammatory, anti-oxidant, phytoesterogenic, and cardio protective properties. However, the issue of rapid elimination of RSV through the metabolic system and its low bio-availability is of paramount importance which is being studied extensively. Therefore, in this article, we scientifically reviewed the molecular targets, biological activities, beneficial and contradicting effects of RSV as evinced by clinical studies for the prevention and treatment of inflammation-mediated chronic disorders.
Collapse
Affiliation(s)
- Ramya Venkat
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| | - Elika Verma
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| | - Uzini Devi Daimary
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| | - Aviral Kumar
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| | - Sosmitha Girisa
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| | - Uma Dutta
- Department of Zoology, Cell and Molecular Biology Laboratory, Cotton University, Guwahati, India
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ajaikumar B Kunnumakkara
- Department of Biosciences and Bioengineering, Cancer Biology Laboratory, Indian Institute of Technology (IIT) Guwahati, Guwahati, India
| |
Collapse
|
6
|
Lee CS, Zhai Y, Shang R, Wong T, Mattison AJ, Cen HH, Johnson JD, Vlodavsky I, Hussein B, Rodrigues B. Flow-Induced Secretion of Endothelial Heparanase Regulates Cardiac Lipoprotein Lipase and Changes Following Diabetes. J Am Heart Assoc 2022; 11:e027958. [PMID: 36416172 PMCID: PMC9851453 DOI: 10.1161/jaha.122.027958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Lipoprotein lipase (LPL)-derived fatty acid is a major source of energy for cardiac contraction. Synthesized in cardiomyocytes, LPL requires translocation to the vascular lumen for hydrolysis of lipoprotein triglyceride, an action mediated by endothelial cell (EC) release of heparanase. We determined whether flow-mediated biophysical forces can cause ECs to secrete heparanase and thus regulate cardiac metabolism. Methods and Results Isolated hearts were retrogradely perfused. Confluent rat aortic ECs were exposed to laminar flow using an orbital shaker. Cathepsin L activity was determined using gelatin-zymography. Diabetes was induced in rats with streptozotocin. Despite the abundance of enzymatically active heparanase in the heart, it was the enzymatically inactive, latent heparanase that was exceptionally responsive to flow-induced release. EC exposed to orbital rotation exhibited a similar pattern of heparanase secretion, an effect that was reproduced by activation of the mechanosensor, Piezo1. The laminar flow-mediated release of heparanase from EC required activation of both the purinergic receptor and protein kinase D, a kinase that assists in vesicular transport of proteins. Heparanase influenced cardiac metabolism by increasing cardiomyocyte LPL displacement along with subsequent replenishment. The flow-induced heparanase secretion was augmented following diabetes and could explain the increased heparin-releasable pool of LPL at the coronary lumen in these diabetic hearts. Conclusions ECs sense fluid shear-stress and communicate this information to subjacent cardiomyocytes with the help of heparanase. This flow-induced mechanosensing and its dynamic control of cardiac metabolism to generate ATP, using LPL-derived fatty acid, is exquisitely adapted to respond to disease conditions, like diabetes.
Collapse
Affiliation(s)
- Chae Syng Lee
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Yajie Zhai
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Rui Shang
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Trevor Wong
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Aurora J. Mattison
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - Haoning Howard Cen
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research CenterRappaport Faculty of Medicine, TechnionHaifaIsrael
| | - Bahira Hussein
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Brian Rodrigues
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| |
Collapse
|
7
|
Janardhanan P, Somasundaran AK, Balakrishnan AJ, Pilankatta R. Sensitization of cancer cells towards Cisplatin and Carboplatin by protein kinase D inhibitors through modulation of ATP7A/B (copper transport ATPases). Cancer Treat Res Commun 2022; 32:100613. [PMID: 35908410 DOI: 10.1016/j.ctarc.2022.100613] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Drug resistance of cancer cells is a significant impediment to effective chemotherapy. One primary reason for this is copper exporters - ATPase copper transporting alpha (ATP7A) and ATPase copper transporting beta (ATP7B). These molecular pumps belong to P-type ATPases and dispose off the Platinum (Pt) based anticancer drugs from cancer cells, causing resistance in them. For the disposal of Pt-drugs, copper exporters require phosphorylation mediated by protein kinase D (PKD) for their activation and trafficking. Even though various research works are underway to overcome resistance to anticancer drugs, the role of PKD is mainly ignored. In this study, we have found a significant upregulation of ATP7A and ATP7B in cervical cancer cells (HeLa) and Liver Hepatocellular Carcinoma cells (HepG2) in the presence of Cisplatin or Carboplatin; both at transcriptional as well as translational levels. Interestingly, the expression of ATP7A and ATP7B were significantly downregulated in the presence of a PKD inhibitor (CID2011756), resulting in the reduction of PKD mediated phosphorylation of ATP7A/7B. This causes enhancement of proteasome-mediated degradation of ATP7A/7B and thereby sensitizes the cells towards Cisplatin and Carboplatin. Similarly, the treatment of Cisplatin resistant HepG2 cells with PKD inhibitor causes enhanced sensitivity towards Cisplatin drug. However, the presence of proteasome inhibitor (MG132) reversed the effect of the PKD inhibitor on the expression level of ATP7A/7B, indicating the necessity of phosphorylation for its stability. Hence, we conclude that the combinatorial usage of Cisplatin with drugs targeting PKD can be developed as an effective chemotherapeutic approach to overcome drug resistance.
Collapse
Affiliation(s)
- Prajit Janardhanan
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Periye, Kasaragod, Kerala 671316, India
| | | | - Anjali Jayasree Balakrishnan
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Periye, Kasaragod, Kerala 671316, India
| | - Rajendra Pilankatta
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Periye, Kasaragod, Kerala 671316, India.
| |
Collapse
|
8
|
Spano D, Colanzi A. Golgi Complex: A Signaling Hub in Cancer. Cells 2022; 11:1990. [PMID: 35805075 PMCID: PMC9265605 DOI: 10.3390/cells11131990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 02/01/2023] Open
Abstract
The Golgi Complex is the central hub in the endomembrane system and serves not only as a biosynthetic and processing center but also as a trafficking and sorting station for glycoproteins and lipids. In addition, it is an active signaling hub involved in the regulation of multiple cellular processes, including cell polarity, motility, growth, autophagy, apoptosis, inflammation, DNA repair and stress responses. As such, the dysregulation of the Golgi Complex-centered signaling cascades contributes to the onset of several pathological conditions, including cancer. This review summarizes the current knowledge on the signaling pathways regulated by the Golgi Complex and implicated in promoting cancer hallmarks and tumor progression.
Collapse
Affiliation(s)
- Daniela Spano
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Antonino Colanzi
- Institute for Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy;
| |
Collapse
|
9
|
Yamazaki Y, Eura Y, Kokame K. V-ATPase V0a1 promotes Weibel-Palade body biogenesis through the regulation of membrane fission. eLife 2021; 10:71526. [PMID: 34904569 PMCID: PMC8718113 DOI: 10.7554/elife.71526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/13/2021] [Indexed: 01/09/2023] Open
Abstract
Membrane fission, the division of a membrane-bound structure into two discrete compartments, is essential for diverse cellular events, such as endocytosis and vesicle/granule biogenesis; however, the process remains unclear. The hemostatic protein von Willebrand factor is produced in vascular endothelial cells and packaged into specialized secretory granules, Weibel–Palade bodies (WPBs) at the trans-Golgi network (TGN). Here, we reported that V0a1, a V-ATPase component, is required for the membrane fission of WPBs. We identified two V0a isoforms in distinct populations of WPBs in cultured endothelial cells, V0a1 and V0a2, on mature and nascent WPBs, respectively. Although WPB buds were formed, WPBs could not separate from the TGN in the absence of V0a1. Screening using dominant–negative forms of known membrane fission regulators revealed protein kinase D (PKD) as an essential factor in biogenesis of WPBs. Further, we showed that the induction of wild-type PKDs in V0a1-depleted cells does not support the segregation of WPBs from the TGN; suggesting a primary role of V0a1 in the membrane fission of WPBs. The identification of V0a1 as a new membrane fission regulator should facilitate the understanding of molecular events that enable membrane fission.
Collapse
Affiliation(s)
- Yasuo Yamazaki
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yuka Eura
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Koichi Kokame
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
10
|
The PKD-Dependent Biogenesis of TGN-to-Plasma Membrane Transport Carriers. Cells 2021; 10:cells10071618. [PMID: 34203456 PMCID: PMC8303525 DOI: 10.3390/cells10071618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 01/30/2023] Open
Abstract
Membrane trafficking is essential for processing and transport of proteins and lipids and to establish cell compartmentation and tissue organization. Cells respond to their needs and control the quantity and quality of protein secretion accordingly. In this review, we focus on a particular membrane trafficking route from the trans-Golgi network (TGN) to the cell surface: protein kinase D (PKD)-dependent pathway for constitutive secretion mediated by carriers of the TGN to the cell surface (CARTS). Recent findings highlight the importance of lipid signaling by organelle membrane contact sites (MCSs) in this pathway. Finally, we discuss our current understanding of multiple signaling pathways for membrane trafficking regulation mediated by PKD, G protein-coupled receptors (GPCRs), growth factors, metabolites, and mechanosensors.
Collapse
|
11
|
Abstract
Exosomes are nanoscale extracellular vesicles that can transport cargos of proteins, lipids, DNA, various RNA species and microRNAs (miRNAs). Exosomes can enter cells and deliver their contents to recipient cell. Owing to their cargo exosomes can transfer different molecules to the target cells and change the phenotype of these cells. The fate of the contents of an exosome depends on its target destination. Various mechanisms for exosome uptake by target cells have been proposed, but the mechanisms responsible for exosomes internalization into cells are still debated. Exosomes exposed cells produce labeled protein kinases, which are expressed by other cells. This means that these kinases are internalized by exosomes, and transported into the cytoplasm of recipient cells. Many studies have confirmed that exosomes are not only secreted by living cells, but also internalized or accumulated by the other cells. The "next cell hypothesis" supports the notion that exosomes constitute communication vehicles between neighboring cells. By this mechanism, exosomes participate in the development of diabetes and its associated complications, critically contribute to the spreading of neuronal damage in Alzheimer's disease, and non-proteolysed form of Fas ligand (mFasL)-bearing exosomes trigger the apoptosis of T lymphocytes. Furthermore, exosomes derived from human B lymphocytes induce antigen-specific major histocompatibility complex (MHC) class II-restricted T cell responses. Interestingly, exosomes secreted by cancer cells have been demonstrated to express tumor antigens, as well as immune suppressive molecules. This process is defined as "exosome-immune suppression" concept. The interplay via the exchange of exosomes between cancer cells and between cancer cells and the tumor stroma promote the transfer of oncogenes and onco-miRNAs from one cell to other. Circulating exosomes that are released from hypertrophic adipocytes are effective in obesity-related complications. On the other hand, the "inflammasome-induced" exosomes can activate inflammatory responses in recipient cells. In this chapter protein kinases-related checkpoints are emphasized considering the regulation of exosome biogenesis, secretory traffic, and their impacts on cell death, tumor growth, immune system, and obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey.
| |
Collapse
|
12
|
Li M, Rehman AU, Liu Y, Chen K, Lu S. Dual roles of ATP-binding site in protein kinases: Orthosteric inhibition and allosteric regulation. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 124:87-119. [PMID: 33632471 DOI: 10.1016/bs.apcsb.2020.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein kinases use ATP to phosphorylate other proteins. Phosphorylation (p) universally orchestrates a fine-tuned network modulating a multitude of biological processes. Moreover, the start of networks, ATP-binding site, has been recognized dual roles to impact protein kinases function: (i) orthosteric inhibition, via being blocked to interference ATP occupying and (ii) allosteric regulation, via being altered first to induce further conformational changes. The above two terminologies are widely used in drug design, which has acquired quite a significant progress in the protein kinases field over the past 2 decades. Most small molecular inhibitors directly compete with ATP to implement orthosteric inhibition, still exhibiting irreplaceable and promising therapeutic effects. Additionally, numerous inhibitors can paradoxically lead protein kinases to hyperphosphorylation, even activation, indicative of the allosteric regulation role of the ATP-binding site. Here, we review the quintessential examples that apply for the dual roles in diverse ways. Our work provides an insight into the molecular mechanisms under the dual roles and will be promisingly instructive for future drug development.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ashfaq Ur Rehman
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yaqin Liu
- Medicinal Chemistry and Bioinformatics Centre, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Kai Chen
- Department of Orthopedics, Changhai Hospital, Naval Military Medical University, Shanghai, China.
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; Medicinal Chemistry and Bioinformatics Centre, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Wang J, Chen Y, Chen L, Duan Y, Kuang X, Peng Z, Li C, Li Y, Xiao Y, Jin H, Tan Q, Zhang S, Zhu B, Tang Y. EGCG modulates PKD1 and ferroptosis to promote recovery in ST rats. Transl Neurosci 2020; 11:173-181. [PMID: 33335755 PMCID: PMC7712186 DOI: 10.1515/tnsci-2020-0119] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background Spinal cord injury (SCI) causes devastating loss of function and neuronal death without effective treatment. (−)-Epigallocatechin-3-gallate (EGCG) has antioxidant properties and plays an essential role in the nervous system. However, the underlying mechanism by which EGCG promotes neuronal survival and functional recovery in complete spinal cord transection (ST) remains unclear. Methods In the present study, we established primary cerebellar granule neurons (CGNs) and a T10 ST rat model to investigate the antioxidant effects of EGCG via its modulation of protein kinase D1 (PKD1) phosphorylation and inhibition of ferroptosis. Results We revealed that EGCG significantly increased the cell survival rate of CGNs and PKD1 phosphorylation levels in comparison to the vehicle control, with a maximal effect observed at 50 µM. EGCG upregulated PKD1 phosphorylation levels and inhibited ferroptosis to reduce the cell death of CGNs under oxidative stress and to promote functional recovery and ERK phosphorylation in rats following complete ST. Conclusion Together, these results lay the foundation for EGCG as a novel strategy for the treatment of SCI related to PKD1 phosphorylation and ferroptosis.
Collapse
Affiliation(s)
- Jianjun Wang
- Affiliated Hospital, Xiangnan University, Chenzhou 423000, Hunan Province, China.,Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Ying Chen
- Jilong Union School of Hengnan County, Hengyang 421000, Hunan Province, China
| | - Long Chen
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Yanzhi Duan
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Xuejun Kuang
- Affiliated Hospital, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Zhao Peng
- Affiliated Hospital, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Conghui Li
- Affiliated Hospital, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Yuanhao Li
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Yang Xiao
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Hao Jin
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Quandan Tan
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Shaofeng Zhang
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Bopei Zhu
- Department of Clinical, Xiangnan University, Chenzhou 423000, Hunan Province, China
| | - Yinjuan Tang
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou 423000, Hunan Province, China
| |
Collapse
|
14
|
Chen S, Jiang Q, Huang P, Hu C, Shen H, Schachner M, Zhao W. The L1 cell adhesion molecule affects protein kinase D1 activity in the cerebral cortex in a mouse model of Alzheimer's disease. Brain Res Bull 2020; 162:141-150. [PMID: 32540419 DOI: 10.1016/j.brainresbull.2020.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is characterized by deposition of β-amyloid protein (Aβ), neurofibrillary tangles and cognitive deficits resulting from neuronal cell death. In search for the molecular underpinnings of the disease, we were interested in the relationship between Aβ, L1 cell adhesion molecule and protein kinase D1 (PKD1), which are not only implicated in neural development and functional maintenance in the adult, but are also neuroprotective under pathological conditions. Based on our observations that L1 and phosphorylated, i.e. activated, protein kinase PKD1 (pPKD1) co-localize in cultured neurons, we investigated the functional relationship between L1 and pPKD1 in the frontal lobe of an AD human cortical tissue microarray, and found increased and positively correlating levels of both molecules when compared to a non-affected human brain. Also in the APPSWE mouse model of AD, L1 and pPKD1 levels were increased in the frontal lobe. To investigate whether L1 influences PKD1-based functions in AD, cultured cortical neurons were stressed with either H2O2 or oligomeric Aβ1-42, in the presence or absence of recombinant L1 extracellular domain, and PKD1 phosphorylation was measured. As indicated by the cell viability assay, L1 maintained neuronal survival under oxidative stress and under application of oligomeric Aβ1-42, when PKD1 activity was inhibited, suggesting that L1 ameliorates some aspects of Aβ1-42 pathology in parallel with reducing PKD1 function.
Collapse
Affiliation(s)
- Shuangxi Chen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China; The First Affiliated Hospital of University of South China, University of South China, No. 69, Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Qiong Jiang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Peizhi Huang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Chengliang Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Huifan Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
15
|
Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan Delgado B, Reynolds LE, Gomez-Escudero J, Newport E, Rajeeve V, Baker AM, Roy-Luzarraga M, Graham TA, Foster J, Wang Y, Campbell JJ, Singh R, Zhang P, Schall TJ, Balkwill FR, Sosabowski J, Cutillas PR, Frezza C, Sancho P, Hodivala-Dilke K. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun 2020; 11:1290. [PMID: 32157087 PMCID: PMC7064590 DOI: 10.1038/s41467-020-15104-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence suggests that cancer cell metabolism can be regulated by cancer-associated fibroblasts (CAFs), but the mechanisms are poorly defined. Here we show that CAFs regulate malignant cell metabolism through pathways under the control of FAK. In breast and pancreatic cancer patients we find that low FAK expression, specifically in the stromal compartment, predicts reduced overall survival. In mice, depletion of FAK in a subpopulation of CAFs regulates paracrine signals that increase malignant cell glycolysis and tumour growth. Proteomic and phosphoproteomic analysis in our mouse model identifies metabolic alterations which are reflected at the transcriptomic level in patients with low stromal FAK. Mechanistically we demonstrate that FAK-depletion in CAFs increases chemokine production, which via CCR1/CCR2 on cancer cells, activate protein kinase A, leading to enhanced malignant cell glycolysis. Our data uncover mechanisms whereby stromal fibroblasts regulate cancer cell metabolism independent of genetic mutations in cancer cells.
Collapse
Affiliation(s)
- Fevzi Demircioglu
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jun Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Juliana Candido
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ana S H Costa
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Pedro Casado
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Beatriz de Luxan Delgado
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jesus Gomez-Escudero
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Emma Newport
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Vinothini Rajeeve
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ann-Marie Baker
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Marina Roy-Luzarraga
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Julie Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Yu Wang
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | | | - Rajinder Singh
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Penglie Zhang
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Thomas J Schall
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Frances R Balkwill
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Pedro R Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Patricia Sancho
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
16
|
Maier D, Nagel AC, Preiss A. Genetic interactions between Protein Kinase D and Lobe mutants during eye development of Drosophila melanogaster. Hereditas 2019; 156:37. [PMID: 31889943 PMCID: PMC6924039 DOI: 10.1186/s41065-019-0113-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Background In Drosophila, the development of the fly eye involves the activity of several, interconnected pathways that first define the presumptive eye field within the eye anlagen, followed by establishment of the dorso-ventral boundary, and the regulation of growth and apoptosis. In Lobe (L) mutant flies, parts of the eye or even the complete eye are absent because the eye field has not been properly defined. Manifold genetic interactions indicate that L influences the activity of several signalling pathways, resulting in a conversion of eye tissue into epidermis, and in the induction of apoptosis. As information on the molecular nature of the L mutation is lacking, the underlying molecular mechanisms are still an enigma. Results We have identified Protein Kinase D (PKD) as a strong modifier of the L mutant phenotype. PKD belongs to the PKC/CAMK class of Ser/Thr kinases that have been involved in diverse cellular processes including stress resistance and growth. Despite the many roles of PKD, Drosophila PKD null mutants are without apparent phenotype apart from sensitivity to oxidative stress. Here we report an involvement of PKD in eye development in the sensitized genetic background of Lobe. Absence of PKD strongly enhanced the dominant eye defects of heterozygous L2 flies, and decreased their viability. Moreover, eye-specific overexpression of an activated isoform of PKD considerably ameliorated the dominant L2 phenotype. This genetic interaction was not allele specific but similarly seen with three additional, weaker L alleles (L1, L5, LG), demonstrating its specificity. Conclusions We propose that PKD-mediated phosphorylation is involved in underlying processes causing the L phenotype, i.e. in the regulation of growth, the epidermal transformation of eye tissue and apoptosis, respectively.
Collapse
Affiliation(s)
- Dieter Maier
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anja C Nagel
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anette Preiss
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
17
|
Wang JN, Fan YP, Chen J, Feng Y, Cui BM, Li XY, Wang LW, Chen HL, Zhang P, Wu HK. [Role of protein kinase D1 in regulating the growth, apoptosis and drug sensitivity of oral squamous carcinoma cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:583-588. [PMID: 31875434 DOI: 10.7518/hxkq.2019.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study aimed to investigate the role of protein kinase D (PKD)1 in regulating the growth, apop-tosis, and drug sensitivity of the squamous carcinoma cell line SCC-25. METHODS The SCC-25 cell line was transfected with either the control-shRNA or PKD1-shRNA plasmids. The stable transfected cells were selected, and the efficiency of PKD1 knockdown was detected by Western blot. The growth and apoptosis of SCC-25 were analyzed with a cell counting kit-8 (CCK8) and flow cytometry. The 50% inhibitory concentrations (IC50) of paclitaxel in the control and PKD1 knockdown cell lines were detected by CCK-8. The expression levels of Bax, Bcl-2, and P-gp were detected by Western blot. RESULTS PKD1 was constitutively expressed and phosphorylated in various cancer cell lines. Inhibiting the expression of PKD1 in SCC-25 cells by RNA interference could inhibit the growth and promote the apoptosis of SCC-25 cells via downregulating Bcl-2 expression. Additionally, inhibiting PKD1 expression could downregulate the expression of P-gp, thereby decreasing both the IC50 and resistance index of paclitaxel. CONCLUSIONS PKD1 plays an important role in regulating the biobehavior of SCC-25. It is a potential therapeutic target for oral squamous carcinoma.
Collapse
Affiliation(s)
- Jing-Nan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ya-Ping Fan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiao Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yun Feng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo-Miao Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiao-Ying Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li-Wei Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hong-Li Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ping Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hong-Kun Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Wu S, Ma S, Yin X, Yi P, Liu J. An integrated PKD1-dependent signaling network amplifies IRE1 prosurvival signaling. J Biol Chem 2019; 294:11119-11130. [PMID: 31167779 DOI: 10.1074/jbc.ra118.003311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Following the accumulation of improperly folded proteins in the endoplasmic reticulum (ER), a condition known as ER stress in this compartment triggers an adaptive signaling pathway referred to as the unfolded protein response (UPR). The UPR aims at restoring ER homeostasis; if the ER stress cannot be resolved, apoptosis is triggered. However, the mechanisms responsible for regulating the balance between cell life and death decisions that occur after exposure to ER stress remain unclear. Protein kinase D1 (PKD1) has been reported to initiate protective signaling against oxidative stress or ischemia, two conditions that impinge on the induction of ER stress. In addition, the high levels of expression of PKD1, observed in highly proliferative cancers and tumors with poor prognosis, contribute to enhanced resistance to chemotherapy. In this study, we show that the ER stress inducers tunicamycin and thapsigargin lead to the activation of PKD1 in human prostate cancer PC-3 cells and in hepatoma HepG2 cells through a PKCδ-dependent mechanism. Moreover, our data indicate that PKD1 is required for the stabilization of inositol-requiring enzyme 1 (IRE1) and the subsequent regulation of its activity. PKD1 activation contributes to the phosphorylation of mitogen-activated protein kinase phosphatase 1, resulting in decreased IRE1-mediated c-Jun N-terminal kinase activation. This study unveils the existence of a novel PKD1-dependent prosurvival mechanism that is activated upon ER stress and selectively enhances IRE1 prosurvival signaling.
Collapse
Affiliation(s)
- Shiyong Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, Hubei, China
| | - Shumin Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, Hubei, China
| | - Xueliang Yin
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, Hubei, China
| | - Ping Yi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, Hubei, China
| | - Jianfeng Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, Hubei, China
| |
Collapse
|
19
|
Bollag WB, Ding KH, Choudhary V, Xu J, Zhong Q, Elsayed R, Bailey LJ, Elsalanty M, Yu K, Johnson MH, McGee-Lawrence ME, Isales CM. Protein kinase D1 conditional null mice show minimal bone loss following ovariectomy. Mol Cell Endocrinol 2018; 474. [PMID: 29530783 PMCID: PMC6733406 DOI: 10.1016/j.mce.2018.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We previously found that 3- and 6-month-old male mice with conditional ablation of protein kinase D1 (PRKD1) in osteoprogenitor cells (expressing Osterix) exhibited reduced bone mass. Others have demonstrated similar effects in young female PRKD1-deficient mice. Here we examined the bone resorptive response of adult female floxed control and conditional knockout (cKO) mice undergoing sham surgery or ovariectomy (OVX). Femoral and tibial bone mineral density (BMD) values were significantly reduced upon OVX in control, but not cKO, females compared to the respective sham-operated mice. Micro-CT analysis showed that OVX significantly increased trabecular number and decreased trabecular spacing in cKO but not control mice. Finally, in control mice serum levels of a marker of bone resorption (pyridinoline crosslinks) and the osteoclast activator RANKL significantly increased upon OVX; however, no such OVX-induced increase was observed in cKO mice. Our results suggest the potential importance of PRKD1 in response to estrogen loss in bone.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Physiology, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States.
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Vivek Choudhary
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Department of Physiology, Augusta University, Augusta, GA 30912, United States
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Ranya Elsayed
- Department of Oral Biology, Augusta University, Augusta, GA 30912, United States
| | - Lakiea J Bailey
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Mohammed Elsalanty
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Oral Biology, Augusta University, Augusta, GA 30912, United States
| | - Kanglun Yu
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - Maribeth H Johnson
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Meghan E McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
20
|
Azoitei N, Cobbaut M, Becher A, Van Lint J, Seufferlein T. Protein kinase D2: a versatile player in cancer biology. Oncogene 2017; 37:1263-1278. [PMID: 29259300 DOI: 10.1038/s41388-017-0052-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022]
Abstract
Protein kinase D2 (PKD2) is a serine/threonine kinase that belongs to the PKD family of calcium-calmodulin kinases, which comprises three isoforms: PKD1, PKD2, and PKD3. PKD2 is activated by many stimuli including growth factors, phorbol esters, and G-protein-coupled receptor agonists. PKD2 participation to uncontrolled growth, survival, neovascularization, metastasis, and invasion has been documented in various tumor types including pancreatic, colorectal, gastric, hepatic, lung, prostate, and breast cancer, as well as glioma multiforme and leukemia. This review discusses the versatile functions of PKD2 from the perspective of cancer hallmarks as described by Hanahan and Weinberg. The PKD2 status, signaling pathways affected in different tumor types and the molecular mechanisms that lead to tumorigenesis and tumor progression are presented. The latest developments of small-molecule inhibitors selective for PKD/PKD2, as well as the need for further chemotherapies that prevent, slow down, or eliminate tumors are also discussed in this review.
Collapse
Affiliation(s)
- Ninel Azoitei
- Center for Internal Medicine I, University of Ulm, Ulm, Germany.
| | - Mathias Cobbaut
- Laboratory for Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - Johan Van Lint
- Laboratory for Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
21
|
Chadha N, Bahia MS, Kaur M, Bahadur R, Silakari O. Computational design of new protein kinase D 1 (PKD1) inhibitors: homology-based active site prediction, energy-optimized pharmacophore, docking and database screening. Mol Divers 2017; 22:47-56. [PMID: 29058231 DOI: 10.1007/s11030-017-9785-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/11/2017] [Indexed: 01/11/2023]
Abstract
Protein kinase D 1 (PKD1) overexpression has a well-validated role in cancer progression and its inhibitors have defined a protective role-play of PKD1 for various cancers such as prostate, pancreatic and noninvasive breast cancers, and more. Therefore, the current research was aimed at designing new PKD1 inhibitors combining different ligand- and structure-based computational drug designing methodologies. Initially, the three-dimensional structure of PKD1's active site was computationally modeled, corrected using molecular dynamic simulations and validated for docking experiments. The highest active PKD1 inhibitor was used to develop a structure-based energetic pharmacophore (e-pharmacophore) model, and a final model was selected with five structural features (Pmodel_AADHR). Pmodel_AADHR was validated and used for database screening to obtain new hits against PKD1. These newly retrieved hits were docked against our PKD1 protein model, and those displaying essential interactions are reported herein as new hits, which could serve as new leads for cancer research, especially pancreatic cancer.
Collapse
Affiliation(s)
- Navriti Chadha
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Malkeet Singh Bahia
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Maninder Kaur
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Renu Bahadur
- Bioinformatics Division, Indian Council of Medical Research (ICMR), New Delhi, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
22
|
Shiwarski DJ, Darr M, Telmer CA, Bruchez MP, Puthenveedu MA. PI3K class II α regulates δ-opioid receptor export from the trans-Golgi network. Mol Biol Cell 2017; 28:2202-2219. [PMID: 28566554 PMCID: PMC5531736 DOI: 10.1091/mbc.e17-01-0030] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/26/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
The interplay between signaling and trafficking by G protein-coupled receptors (GPCRs) has focused mainly on endocytic trafficking. Whether and how surface delivery of newly synthesized GPCRs is regulated by extracellular signals is less understood. Here we define a signaling-regulated checkpoint at the trans-Golgi network (TGN) that controls the surface delivery of the delta opioid receptor (δR). In PC12 cells, inhibition of phosphoinositide-3 kinase (PI3K) activity blocked export of newly synthesized δR from the Golgi and delivery to the cell surface, similar to treatment with nerve growth factor (NGF). Depletion of class II phosphoinositide-3 kinase α (PI3K C2A), but not inhibition of class I PI3K, blocked δR export to comparable levels and attenuated δR-mediated cAMP inhibition. NGF treatment displaced PI3K C2A from the Golgi and optogenetic recruitment of the PI3K C2A kinase domain to the TGN-induced δR export downstream of NGF. Of importance, PI3K C2A expression promotes export of endogenous δR in primary trigeminal ganglion neurons. Taken together, our results identify PI3K C2A as being required and sufficient for δR export and surface delivery in neuronal cells and suggest that it could be a key modulator of a novel Golgi export checkpoint that coordinates GPCR delivery to the surface.
Collapse
Affiliation(s)
- Daniel J Shiwarski
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213.,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Marlena Darr
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Cheryl A Telmer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Marcel P Bruchez
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213.,Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213.,Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Manojkumar A Puthenveedu
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213 .,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
23
|
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
|
24
|
Jain A, Holthuis JCM. Membrane contact sites, ancient and central hubs of cellular lipid logistics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1450-1458. [PMID: 28554771 DOI: 10.1016/j.bbamcr.2017.05.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 12/26/2022]
Abstract
Membrane contact sites (MCSs) are regions where two organelles are closely apposed to facilitate molecular communication and promote a functional integration of compartmentalized cellular processes. There is growing evidence that MCSs play key roles in controlling intracellular lipid flows and distributions. Strikingly, even organelles connected by vesicular trafficking exchange lipids en bulk via lipid transfer proteins that operate at MCSs. Herein, we describe how MCSs developed into central hubs of lipid logistics during the evolution of eukaryotic cells. We then focus on how modern eukaryotes exploit MCSs to help solve a major logistical problem, namely to preserve the unique lipid mixtures of their early and late secretory organelles in the face of extensive vesicular trafficking. This article is part of a Special Issue entitled: Membrane Contact Sites edited by Christian Ungermann and Benoit Kornmann.
Collapse
Affiliation(s)
- Amrita Jain
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany; Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
25
|
Cobbaut M, Derua R, Döppler H, Lou HJ, Vandoninck S, Storz P, Turk BE, Seufferlein T, Waelkens E, Janssens V, Van Lint J. Differential regulation of PKD isoforms in oxidative stress conditions through phosphorylation of a conserved Tyr in the P+1 loop. Sci Rep 2017; 7:887. [PMID: 28428613 PMCID: PMC5430542 DOI: 10.1038/s41598-017-00800-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
Protein kinases are essential molecules in life and their crucial function requires tight regulation. Many kinases are regulated via phosphorylation within their activation loop. This loop is embedded in the activation segment, which additionally contains the Mg2+ binding loop and a P + 1 loop that is important in substrate binding. In this report, we identify Abl-mediated phosphorylation of a highly conserved Tyr residue in the P + 1 loop of protein kinase D2 (PKD2) during oxidative stress. Remarkably, we observed that the three human PKD isoforms display very different degrees of P + 1 loop Tyr phosphorylation and we identify one of the molecular determinants for this divergence. This is paralleled by a different activation mechanism of PKD1 and PKD2 during oxidative stress. Tyr phosphorylation in the P + 1 loop of PKD2 increases turnover for Syntide-2, while substrate specificity and the role of PKD2 in NF-κB signaling remain unaffected. Importantly, Tyr to Phe substitution renders the kinase inactive, jeopardizing its use as a non-phosphorylatable mutant. Since large-scale proteomics studies identified P + 1 loop Tyr phosphorylation in more than 70 Ser/Thr kinases in multiple conditions, our results do not only demonstrate differential regulation/function of PKD isoforms under oxidative stress, but also have implications for kinase regulation in general.
Collapse
Affiliation(s)
- Mathias Cobbaut
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Rita Derua
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sandy Vandoninck
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Etienne Waelkens
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Veerle Janssens
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium. .,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium.
| |
Collapse
|
26
|
Investigation of the Role of Protein Kinase D in Human Rhinovirus Replication. J Virol 2017; 91:JVI.00217-17. [PMID: 28228588 PMCID: PMC5391474 DOI: 10.1128/jvi.00217-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/19/2022] Open
Abstract
Picornavirus replication is known to cause extensive remodeling of Golgi and endoplasmic reticulum membranes, and a number of the host proteins involved in the viral replication complex have been identified, including oxysterol binding protein (OSBP) and phosphatidylinositol 4-kinase III beta (PI4KB). Since both OSBP and PI4KB are substrates for protein kinase D (PKD) and PKD is known to be involved in the control of Golgi membrane vesicular and lipid transport, we hypothesized that PKD played a role in viral replication. We present multiple lines of evidence in support of this hypothesis. First, infection of HeLa cells with human rhinovirus (HRV) induced the phosphorylation of PKD. Second, PKD inhibitors reduced HRV genome replication, protein expression, and titers in a concentration-dependent fashion and also blocked the replication of poliovirus (PV) and foot-and-mouth disease virus (FMDV) in a variety of cells. Third, HRV replication was significantly reduced in HeLa cells overexpressing wild-type and mutant forms of PKD1. Fourth, HRV genome replication was reduced in HAP1 cells in which the PKD1 gene was knocked out by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9. Although we have not identified the molecular mechanism through which PKD regulates viral replication, our data suggest that this is not due to enhanced interferon signaling or an inhibition of clathrin-mediated endocytosis, and PKD inhibitors do not need to be present during viral uptake. Our data show for the first time that targeting PKD with small molecules can inhibit the replication of HRV, PV, and FMDV, and therefore, PKD may represent a novel antiviral target for drug discovery. IMPORTANCE Picornaviruses remain an important family of human and animal pathogens for which we have a very limited arsenal of antiviral agents. HRV is the causative agent of the common cold, which in itself is a relatively trivial infection; however, in asthma and chronic obstructive pulmonary disease (COPD) patients, this virus is a major cause of exacerbations resulting in an increased use of medication, worsening symptoms, and, frequently, hospital admission. Thus, HRV represents a substantial health care and economic burden for which there are no approved therapies. We sought to identify a novel host target as a potential anti-HRV therapy. HRV infection induces the phosphorylation of PKD, and inhibitors of this kinase effectively block HRV replication at an early stage of the viral life cycle. Moreover, PKD inhibitors also block PV and FMDV replication. This is the first description that PKD may represent a target for antiviral drug discovery.
Collapse
|
27
|
Verschueren K, Cobbaut M, Demaerel J, Saadah L, Voet ARD, Van Lint J, De Borggraeve WM. Discovery of a potent protein kinase D inhibitor: insights in the binding mode of pyrazolo[3,4- d]pyrimidine analogues. MEDCHEMCOMM 2017; 8:640-646. [PMID: 28890776 PMCID: PMC5567267 DOI: 10.1039/c6md00675b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/31/2017] [Indexed: 12/17/2022]
Abstract
In this study, we set out to rationally optimize PKD inhibitors based on the pyrazolo[3,4-d]pyrimidine scaffold. The lead compound for this study was 1-NM-PP1, which was previously found by us and others to inhibit PKD. In our screening we identified one compound (3-IN-PP1) displaying a 10-fold increase in potency over 1-NM-PP1, opening new possibilities for specific protein kinase inhibitors for kinases that show sensitivity towards pyrazolo[3,4-d]pyrimidine derived compounds. Interestingly the observed SAR was not in complete agreement with the commonly observed binding mode where the pyrazolo[3,4-d]pyrimidine compounds are bound in a similar fashion as PKD's natural ligand ATP. Therefore we suggest an alternate binding mode where the compounds are flipped 180 degrees. This possible alternate binding mode for pyrazolo[3,4-d]pyrimidine based compounds could pave the way for a new class of specific protein kinase inhibitors for kinases sensitive towards pyrazolo[3,4-d]pyrmidines.
Collapse
Affiliation(s)
- Klaas Verschueren
- Department of Chemistry , Molecular Design and Synthesis , KU Leuven , Celestijnenlaan 200F , 3001 Leuven , Belgium .
| | - Mathias Cobbaut
- Department of Cellular and Molecular Medicine , Laboratory of Protein Phosphorylation and Proteomics , KU Leuven , Herestraat 49 box 901 , 3000 Leuven , Belgium
| | - Joachim Demaerel
- Department of Chemistry , Molecular Design and Synthesis , KU Leuven , Celestijnenlaan 200F , 3001 Leuven , Belgium .
| | - Lina Saadah
- Department of Cellular and Molecular Medicine , Laboratory of Protein Phosphorylation and Proteomics , KU Leuven , Herestraat 49 box 901 , 3000 Leuven , Belgium
| | - Arnout R D Voet
- Department of Chemistry , Laboratory of Biomolecular Modeling and Design , KU Leuven , Celestijnenlaan 200G , 3001 Leuven , Belgium
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine , Laboratory of Protein Phosphorylation and Proteomics , KU Leuven , Herestraat 49 box 901 , 3000 Leuven , Belgium
| | - Wim M De Borggraeve
- Department of Chemistry , Molecular Design and Synthesis , KU Leuven , Celestijnenlaan 200F , 3001 Leuven , Belgium .
| |
Collapse
|
28
|
Kang HB, Lee HR, Jee DJ, Shin SH, Nah SS, Yoon SY, Kim JW. PRDM1, a Tumor-Suppressor Gene, is Induced by Genkwadaphnin in Human Colon Cancer SW620 Cells. J Cell Biochem 2016; 117:172-9. [PMID: 26096175 DOI: 10.1002/jcb.25262] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/16/2015] [Indexed: 12/30/2022]
Abstract
Genkwadaphnin (GD-1) is isolated from the flower buds of Daphne genkwa Siebold et Zuccarini (Thymelaeaceae), and it has been used as a traditional Korean and Chinese medicine. In this study, the authors observe that GD-1 inhibits the growth of the colon cancer cell line, SW620, through the up-regulation of p21 expression in a PRDM1-dependent manner. After treatment with GD-1, the transcriptional repressor PRDM1 is prominently induced in SW620 cells. Furthermore, GD-1 induce the phosphorylation of PKD1 and MEK and subsequently provide PRDM1 enhancement, resulting in the suppression of c-Myc expression and the up-regulation of p21. PKD1 knockdown using siRNA abrogates PRDM1 expression by GD-1 and subsequently disrupts the regulation of c-Myc and p21 expression. Treating SW620 cells with GD-1 inhibits cell-cycle progression and is characterized by the down-regulation of c-Myc followed by the up-regulation of p21 expression. The up-regulation of p21 by GD-1 induces the growth arrest of the SW620 colon cancer cell line. Based on these data, the authors propose that GD-1 has tumor-suppressor activity that may contribute to the anti-tumor effects of PRDM1 in colon cancer.
Collapse
Affiliation(s)
- Ho-Bum Kang
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Ha-Reum Lee
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Da Jung Jee
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Su-Hyun Shin
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Soonchunhyang Medical Science Research Institute, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Seong-Su Nah
- Head of Rheumatology, Department of Internal Medicine, Soonchunhyang University Choenan hospital College of Medicine, Choenan, 330-721, Republic of Korea
| | - Sun Young Yoon
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon, 305-732, Republic of Korea
| | - Jae Wha Kim
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|
29
|
Tsutsuki H, Yahiro K, Ogura K, Ichimura K, Iyoda S, Ohnishi M, Nagasawa S, Seto K, Moss J, Noda M. Subtilase cytotoxin produced by locus of enterocyte effacement-negative Shiga-toxigenic Escherichia coli induces stress granule formation. Cell Microbiol 2016; 18:1024-40. [PMID: 26749168 PMCID: PMC10068837 DOI: 10.1111/cmi.12565] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/24/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022]
Abstract
Subtilase cytotoxin (SubAB) is mainly produced by locus of enterocyte effacement (LEE)-negative strains of Shiga-toxigenic Escherichia coli (STEC). SubAB cleaves an endoplasmic reticulum (ER) chaperone, BiP/Grp78, leading to induction of ER stress. This stress causes activation of ER stress sensor proteins and induction of caspase-dependent apoptosis. We found that SubAB induces stress granules (SG) in various cells. Aim of this study was to explore the mechanism by which SubAB induced SG formation. Here, we show that SubAB-induced SG formation is regulated by activation of double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK). The culture supernatant of STEC O113:H21 dramatically induced SG in Caco2 cells, although subAB knockout STEC O113:H21 culture supernatant did not. Treatment with phorbol 12-myristate 13-acetate (PMA), a protein kinase C (PKC) activator, and lysosomal inhibitors, NH4 Cl and chloroquine, suppressed SubAB-induced SG formation, which was enhanced by PKC and PKD inhibitors. SubAB attenuated the level of PKD1 phosphorylation. Depletion of PKCδ and PKD1 by siRNA promoted SG formation in response to SubAB. Furthermore, death-associated protein 1 (DAP1) knockdown increased basal phospho-PKD1(S916) and suppressed SG formation by SubAB. However, SG formation by an ER stress inducer, Thapsigargin, was not inhibited in PMA-treated cells. Our findings show that SubAB-induced SG formation is regulated by the PERK/DAP1 signalling pathway, which may be modulated by PKCδ/PKD1, and different from the signal transduction pathway that results in Thapsigargin-induced SG formation.
Collapse
Affiliation(s)
- Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kohei Ogura
- Pathogenic Microbe Laboratory, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Kimitoshi Ichimura
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sunao Iyoda
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Ohnishi
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sayaka Nagasawa
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuko Seto
- Division of Bacteriology, Osaka Prefectural Institute of Public Health, Osaka, Japan
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Masatoshi Noda
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
30
|
Manukjan G, Ripperger T, Santer L, von Neuhoff N, Ganser A, Schambach A, Schlegelberger B, Steinemann D. Expression of the ETS transcription factor GABPα is positively correlated to the BCR-ABL1/ABL1 ratio in CML patients and affects imatinib sensitivity in vitro. Exp Hematol 2015; 43:880-90. [PMID: 26072332 DOI: 10.1016/j.exphem.2015.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/18/2015] [Accepted: 05/23/2015] [Indexed: 11/18/2022]
Abstract
In Philadelphia-positive chronic myeloid leukemia (CML), imatinib resistance frequently emerges because of point mutations in the ABL1 kinase domain, but may also be the consequence of uncontrolled upstream signaling. Recently, the heteromeric transcription factor GA-binding protein (GABP) was found to promote CML-like myeloproliferative disease in mice. In a cohort of 70 CML patients, we found that expression of the GABP α subunit (GABPα) is positively correlated to the BCR-ABL1/ABL1 ratio. Moreover, significantly higher GABPα expression was detected in blast crisis than in chronic phase CML after performing data mining on 91 CML patients. In functional studies, imatinib sensitivity is enhanced after GABPα knockdown in tyrosine kinase inhibitors (TKI)-sensitive K-562, as well as by overexpression of a deletion mutant in TKI-resistant NALM-1 cells. Moreover, in K-562 cells, GABP-dependent expression variations of PRKD2 and RAC2, relevant signaling mediators in CML, were observed. Notably, protein kinase D2 (Prkd2) was reported to be a GABP target gene in mice. In line with this, we detected a positive correlation between GABPA and PRKD2 expression in primary human CML, indicating that the effects of GABP are mediated by PRKD2. These findings illustrate an important role for GABP in disease development and imatinib sensitivity in human CML.
Collapse
MESH Headings
- Drug Resistance, Neoplasm
- Female
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- GA-Binding Protein Transcription Factor/genetics
- GA-Binding Protein Transcription Factor/metabolism
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- Humans
- Imatinib Mesylate/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Male
- Middle Aged
- Proto-Oncogene Proteins c-ets/biosynthesis
- Proto-Oncogene Proteins c-ets/genetics
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
- RAC2 GTP-Binding Protein
Collapse
Affiliation(s)
- Georgi Manukjan
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany.
| | - Tim Ripperger
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Laura Santer
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Nils von Neuhoff
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Doris Steinemann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
31
|
Bonfim-Melo A, Zanetti BF, Ferreira ÉR, Vandoninck S, Han SW, Van Lint J, Mortara RA, Bahia D. Trypanosoma cruziextracellular amastigotes trigger the protein kinase D1-cortactin-actin pathway during cell invasion. Cell Microbiol 2015; 17:1797-810. [DOI: 10.1111/cmi.12472] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 05/31/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Alexis Bonfim-Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Bianca Ferrarini Zanetti
- Interdisciplinary Center for Gene Therapy (CINTERGEN); Universidade Federal de São Paulo (UNIFESP); São Paulo Brazil
| | - Éden Ramalho Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Sandy Vandoninck
- Department of Cellular and Molecular Medicine; University of Leuven; Leuven Belgium
| | - Sang Won Han
- Interdisciplinary Center for Gene Therapy (CINTERGEN); Universidade Federal de São Paulo (UNIFESP); São Paulo Brazil
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine; University of Leuven; Leuven Belgium
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
- Departamento de Biologia Geral, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais (ICB-UFMG); Belo Horizonte Brazil
| |
Collapse
|
32
|
Müller M, Schröer J, Azoitei N, Eiseler T, Bergmann W, Köhntop R, Lin Q, Costa IG, Zenke M, Genze F, Weidgang C, Seufferlein T, Liebau S, Kleger A. A time frame permissive for Protein Kinase D2 activity to direct angiogenesis in mouse embryonic stem cells. Sci Rep 2015; 5:11742. [PMID: 26148697 PMCID: PMC4493579 DOI: 10.1038/srep11742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 05/15/2015] [Indexed: 01/18/2023] Open
Abstract
The protein kinase D isoenzymes PKD1/2/3 are prominent downstream targets of PKCs (Protein Kinase Cs) and phospholipase D in various biological systems. Recently, we identified PKD isoforms as novel mediators of tumour cell-endothelial cell communication, tumour cell motility and metastasis. Although PKD isoforms have been implicated in physiological/tumour angiogenesis, a role of PKDs during embryonic development, vasculogenesis and angiogenesis still remains elusive. We investigated the role of PKDs in germ layer segregation and subsequent vasculogenesis and angiogenesis using mouse embryonic stem cells (ESCs). We show that mouse ESCs predominantly express PKD2 followed by PKD3 while PKD1 displays negligible levels. Furthermore, we demonstrate that PKD2 is specifically phosphorylated/activated at the time of germ layer segregation. Time-restricted PKD2-activation limits mesendoderm formation and subsequent cardiovasculogenesis during early differentiation while leading to branching angiogenesis during late differentiation. In line, PKD2 loss-of-function analyses showed induction of mesendodermal differentiation in expense of the neuroectodermal germ layer. Our in vivo findings demonstrate that embryoid bodies transplanted on chicken chorioallantoic membrane induced an angiogenic response indicating that timed overexpression of PKD2 from day 4 onwards leads to augmented angiogenesis in differentiating ESCs. Taken together, our results describe novel and time-dependent facets of PKD2 during early cell fate determination.
Collapse
Affiliation(s)
- Martin Müller
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Jana Schröer
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Ninel Azoitei
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Wendy Bergmann
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Ralf Köhntop
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | - Qiong Lin
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Ivan G Costa
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, Aachen, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | | | - Clair Weidgang
- Department of Internal Medicine I, Ulm University, Ulm, Germany
| | | | - Stefan Liebau
- Institute of Neuroanatomy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | | |
Collapse
|
33
|
Protein kinase D1/2 is involved in the maturation of multivesicular bodies and secretion of exosomes in T and B lymphocytes. Cell Death Differ 2015; 23:99-109. [PMID: 26045048 DOI: 10.1038/cdd.2015.72] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 04/22/2015] [Accepted: 04/28/2015] [Indexed: 12/26/2022] Open
Abstract
Multivesicular bodies (MVBs) are endocytic compartments that enclose intraluminal vesicles (ILVs) formed by inward budding from the limiting membrane of endosomes. In T lymphocytes, these ILV contain Fas ligand (FasL) and are secreted as 'lethal exosomes' following activation-induced fusion of the MVB with the plasma membrane. Diacylglycerol (DAG) and diacylglycerol kinase α (DGKα) regulate MVB maturation and polarized traffic, as well as subsequent secretion of pro-apoptotic exosomes, but the molecular basis underlying these phenomena remains unclear. Here we identify protein kinase D (PKD) family members as DAG effectors involved in MVB genesis and secretion. We show that the inducible secretion of exosomes is enhanced when a constitutively active PKD1 mutant is expressed in T lymphocytes, whereas exosome secretion is impaired in PKD2-deficient mouse T lymphoblasts and in PKD1/3-null B cells. Analysis of PKD2-deficient T lymphoblasts showed the presence of large, immature MVB-like vesicles and demonstrated defects in cytotoxic activity and in activation-induced cell death. Using pharmacological and genetic tools, we show that DGKα regulates PKD1/2 subcellular localization and activation. Our studies demonstrate that PKD1/2 is a key regulator of MVB maturation and exosome secretion, and constitutes a mediator of the DGKα effect on MVB secretory traffic.
Collapse
|
34
|
Varga A, Gyulavári P, Greff Z, Futosi K, Németh T, Simon-Szabó L, Kerekes K, Szántai-Kis C, Brauswetter D, Kokas M, Borbély G, Erdei A, Mócsai A, Kéri G, Vántus T. Targeting vascular endothelial growth factor receptor 2 and protein kinase D1 related pathways by a multiple kinase inhibitor in angiogenesis and inflammation related processes in vitro. PLoS One 2015; 10:e0124234. [PMID: 25874616 PMCID: PMC4396990 DOI: 10.1371/journal.pone.0124234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/27/2015] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence suggests that the vascular endothelial growth factor receptor 2 (VEGFR2) and protein kinase D1 (PKD1) signaling axis plays a critical role in normal and pathological angiogenesis and inflammation related processes. Despite all efforts, the currently available therapeutic interventions are limited. Prior studies have also proved that a multiple target inhibitor can be more efficient compared to a single target one. Therefore, development of novel inflammatory pathway-specific inhibitors would be of great value. To test this possibility, we screened our molecular library using recombinant kinase assays and identified the previously described compound VCC251801 with strong inhibitory effect on both VEGFR2 and PKD1. We further analyzed the effect of VCC251801 in the endothelium-derived EA.hy926 cell line and in different inflammatory cell types. In EA.hy926 cells, VCC251801 potently inhibited the intracellular activation and signaling of VEGFR2 and PKD1 which inhibition eventually resulted in diminished cell proliferation. In this model, our compound was also an efficient inhibitor of in vitro angiogenesis by interfering with endothelial cell migration and tube formation processes. Our results from functional assays in inflammatory cellular models such as neutrophils and mast cells suggested an anti-inflammatory effect of VCC251801. The neutrophil study showed that VCC251801 specifically blocked the immobilized immune-complex and the adhesion dependent TNF-α -fibrinogen stimulated neutrophil activation. Furthermore, similar results were found in mast cell degranulation assay where VCC251801 caused significant reduction of mast cell response. In summary, we described a novel function of a multiple kinase inhibitor which strongly inhibits the VEGFR2-PKD1 signaling and might be a novel inhibitor of pathological inflammatory pathways.
Collapse
Affiliation(s)
- Attila Varga
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | - Pál Gyulavári
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | | | - Krisztina Futosi
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Tamás Németh
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Laura Simon-Szabó
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Krisztina Kerekes
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Diána Brauswetter
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Márton Kokas
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Gábor Borbély
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | - Anna Erdei
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - György Kéri
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
- Vichem Chemie Research Ltd., Budapest, Hungary
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Tibor Vántus
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
35
|
Sin YY, Martin TP, Wills L, Currie S, Baillie GS. Small heat shock protein 20 (Hsp20) facilitates nuclear import of protein kinase D 1 (PKD1) during cardiac hypertrophy. Cell Commun Signal 2015; 13:16. [PMID: 25889640 PMCID: PMC4356135 DOI: 10.1186/s12964-015-0094-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 02/13/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Nuclear import of protein kinase D1 (PKD1) is an important event in the transcriptional regulation of cardiac gene reprogramming leading to the hypertrophic growth response, however, little is known about the molecular events that govern this event. We have identified a novel complex between PKD1 and a heat shock protein (Hsp), Hsp20, which has been implicated as cardioprotective. This study aims to characterize the role of the complex in PKD1-mediated myocardial regulatory mechanisms that depend on PKD1 nuclear translocation. RESULTS In mapping the Hsp20 binding sites on PKD1 within its catalytic unit using peptide array analysis, we were able to develop a cell-permeable peptide that disrupts the Hsp20-PKD1 complex. We use this peptide to show that formation of the Hsp20-PKD1 complex is essential for PKD1 nuclear translocation, signaling mechanisms leading to hypertrophy, activation of the fetal gene programme and pathological cardiac remodeling leading to cardiac fibrosis. CONCLUSIONS These results identify a new signaling complex that is pivotal to pathological remodelling of the heart that could be targeted therapeutically.
Collapse
Affiliation(s)
- Yuan Yan Sin
- Institute of Cardiovascular and Medical sciences, CMVLS, University of Glasgow, Glasgow, G128QQ, UK.
| | - Tamara P Martin
- Institute of Cardiovascular and Medical sciences, CMVLS, University of Glasgow, Glasgow, G128QQ, UK.
| | - Lauren Wills
- Institute of Cardiovascular and Medical sciences, CMVLS, University of Glasgow, Glasgow, G128QQ, UK.
| | - Susan Currie
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Hamnett building, 161 Cathedral Street, Glasgow, G4 ORE, UK.
| | - George S Baillie
- Institute of Cardiovascular and Medical sciences, CMVLS, University of Glasgow, Glasgow, G128QQ, UK.
| |
Collapse
|
36
|
Xu X, Prough RA, Samuelson DJ. Differential 12-O-Tetradecanoylphorbol-13-acetate-induced activation of rat mammary carcinoma susceptibility Fbxo10 variant promoters via a PKC-AP1 pathway. Mol Carcinog 2015; 54:134-47. [PMID: 24008983 PMCID: PMC9733134 DOI: 10.1002/mc.22081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/26/2013] [Accepted: 08/07/2013] [Indexed: 12/13/2022]
Abstract
Rat mammary carcinoma susceptibility 5a1 (Mcs5a1), which is concordant to human MCS5A1 breast cancer risk locus, mediates susceptibility by a non-mammary cell-autonomous mechanism associated with T cell differential expression of F-box protein 10 (Fbxo10). Human FBXO10, an evolutionarily conserved ubiquitin ligase gene, was shown to have a potential role in regulating cell death by controlling the degradation of Bcl-2, a key protein involved in apoptosis. Breast cancer susceptibility is controlled by interactions between environmental and genetic factors; therefore, we sought to determine if breast cancer risk-associated environmental chemicals interact with Mcs5a1 variants using luciferase reporter constructs containing 4.2 kb Fbxo10 promoters based on alleles of mammary cancer susceptible Wistar Furth (WF) and resistant Wistar Kyoto (WKY) rat strains. 12-O-Tetradecanoylphorbol-13-acetate (TPA) induced activation of a 4.2 kb WF Fbxo10 promoter region, but lower levels of activation of the homologous WKY Fbxo10 promoter region. Using general and specific protein kinase inhibitors, we identified a protein kinase C (PKC) pathway that mediated TPA activation. We narrowed the possible PKCs to a member of the atypical PKC isoforms, namely PKCµ. We also determined that activator protein 1 (AP1) family member c-Fos mediated TPA activation of the 4.2 kb WF Fbxo10 promoter. TPA was shown to induce endogenous FBXO10 mRNA and FBXO10 protein in Jurkat cells, a human T cell line, with a maximal level of expression from 1.5 to 2.5 h after exposure. These results indicate that FBXO10/Fbxo10 expression is regulated by a PKC-dependent pathway acting through c-Fos, which binds AP1-specific DNA elements in Mcs5a1.
Collapse
Affiliation(s)
- Xin Xu
- Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Russell A. Prough
- Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky,Center for Genetics & Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - David J. Samuelson
- Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky,James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky,Center for Genetics & Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky,Correspondence to: Center for Genetics and Molecular Medicine, Department of Biochemistry & Molecular Biology, University of Louisville School of Medicine, 319 Abraham Flexner Way, HSC-A, Room 708, Louisville, KY 40292
| |
Collapse
|
37
|
PKD1 is downregulated in non-small cell lung cancer and mediates the feedback inhibition of mTORC1-S6K1 axis in response to phorbol ester. Int J Biochem Cell Biol 2015; 60:34-42. [PMID: 25578563 DOI: 10.1016/j.biocel.2014.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 11/05/2014] [Accepted: 12/25/2014] [Indexed: 12/27/2022]
Abstract
Protein kinase D1 (PKD1) is increasingly implicated in multiple biological and molecular events that regulate the proliferation or invasiveness in several cancers. However, little is known about the expression and functions of PKD1 in non-small cell lung cancer (NSCLC). In the present study, 34 pairs of human NSCLC and matched normal bronchiolar epitheliums were enrolled and evaluated for PKD1 expression by quantitative real-time PCR. We showed that PKD1 was downregulated in 26 of 34 cancer tissues in comparison with matched normal epitheliums. Moreover, patients with venous invasion or lymph node metastasis showed significant lower expression of PKD1. Exposure of NSCLC A549 and H520 cells to the PKD family inhibitor kb NB 142-70(Kb), at concentrations that inhibited PKD1 activation, strikingly potentiated S6K1 phosphorylation at Thr(389) and S6 phosphorylation at Ser(235/236) in response to phorbol ester (PMA). Knockdown of PKD1 with siRNAs strikingly enhanced S6K1 phosphorylation whereas constitutively active PKD1 resulted in the S6K1 activity inhibition. Furthermore, the PI3K inhibitors LY294002, BKM120 and MEK inhibitors U0126, PD0325901 blocked the enhanced S6K1 activity induced by Kb. Collectively, our results identify decreased expression of the PKD1 as a marker for NSCLC and the loss of PKD1 expression increases the malignant potential of NSCLC cells. This may be due to the function of PKD1 as a negative regulator of mTORC1-S6K1. Our results suggest that re-expression or activation of PKD1 might serve as a potential therapeutic target for NSCLC treatment.
Collapse
|
38
|
Chen SX, Hu CL, Liao YH, Zhao WJ. L1 modulates PKD1 phosphorylation in cerebellar granule neurons. Neurosci Lett 2015; 584:331-6. [PMID: 25445362 DOI: 10.1016/j.neulet.2014.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/16/2014] [Accepted: 11/10/2014] [Indexed: 02/05/2023]
Abstract
The neural cell adhesion molecule L1 (L1CAM) is crucial for the development of the nervous system, with an essential role in regulating multiple cellular activities. Protein kinase D1 (PKD1) serves as a key kinase given its diverse array of functions within the cell. Here, we investigated various aspects of the functional relationship between L1 and phosphorylated PKD1 (pPKD1) in cerebellar granule neurons. To study the relationship between L1 and PKD1 phosphorylation, human cerebellar tissue microarrays were subject to immunofluorescence staining. We observed a positive correlation between L1 protein levels and PKD1 phosphorylation. In addition, L1 also co-localized with pPKD1. To analyze the regulatory role of L1 on PKD1 phosphorylation, primary mouse cerebellar granule neurons were treated with various concentrations of rL1 for 48 h. Using Western blot, we revealed that L1 significantly increased PKD1 phosphorylation compared with vehicle control, with the maximal effect observed at 5 nM. ERK1/2 phosphorylation was significantly increased by 2.5 nM and 10nM L1, with no apparent change in SRC phosphorylation. However, SRC expression was markedly reduced by 10nM rL1. AKT1 expression and phosphorylation levels were significantly increased by rL1, with the maximal effect observed at 2.5 and 5 nM, respectively. Our combined data revealed a positive relationship between L1 and pPKD1 in both cultured cerebellar neurons and human cerebellar tissue, suggesting that L1 functions in the modulation of PKD1 phosphorylation.
Collapse
Affiliation(s)
- Shuang-xi Chen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Rd, Jinping District, Shantou, Guangdong Province 515041, PR China
| | - Cheng-liang Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Rd, Jinping District, Shantou, Guangdong Province 515041, PR China
| | - Yong-hong Liao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Rd, Jinping District, Shantou, Guangdong Province 515041, PR China
| | - Wei-jiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Rd, Jinping District, Shantou, Guangdong Province 515041, PR China.
| |
Collapse
|
39
|
Zhou X, Xue P, Yang M, Shi H, Lu D, Wang Z, Shi Q, Hu J, Xie S, Zhan W, Yu R. Protein kinase D2 promotes the proliferation of glioma cells by regulating Golgi phosphoprotein 3. Cancer Lett 2014; 355:121-9. [PMID: 25218347 DOI: 10.1016/j.canlet.2014.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 01/16/2023]
Abstract
Protein kinase D2 (PKD2) has been demonstrated to promote tumorigenesis in many types of cancers. However, how PKD2 regulates cancer cell growth is largely unknown. In this study, we found that over-expression of PKD2 promoted glioma cell growth but down-regulation of PKD2 inhibited it. Further investigation indicated that PKD2 down-regulation decreased the protein level of Golgi phosphoprotein 3(GOLPH3) as well as p-AKT level. On the contrary, over-expression of PKD2 increased the protein level of GOLPH3 and p-AKT. In addition, GOLPH3 exhibited similar effect on glioma cell growth to that of PKD2. Importantly, GOLPH3 down-regulation partially abolished glioma cell proliferation induced by PKD2 over-expression, while over-expression of GOLPH3 also partially rescued the inhibition effect of PKD2 down-regulation on glioma cell growth. Interestingly, the level of PKD2 and GOLPH3 significantly increased and was positively correlated in a cohort of glioma patients, as well as in patients from TCGA database. Taken together, these results reveal that PKD2 promotes glioma cell proliferation by regulating GOLPH3 and then AKT activation. Our findings indicate that both PKD2 and GOLPH3 play important roles in the progression of human gliomas and PKD2-GOLPH3-AKT signaling pathway might be a potential glioma therapeutic target.
Collapse
Affiliation(s)
- Xiuping Zhou
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China.
| | - Pengfei Xue
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Minglin Yang
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hengliang Shi
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Dong Lu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zhaohao Wang
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Qiong Shi
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jinxia Hu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Shao Xie
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Wenjian Zhan
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China; Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou, Jiangsu, China.
| |
Collapse
|
40
|
Chen Q, de Lecea L, Hu Z, Gao D. The hypocretin/orexin system: an increasingly important role in neuropsychiatry. Med Res Rev 2014; 35:152-97. [PMID: 25044006 DOI: 10.1002/med.21326] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hypocretins, also named as orexins, are excitatory neuropeptides secreted by neurons specifically located in lateral hypothalamus and perifornical areas. Orexinergic fibers are extensively distributed in various brain regions and involved in a number of physiological functions, such as arousal, cognition, stress, appetite, and metabolism. Arousal is the most important function of orexin system as dysfunction of orexin signaling leads to narcolepsy. In addition to narcolepsy, orexin dysfunction is associated with serious neural disorders, including addiction, depression, and anxiety. However, some results linking orexin with these disorders are still contradictory, which may result from differences of detection methods or the precision of tools used in measurements; strategies targeted to orexin system (e.g., antagonists to orexin receptors, gene delivery, and cell transplantation) are promising new tools for treatment of neuropsychiatric disorders, though studies are still in a stage of preclinical or clinical research.
Collapse
Affiliation(s)
- Quanhui Chen
- Department of Physiology, Third Military Medical University, Chongqing 400038, China; Department of Sleep and Psychology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | |
Collapse
|
41
|
Kim JH, Kim WS, Park C. PKD1 is critical for Epstein-Barr virus LMP1-induced protection of malignant B cells from cell death induced by rituximab. Leuk Lymphoma 2014; 56:194-201. [PMID: 24707946 DOI: 10.3109/10428194.2014.911860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Protein kinase D1 (PKD1 or PKCμ) is a serine/threonine kinase that contributes to malignant progression. Although B and T cells express multiple PKCs, modulation of PKC in association with EBV has not been evaluated. In this study we examined the effects of PKD1 as a cellular target of EBV latent membrane protein-1 (LMP1) on the response of malignant B cells to rituximab and doxorubicin. LMP1 up-regulated PKD1 in malignant B cells but not in T cells. Interestingly, LMP1 stabilized PKD1 protein through direct interaction, which contributed to the survival of malignant B cells. In the absence of PKD1, LMP1 was unable to up-regulate Mcl-1. Also, PH domain and activation loop of PKD1 was critical for LMP1-mediated cell survival. PKD1 knockdown was found to be an efficient strategy to overcome resistance caused by LMP1 expression. Therefore, PKD1 could be a molecular target for therapeutic intervention in EBV-associated B cell lymphoma treatment.
Collapse
Affiliation(s)
- Joo Hyun Kim
- Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul , Korea
| | | | | |
Collapse
|
42
|
Wille C, Seufferlein T, Eiseler T. Protein Kinase D family kinases: roads start to segregate. BIOARCHITECTURE 2014; 4:111-5. [PMID: 24847910 PMCID: PMC4201600 DOI: 10.4161/bioa.29273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Highly invasive pancreatic tumors are often recognized in late stages due to a lack of clear symptoms and pose major challenges for treatment and disease management. Broad-band Protein Kinase D (PKD) inhibitors have recently been proposed as additional treatment option for this disease. PKDs are implicated in the control of cancer cell motility, angiogenesis, proliferation and metastasis. In particular, PKD2 expression is elevated in pancreatic cancer, whereas PKD1 expression is comparably lower. In our recent study we report that both kinases control PDAC cell invasive properties in an isoform-specific, but opposing manner. PKD1 selectively mediates anti-migratory/anti-invasive features by preferential regulation of the actin-regulatory Cofilin-phosphatase Slingshot1L (SSH1L). PKD2, on the other hand enhances invasion and angiogenesis of PDAC cells in 3D-ECM cultures and chorioallantois tumor models by stimulating expression and secretion of matrix-metalloproteinase 7 and 9 (MMP7/9). MMP9 also enhances PKD2-mediated tumor angiogenesis releasing extracellular matrix-bound VEGF-A. We thus suggest high PKD2 expression and loss of PKD1 may be beneficial for tumor cells to enhance their matrix-invading abilities. In our recent study we demonstrate for the first time PKD1 and 2 isoform-selective effects on pancreatic cancer cell invasion, in-vitro and in-vivo, defining isoform-specific regulation of PKDs as a major future issue.
Collapse
Affiliation(s)
- Christoph Wille
- Department of Internal Medicine I; Ulm University; Ulm, Germany
| | | | - Tim Eiseler
- Department of Internal Medicine I; Ulm University; Ulm, Germany
| |
Collapse
|
43
|
Li G, Wang Y. Protein kinase D: a new player among the signaling proteins that regulate functions in the nervous system. Neurosci Bull 2014; 30:497-504. [PMID: 24526660 DOI: 10.1007/s12264-013-1403-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/07/2013] [Indexed: 10/25/2022] Open
Abstract
Protein kinase D (PKD) is an evolutionarily-conserved family of protein kinases. It has structural, regulatory, and enzymatic properties quite different from the PKC family. Many stimuli induce PKD signaling, including G-protein-coupled receptor agonists and growth factors. PKD1 is the most studied member of the family. It functions during cell proliferation, differentiation, secretion, cardiac hypertrophy, immune regulation, angiogenesis, and cancer. Previously, we found that PKD1 is also critically involved in pain modulation. Since then, a series of studies performed in our lab and by other groups have shown that PKDs also participate in other processes in the nervous system including neuronal polarity establishment, neuroprotection, and learning. Here, we discuss the connections between PKD structure, enzyme function, and localization, and summarize the recent findings on the roles of PKD-mediated signaling in the nervous system.
Collapse
Affiliation(s)
- Gang Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | | |
Collapse
|
44
|
Atik N, Kunii M, Avriyanti E, Furumoto N, Inami K, Yoshimura SI, Harada R, Harada A. The role of PKD in cell polarity, biosynthetic pathways, and organelle/F-actin distribution. Cell Struct Funct 2014; 39:61-77. [PMID: 24492625 DOI: 10.1247/csf.13020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Protein Kinase D (PKD) 1, 2, and 3 are members of the PKD family. PKDs influence many cellular processes, including cell polarity, structure of the Golgi, polarized transport from the Golgi to the basolateral plasma membrane, and actin polymerization. However, the role of the PKD family in cell polarity has not yet been elucidated in vivo. Here, we show that KO mice displayed similar localization of the apical and basolateral proteins, transport of VSV-G and a GPI-anchored protein, and similar localization of actin filaments. As DKO mice were embryonic lethal, we generated MEFs that lacked all PKD isoforms from the PKD1 and PKD2 double floxed mice using Cre recombinase and PKD3 siRNA. We observed a similar localization of various organelles, a similar time course in the transport of VSV-G and a GPI-anchored protein, and a similar distribution of F-actin in the PKD-null MEFs. Collectively, our results demonstrate that the complete deletion of PKDs does not affect the transport of VSV-G or a GPI-anchored protein, and the distribution of F-actin. However, simultaneous deletion of PKD1 and PKD2 affect embryonic development, demonstrating their functional redundancy during development.
Collapse
Affiliation(s)
- Nur Atik
- Department of Cell Biology, Graduate School of Medicine, Osaka University
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Olayioye MA, Barisic S, Hausser A. Multi-level control of actin dynamics by protein kinase D. Cell Signal 2013; 25:1739-47. [PMID: 23688773 DOI: 10.1016/j.cellsig.2013.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/24/2013] [Accepted: 04/30/2013] [Indexed: 11/26/2022]
Abstract
Dynamic actin remodeling is fundamental to processes such as cell motility, vesicle trafficking, and cytokinesis. Protein kinase D (PKD) is a serine-threonine kinase known to be involved in diverse biological functions ranging from vesicle fission at the Golgi complex to regulation of cell motility and invasion. This review addresses the role of PKD in the organization of the actin cytoskeleton with a particular emphasis on the substrates associated with this function. We further highlight the multi-level control of actin dynamics by PKD and suggest that the tight spatio-temporal control of PKD activity is critical for the coordination of directed cell migration.
Collapse
Affiliation(s)
- Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | | | | |
Collapse
|
46
|
Xie X, Zhang SS, Wen J, Yang H, Luo KJ, Yang F, Hu Y, Fu JH. Protein kinase D1 mRNA level may predict cancer-specific survival in heavy smokers with esophageal squamous cell cancers. Dis Esophagus 2013; 27:188-95. [PMID: 23621299 DOI: 10.1111/dote.12077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein kinase D1 (PRKD1) is a kinase that regulates various pathways, which involve in cell proliferation, apoptosis, cell adhesion and invasion. Although PRKD1 expression has been observed in many cancers, its role in esophageal squamous cell cancer (ESCC) has not been well reported. As its dysregulation in cancers is organ specific, we sought to investigate the potential role of PRKD1 in the progression of ESCC. Samples were collected from 178 patients with completely resected ESCCs at Sun Yat-sen University Cancer Center, including 47 pairs of tumorous and non-tumorous tissues. PRKD1 mRNA expression was investigated by quantitative real-time polymerase chain reaction. Receiver operating characteristic (ROC) curve analysis was used to search for a feasible cut-off point of PRKD1 mRNA levels for predicting cancer-specific survival. Kaplan-Meier and multivariate Cox regression analysis were used to assess the prognostic value of PRKD1 mRNA level in ESCC patients. In result, upregulation of PRKD1 mRNA was detected in 55.3% (26/47) of ESCC tissues compared with paired non-tumorous ones (P = 0.011). ROC analysis indicated 3.28 as a cut-off point, and thus 72 and 106 tumors with low and high PRKD1 mRNA expression were categorized. High-PRKD1 mRNA expression in tumors appeared with more frequency in heavy smokers (P = 0.002) and patients with advanced pathological T category (P = 0.034). Kaplan-Meier analysis indicated that patients with low-PRKD1 mRNA had a longer cancer-specific survival than the ones with high-PRKD1 level (P = 0.044). Multivariate analysis showed that tumorous PRKD1 mRNA expression was an independent prognostic factor (hazard ratio: 1.538, 95% confidence interval: 1.018-2.323, P = 0.041) in resected ESCC. Subgroup analysis revealed that the discernibility of PRKD1 mRNA level on ESCC outcomes was only pronounced in heavy smokers (P = 0.002), but not in non-heavy smokers (P = 0.870). PRKD1 might play a potential oncogenic role in ESCC. It might be an independent biomarker to predict prognosis in heavy smokers with ESCC.
Collapse
Affiliation(s)
- X Xie
- State Key Laboratory of Oncology in South China, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangdong Esophageal Cancer Research Institute, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Vera-Ramirez L, Pérez-Lopez P, Varela-Lopez A, Ramirez-Tortosa M, Battino M, Quiles JL. Curcumin and liver disease. Biofactors 2013; 39:88-100. [PMID: 23303639 DOI: 10.1002/biof.1057] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 09/13/2012] [Indexed: 12/12/2022]
Abstract
Liver diseases pose a major medical problem worldwide and a wide variety of herbs have been studied for the management of liver-related diseases. In this respect, curcumin has long been used in traditional medicine, and in recent years it has been the object of increasing research interest. In combating liver diseases, it seems clear that curcumin exerts a hypolipidic effect, which prevents the fatty acid accumulation in the hepatocytes that may result from metabolic imbalances, and which may cause nonalcoholic steatohepatitis. Another crucial protective activity of curcumin, not only in the context of chronic liver diseases but also regarding carcinogenesis and other age-related processes, is its potent antioxidant activity, which affects multiple processes and signaling pathways. The effects of curcumin on NF-κβ are crucial to our understanding of the potent hepatoprotective role of this herb-derived micronutrient. Because curcumin is a micronutrient that is closely related to cellular redox balance, its properties and activity give rise to a series of molecular reactions that in every case and biological situation affect the mitochondria.
Collapse
Affiliation(s)
- Laura Vera-Ramirez
- GENyO Center Pfizer-University of Granada & Andalusian Government Centre for Genomics & Oncology, Granada, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Bourzac JF, L'Ériger K, Larrivée JF, Arguin G, Bilodeau MS, Stankova J, Gendron FP. Glucose transporter 2 expression is down regulated following P2X7 activation in enterocytes. J Cell Physiol 2012; 228:120-9. [PMID: 22566162 DOI: 10.1002/jcp.24111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
With the diabetes epidemic affecting the world population, there is an increasing demand for means to regulate glycemia. Dietary glucose is first absorbed by the intestine before entering the blood stream. Thus, the regulation of glucose absorption by intestinal epithelial cells (IECs) could represent a way to regulate glycemia. Among the molecules involved in glycemia homeostasis, extracellular ATP, a paracrine signaling molecule, was reported to induce insulin secretion from pancreatic β cells by activating P2Y and P2X receptors. In rat's jejunum, P2X7 expression was previously immunolocalized to the apex of villi, where it has been suspected to play a role in apoptosis. However, using an antibody recognizing the receptor extracellular domain and thus most of the P2X7 isoforms, we showed that expression of this receptor is apparent in the top two-thirds of villi. These data suggest a different role for this receptor in IECs. Using the non-cancerous IEC-6 cells and differentiated Caco-2 cells, glucose transport was reduced by more than 30% following P2X7 stimulation. This effect on glucose transport was not due to P2X7-induced cell apoptosis, but rather was the consequence of glucose transporter 2 (Glut2)'s internalization. The signaling pathway leading to P2X7-dependent Glut2 internalization involved the calcium-independent activation of phospholipase Cγ1 (PLCγ1), PKCδ, and PKD1. Although the complete mechanism regulating Glut2 internalization following P2X7 activation is not fully understood, modulation of P2X7 receptor activation could represent an interesting approach to regulate intestinal glucose absorption.
Collapse
Affiliation(s)
- Jean-François Bourzac
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Xiang SY, Dusaban SS, Brown JH. Lysophospholipid receptor activation of RhoA and lipid signaling pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:213-22. [PMID: 22986288 DOI: 10.1016/j.bbalip.2012.09.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 09/08/2012] [Accepted: 09/08/2012] [Indexed: 01/08/2023]
Abstract
The lysophospholipids sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) signal through G-protein coupled receptors (GPCRs) which couple to multiple G-proteins and their effectors. These GPCRs are quite efficacious in coupling to the Gα(12/13) family of G-proteins, which stimulate guanine nucleotide exchange factors (GEFs) for RhoA. Activated RhoA subsequently regulates downstream enzymes that transduce signals which affect the actin cytoskeleton, gene expression, cell proliferation and cell survival. Remarkably many of the enzymes regulated downstream of RhoA either use phospholipids as substrates (e.g. phospholipase D, phospholipase C-epsilon, PTEN, PI3 kinase) or are regulated by phospholipid products (e.g. protein kinase D, Akt). Thus lysophospholipids signal from outside of the cell and control phospholipid signaling processes within the cell that they target. Here we review evidence suggesting an integrative role for RhoA in responding to lysophospholipids upregulated in the pathophysiological environment, and in transducing this signal to cellular responses through effects on phospholipid regulatory or phospholipid regulated enzymes. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- Sunny Yang Xiang
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
50
|
Onodera Y, Nam JM, Hashimoto A, Norman JC, Shirato H, Hashimoto S, Sabe H. Rab5c promotes AMAP1-PRKD2 complex formation to enhance β1 integrin recycling in EGF-induced cancer invasion. ACTA ACUST UNITED AC 2012; 197:983-96. [PMID: 22734003 PMCID: PMC3384417 DOI: 10.1083/jcb.201201065] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
EGF signaling activates Rab5c and promotes the intracellular association of AMAP1 and PRKD2 to enhance β1 integrin recycling and promote the invasiveness of breast cancer cells. Epidermal growth factor receptor (EGFR) signaling is one of the crucial factors in breast cancer malignancy. Breast cancer cells often overexpress Arf6 and its effector, AMAP1/ASAP1/DDEF1; in these cells, EGFR signaling may activate the Arf6 pathway to induce invasion and metastasis. Active recycling of some integrins is crucial for invasion and metastasis. Here, we show that the Arf6–AMAP1 pathway links to the machinery that recycles β1 integrins, such as α3β1, to promote cell invasion upon EGFR stimulation. We found that AMAP1 had the ability to bind directly to PRKD2 and hence to make a complex with the cytoplasmic tail of the β1 subunit. Moreover, GTP-Rab5c also bound to AMAP1, and activation of Rab5c by EGFR signaling was necessary to promote the intracellular association of AMAP1 and PRKD2. Our results suggest a novel mechanism by which EGFR signaling promotes the invasiveness of some breast cancer cells via integrin recycling.
Collapse
Affiliation(s)
- Yasuhito Onodera
- Department of Molecular Biology and 2 Department of Radiation Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|