1
|
Jones I, Arias-Garcia M, Pascual-Vargas P, Beykou M, Dent L, Chaudhuri TP, Roumeliotis T, Choudhary J, Sero J, Bakal C. YAP activation is robust to dilution. Mol Omics 2024; 20:554-569. [PMID: 39282972 PMCID: PMC11403994 DOI: 10.1039/d4mo00100a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
The concentration of many transcription factors exhibits high cell-to-cell variability due to differences in synthesis, degradation, and cell size. Whether the functions of these factors are robust to fluctuations in concentration, and how this may be achieved, is poorly understood. Across two independent panels of breast cancer cells, we show that the average whole cell concentration of YAP decreases as a function of cell area. However, the nuclear concentration distribution remains constant across cells grouped by size, across a 4-8 fold size range, implying unperturbed nuclear translocation despite the falling cell wide concentration. Both the whole cell and nuclear concentration was higher in cells with more DNA and CycA/PCNA expression suggesting periodic synthesis of YAP across the cell cycle offsets dilution due to cell growth and/or cell spreading. The cell area - YAP scaling relationship extended to melanoma and RPE cells. Integrative analysis of imaging and phospho-proteomic data showed the average nuclear YAP concentration across cell lines was predicted by differences in RAS/MAPK signalling, focal adhesion maturation, and nuclear transport processes. Validating the idea that RAS/MAPK and cell cycle regulate YAP translocation, chemical inhibition of MEK or CDK4/6 increased the average nuclear YAP concentration. Together, this study provides an example case, where cytoplasmic dilution of a protein, for example through cell growth, does not limit a cognate cellular function. Here, that same proteins translocation into the nucleus.
Collapse
Affiliation(s)
- Ian Jones
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Mar Arias-Garcia
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Patricia Pascual-Vargas
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Melina Beykou
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Lucas Dent
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Tara Pal Chaudhuri
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Theodoros Roumeliotis
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Jyoti Choudhary
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Julia Sero
- Institute for Mathematical Innovation, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Chris Bakal
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
2
|
Gurbi B, Dános K, Birtalan E, Krenács T, Kovács B, Tamás L, Csala M, Varga A. Potential Prognostic Role of Protein Kinase D Isoforms in Head and Neck Cancers. Int J Mol Sci 2024; 25:10274. [PMID: 39408603 PMCID: PMC11477009 DOI: 10.3390/ijms251910274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are among the most common malignancies in men worldwide. Nevertheless, their clinical management is hampered by the limited availability of reliable predictive and prognostic biomarkers. Protein kinase D (PKD) isoforms contribute to major cellular processes. However, their potential role in HNSCC has not been studied systematically, which is the focus of this study. A total of 63 therapy-naive patients with squamous cell carcinoma were consecutively enrolled. Tissue microarray duplicate cores from each case were tested in situ for PKD1, PKD2, and PKD3 expression using immunohistochemistry, and the results were correlated with clinicopathological parameters. We found a high frequency of PKD1/PKD2 positive cases in oropharyngeal and PKD2 positive cases in laryngeal localizations. Only high PKD2 levels were statistically linked to elevated tumor grades, more advanced TNM (3-4) tumor stages, and p16INK4a expression, while elevated PKD3 levels were associated with favorable disease-specific survival. Both PKD2 and PKD3 have been proposed to promote tumor cell proliferation, migration/invasion, and angiogenesis. However, the role of PKD3 was elusive in some cancers. Our findings suggest that testing for PKD isotypes with immunohistochemistry may support the diagnostic estimation of tumor progression and prognosis in HNSCC with a potential therapeutic relevance.
Collapse
Affiliation(s)
- Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (B.G.); (B.K.); (A.V.)
| | - Kornél Dános
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Semmelweis University, 1083 Budapest, Hungary; (K.D.); (E.B.); (L.T.)
| | - Ede Birtalan
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Semmelweis University, 1083 Budapest, Hungary; (K.D.); (E.B.); (L.T.)
| | - Tibor Krenács
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Borbála Kovács
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (B.G.); (B.K.); (A.V.)
| | - László Tamás
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Semmelweis University, 1083 Budapest, Hungary; (K.D.); (E.B.); (L.T.)
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (B.G.); (B.K.); (A.V.)
| | - Attila Varga
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (B.G.); (B.K.); (A.V.)
| |
Collapse
|
3
|
Gutiérrez-Galindo E, Yilmaz ZH, Hausser A. Membrane trafficking in breast cancer progression: protein kinase D comes into play. Front Cell Dev Biol 2023; 11:1173387. [PMID: 37293129 PMCID: PMC10246754 DOI: 10.3389/fcell.2023.1173387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Protein kinase D (PKD) is a serine/threonine kinase family that controls important cellular functions, most notably playing a key role in the secretory pathway at the trans-Golgi network. Aberrant expression of PKD isoforms has been found mainly in breast cancer, where it promotes various cellular processes such as growth, invasion, survival and stem cell maintenance. In this review, we discuss the isoform-specific functions of PKD in breast cancer progression, with a particular focus on how the PKD controlled cellular processes might be linked to deregulated membrane trafficking and secretion. We further highlight the challenges of a therapeutic approach targeting PKD to prevent breast cancer progression.
Collapse
Affiliation(s)
| | - Zeynep Hazal Yilmaz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
4
|
Varga A, Nguyen MT, Pénzes K, Bátai B, Gyulavári P, Gurbi B, Murányi J, Csermely P, Csala M, Vántus T, Sőti C. Protein Kinase D3 (PKD3) Requires Hsp90 for Stability and Promotion of Prostate Cancer Cell Migration. Cells 2023; 12:cells12020212. [PMID: 36672148 PMCID: PMC9857065 DOI: 10.3390/cells12020212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Prostate cancer metastasis is a significant cause of mortality in men. PKD3 facilitates tumor growth and metastasis, however, its regulation is largely unclear. The Hsp90 chaperone stabilizes an array of signaling client proteins, thus is an enabler of the malignant phenotype. Here, using different prostate cancer cell lines, we report that Hsp90 ensures PKD3 conformational stability and function to promote cancer cell migration. We found that pharmacological inhibition of either PKDs or Hsp90 dose-dependently abrogated the migration of DU145 and PC3 metastatic prostate cancer cells. Hsp90 inhibition by ganetespib caused a dose-dependent depletion of PKD2, PKD3, and Akt, which are all involved in metastasis formation. Proximity ligation assay and immunoprecipitation experiments demonstrated a physical interaction between Hsp90 and PKD3. Inhibition of the chaperone-client interaction induced misfolding and proteasomal degradation of PKD3. PKD3 siRNA combined with ganetespib treatment demonstrated a specific involvement of PKD3 in DU145 and PC3 cell migration, which was entirely dependent on Hsp90. Finally, ectopic expression of PKD3 enhanced migration of non-metastatic LNCaP cells in an Hsp90-dependent manner. Altogether, our findings identify PKD3 as an Hsp90 client and uncover a potential mechanism of Hsp90 in prostate cancer metastasis. The molecular interaction revealed here may regulate other biological and pathological functions.
Collapse
Affiliation(s)
- Attila Varga
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (A.V.); (C.S.)
| | - Minh Tu Nguyen
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Kinga Pénzes
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Bence Bátai
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- HCEMM-SU Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Pál Gyulavári
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- IQVIA Hungary, 1117 Budapest, Hungary
| | - Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - József Murányi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Péter Csermely
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Tibor Vántus
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (A.V.); (C.S.)
| |
Collapse
|
5
|
Ramos-Alvarez I, Lee L, Jensen RT. Cofilin activation in pancreatic acinar cells plays a pivotal convergent role for mediating CCK-stimulated enzyme secretion and growth. Front Physiol 2023; 14:1147572. [PMID: 37138671 PMCID: PMC10149936 DOI: 10.3389/fphys.2023.1147572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/05/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction: The actin regulatory protein, cofilin plays a key signaling role in many cells for numerous cellular responses including in proliferation, development, motility, migration, secretion and growth. In the pancreas it is important in islet insulin secretion, growth of pancreatic cancer cells and in pancreatitis. However, there are no studies on its role or activation in pancreatic acinar cells. Methods: To address this question, we studied the ability of CCK to activate cofilin in pancreatic acinar cells, AR42J cells and CCK1-R transfected Panc-1 cells, the signaling cascades involved and its effect on enzyme secretion and MAPK activation, a key mediator of pancreatic growth. Results: CCK (0.3 and 100 nM), TPA, carbachol, Bombesin, secretin and VIP decreased phospho-cofilin (i.e., activate cofilin) and both phospho-kinetic and inhibitor studies of cofilin, LIM kinase (LIMK) and Slingshot Protein Phosphatase (SSH1) demonstrated these conventional activators of cofilin were not involved. Serine phosphatases inhibitors (calyculin A and okadaic acid), however inhibited CCK/TPA-cofilin activation. Studies of various CCK-activated signaling cascades showed activation of PKC/PKD, Src, PAK4, JNK, ROCK mediated cofilin activation, but not PI3K, p38, or MEK. Furthermore, using both siRNA and cofilin inhibitors, cofilin activation was shown to be essential for CCK-mediated enzyme secretion and MAPK activation. Conclusion: These results support the conclusion that cofilin activation plays a pivotal convergent role for various cell signaling cascades in CCK mediated growth/enzyme secretion in pancreatic acini.
Collapse
Affiliation(s)
- Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- National Kyushu Cancer Center, Department of Hepato-Biliary-Pancreatology, Fukuoka, Japan
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Robert T. Jensen,
| |
Collapse
|
6
|
Yuan Y, Zhang H, Li D, Li Y, Lin F, Wang Y, Song H, Liu X, Li F, Zhang J. PAK4 in cancer development: Emerging player and therapeutic opportunities. Cancer Lett 2022; 545:215813. [DOI: 10.1016/j.canlet.2022.215813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
|
7
|
Bamburg JR, Minamide LS, Wiggan O, Tahtamouni LH, Kuhn TB. Cofilin and Actin Dynamics: Multiple Modes of Regulation and Their Impacts in Neuronal Development and Degeneration. Cells 2021; 10:cells10102726. [PMID: 34685706 PMCID: PMC8534876 DOI: 10.3390/cells10102726] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023] Open
Abstract
Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-β in Alzheimer’s disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Correspondence: ; Tel.: +1-970-988-9120; Fax: +1-970-491-0494
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - Lubna H. Tahtamouni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Biology and Biotechnology, The Hashemite University, Zarqa 13115, Jordan
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Chemistry and Biochemistry, University of Alaska, Fairbanks, AK 99775, USA
| |
Collapse
|
8
|
Steinberg SF. Decoding the Cardiac Actions of Protein Kinase D Isoforms. Mol Pharmacol 2021; 100:558-567. [PMID: 34531296 DOI: 10.1124/molpharm.121.000341] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Protein kinase D (PKD) consists of a family of three structurally related enzymes that play key roles in a wide range of biological functions that contribute to the evolution of cardiac hypertrophy and heart failure. PKD1 (the founding member of this enzyme family) has been implicated in the phosphorylation of substrates that regulate cardiac hypertrophy, contraction, and susceptibility to ischemia/reperfusion injury, and de novo PRKD1 (protein kinase D1 gene) mutations have been identified in patients with syndromic congenital heart disease. However, cardiomyocytes coexpress all three PKDs. Although stimulus-specific activation patterns for PKD1, PKD2, and PKD3 have been identified in cardiomyocytes, progress toward identifying PKD isoform-specific functions in the heart have been hampered by significant gaps in our understanding of the molecular mechanisms that regulate PKD activity. This review incorporates recent conceptual breakthroughs in our understanding of various alternative mechanisms for PKD activation, with an emphasis on recent evidence that PKDs activate certain effector responses as dimers, to consider the role of PKD isoforms in signaling pathways that drive cardiac hypertrophy and ischemia/reperfusion injury. The focus is on whether the recently identified activation mechanisms that enhance the signaling repertoire of PKD family enzymes provide novel therapeutic strategies to target PKD enzymes and prevent or slow the evolution of cardiac injury and pathological cardiac remodeling. SIGNIFICANCE STATEMENT: PKD isoforms regulate a large number of fundamental biological processes, but the understanding of the biological actions of individual PKDs (based upon studies using adenoviral overexpression or gene-silencing methods) remains incomplete. This review focuses on dimerization, a recently identified mechanism for PKD activation, and the notion that this mechanism provides a strategy to develop novel PKD-targeted pharmaceuticals that restrict proliferation, invasion, or angiogenesis in cancer and prevent or slow the evolution of cardiac injury and pathological cardiac remodeling.
Collapse
|
9
|
Zhang X, Connelly J, Chao Y, Wang QJ. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021; 11:biom11030483. [PMID: 33807058 PMCID: PMC8005150 DOI: 10.3390/biom11030483] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine protein kinases operating in the signaling network of the second messenger diacylglycerol. The three family members, PKD1, PKD2, and PKD3, are activated by a variety of extracellular stimuli and transduce cell signals affecting many aspects of basic cell functions including secretion, migration, proliferation, survival, angiogenesis, and immune response. Dysregulation of PKD in expression and activity has been detected in many human diseases. Further loss- or gain-of-function studies at cellular levels and in animal models provide strong support for crucial roles of PKD in many pathological conditions, including cancer, metabolic disorders, cardiac diseases, central nervous system disorders, inflammatory diseases, and immune dysregulation. Complexity in enzymatic regulation and function is evident as PKD isoforms may act differently in different biological systems and disease models, and understanding the molecular mechanisms underlying these differences and their biological significance in vivo is essential for the development of safer and more effective PKD-targeted therapies. In this review, to provide a global understanding of PKD function, we present an overview of the PKD family in several major human diseases with more focus on cancer-associated biological processes.
Collapse
|
10
|
Werle SD, Schwab JD, Tatura M, Kirchhoff S, Szekely R, Diels R, Ikonomi N, Sipos B, Sperveslage J, Gress TM, Buchholz M, Kestler HA. Unraveling the Molecular Tumor-Promoting Regulation of Cofilin-1 in Pancreatic Cancer. Cancers (Basel) 2021; 13:725. [PMID: 33578795 PMCID: PMC7916621 DOI: 10.3390/cancers13040725] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/26/2021] [Accepted: 02/07/2021] [Indexed: 12/24/2022] Open
Abstract
Cofilin-1 (CFL1) overexpression in pancreatic cancer correlates with high invasiveness and shorter survival. Besides a well-documented role in actin remodeling, additional cellular functions of CFL1 remain poorly understood. Here, we unraveled molecular tumor-promoting functions of CFL1 in pancreatic cancer. For this purpose, we first show that a knockdown of CFL1 results in reduced growth and proliferation rates in vitro and in vivo, while apoptosis is not induced. By mechanistic modeling we were able to predict the underlying regulation. Model simulations indicate that an imbalance in actin remodeling induces overexpression and activation of CFL1 by acting on transcription factor 7-like 2 (TCF7L2) and aurora kinase A (AURKA). Moreover, we could predict that CFL1 impacts proliferation and apoptosis via the signal transducer and activator of transcription 3 (STAT3). These initial model-based regulations could be substantiated by studying protein levels in pancreatic cancer cell lines and human datasets. Finally, we identified the surface protein CD44 as a promising therapeutic target for pancreatic cancer patients with high CFL1 expression.
Collapse
Affiliation(s)
- Silke D. Werle
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Julian D. Schwab
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Marina Tatura
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Sandra Kirchhoff
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Robin Szekely
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Ramona Diels
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Bence Sipos
- Institute of Pathology, University of Tübingen, 72076 Tübingen, Germany; (B.S.); (J.S.)
| | - Jan Sperveslage
- Institute of Pathology, University of Tübingen, 72076 Tübingen, Germany; (B.S.); (J.S.)
| | - Thomas M. Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Malte Buchholz
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| |
Collapse
|
11
|
Liu Y, Song H, Zhou Y, Ma X, Xu J, Yu Z, Chen L. The oncogenic role of protein kinase D3 in cancer. J Cancer 2021; 12:735-739. [PMID: 33403031 PMCID: PMC7778554 DOI: 10.7150/jca.50899] [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] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/30/2020] [Indexed: 01/12/2023] Open
Abstract
Protein kinase D3 (PRKD3), a serine/threonine kinase, belongs to protein kinase D family, which contains three members: PRKD1, PRKD2, and PRKD3. PRKD3 is activated by many stimuli including phorbol esters, and G-protein-coupled receptor agonists. PRKD3 promotes cancer cell proliferation, growth, migration, and invasion in various tumor types including colorectal, gastric, hepatic, prostate, and breast cancer. Accumulating data supports that PRKD3 is a promising therapeutic target for treatment of cancer. This review discusses the functions and mechanisms of PRKD3 in promoting tumorigenesis and tumor progression of various tumor types as well as the latest developments of small-molecule inhibitors selection for PRKD/PRKD3.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Institute of cancer, Department of biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, P. R.China
| | - Yehui Zhou
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, P. R. China
| | - Xinxing Ma
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, P. R. China
| | - Jing Xu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, P. R.China
| | - Zhenghong Yu
- Department of Rheumatology and Immunology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, P. R.China
| | - Liming Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Institute of cancer, Department of biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, P. R. China
| |
Collapse
|
12
|
Reinhardt R, Truebestein L, Schmidt HA, Leonard TA. It Takes Two to Tango: Activation of Protein Kinase D by Dimerization. Bioessays 2020; 42:e1900222. [PMID: 31997382 DOI: 10.1002/bies.201900222] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/10/2020] [Indexed: 12/23/2022]
Abstract
The recent discovery and structure determination of a novel ubiquitin-like dimerization domain in protein kinase D (PKD) has significant implications for its activation. PKD is a serine/threonine kinase activated by the lipid second messenger diacylglycerol (DAG). It is an essential and highly conserved protein that is implicated in plasma membrane directed trafficking processes from the trans-Golgi network. However, many open questions surround its mechanism of activation, its localization, and its role in the biogenesis of cargo transport carriers. In reviewing this field, the focus is primarily on the mechanisms that control the activation of PKD at precise locations in the cell. In light of the new structural findings, the understanding of the mechanisms underlying PKD activation is critically evaluated, with particular emphasis on the role of dimerization in PKD autophosphorylation, and the provenance and recognition of the DAG that activates PKD.
Collapse
Affiliation(s)
- Ronja Reinhardt
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter, 1030, Vienna, Austria.,Department of Medical Biochemistry, Medical University of Vienna, 1030, Vienna, Austria
| | - Linda Truebestein
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter, 1030, Vienna, Austria.,Department of Medical Biochemistry, Medical University of Vienna, 1030, Vienna, Austria
| | - Heiko A Schmidt
- Center for Integrative Bioinformatics Vienna, Max Perutz Labs, University of Vienna and Medical University of Vienna, Vienna Biocenter, 1030, Vienna, Austria
| | - Thomas A Leonard
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter, 1030, Vienna, Austria.,Department of Medical Biochemistry, Medical University of Vienna, 1030, Vienna, Austria
| |
Collapse
|
13
|
Liu Y, Li J, Ma Z, Zhang J, Wang Y, Yu Z, Lin X, Xu Z, Su Q, An L, Zhou Y, Ma X, Yang Y, Wang F, Chen Q, Zhang Y, Wang J, Zheng H, Shi A, Yu S, Zhang J, Zhao W, Chen L. Oncogenic functions of protein kinase D2 and D3 in regulating multiple cancer-related pathways in breast cancer. Cancer Med 2019; 8:729-741. [PMID: 30652415 PMCID: PMC6504119 DOI: 10.1002/cam4.1938] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/16/2022] Open
Abstract
Protein Kinase D (PKD) family contains PKD1, PKD2, and PKD3 in human. Compared to consistent tumor-suppressive functions of PKD1 in breast cancer, how PKD2/3 functions in breast cancer are not fully understood. In the current study, we found that PKD2 and PKD3 but not PKD1 were preferentially overexpressed in breast cancer and involved in regulating cell proliferation and metastasis. Integrated phosphoproteome, transcriptome, and interactome showed that PKD2 was associated with multiple cancer-related pathways, including adherent junction, regulation of actin cytoskeleton, and cell cycle-related pathways. ELAVL1 was identified as a common hub-node in networks of PKD2/3-regulated phosphoproteins and genes. Silencing ELAVL1 inhibited breast cancer growth in vitro and in vivo. Direct interaction between ELAVL1 and PKD2 or PKD3 was demonstrated. Suppression of PKD2 led to ELAVL1 translocation from the cytoplasm to the nucleus without significant affecting ELAVL1 expression. Taken together, we characterized the oncogenic functions of PKD2/3 in breast cancer and their association with cancer-related pathways, which shed lights on the oncogenic roles and mechanisms of PKDs in breast cancer.
Collapse
Affiliation(s)
- Yan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.,Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, China
| | - Jian Li
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhifang Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Jun Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yuzhi Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhenghong Yu
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xue Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zhi Xu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Su
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Li An
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yehui Zhou
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Xinxing Ma
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yiwen Yang
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Feifei Wang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Qingfei Chen
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yunchao Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Jilinlin Wang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Huilin Zheng
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Aihua Shi
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Shuang Yu
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Weiyong Zhao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liming Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| |
Collapse
|
14
|
Durand N, Borges S, Hall T, Bastea L, Döppler H, Edenfield BH, Thompson EA, Geiger X, Storz P. The phosphorylation status of PIP5K1C at serine 448 can be predictive for invasive ductal carcinoma of the breast. Oncotarget 2018; 9:36358-36370. [PMID: 30555634 PMCID: PMC6284740 DOI: 10.18632/oncotarget.26357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 10/31/2018] [Indexed: 11/25/2022] Open
Abstract
Phosphatidylinositol-4-phosphate 5-kinase type-1C (PIP5K1C) is a lipid kinase that regulates focal adhesion dynamics and cell attachment through site-specific formation of phosphatidylinositol-4,5-bisphosphate (PI4,5P2). By comparing normal breast tissue to carcinoma in situ and invasive ductal carcinoma subtypes, we here show that the phosphorylation status of PIP5K1C at serine residue 448 (S448) can be predictive for breast cancer progression to an aggressive phenotype, while PIP5K1C expression levels are not indicative for this event. PIP5K1C phosphorylation at S448 is downregulated in invasive ductal carcinoma, and similarly, the expression levels of PKD1, the kinase that phosphorylates PIP5K1C at this site, are decreased. Overall, since PKD1 is a negative regulator of cell migration and invasion in breast cancer, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.
Collapse
Affiliation(s)
- Nisha Durand
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tavia Hall
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Brandy H Edenfield
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
15
|
Ramos-Alvarez I, Jensen RT. P21-activated kinase 4 in pancreatic acinar cells is activated by numerous gastrointestinal hormones/neurotransmitters and growth factors by novel signaling, and its activation stimulates secretory/growth cascades. Am J Physiol Gastrointest Liver Physiol 2018; 315:G302-G317. [PMID: 29672153 PMCID: PMC6139648 DOI: 10.1152/ajpgi.00005.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 01/31/2023]
Abstract
p21-activated kinases (PAKs) are highly conserved serine/threonine protein kinases, which are divided into two groups: group-I (PAKs1-3) and group-II (PAKs4-6). In various tissues, Group-II PAKs play important roles in cytoskeletal dynamics and cell growth as well as neoplastic development/progression. However, little is known about Group-II PAK's role in a number of physiological events, including their ability to be activated by gastrointestinal (GI) hormones/neurotransmitters/growth factors (GFs). We used rat pancreatic acini to explore the ability of GI hormones/neurotransmitters/GFs to activate Group-II-PAKs and the signaling cascades involved. Only PAK4 was detected in pancreatic acini. PAK4 was activated by endothelin, secretagogues-stimulating phospholipase C (bombesin, CCK-8, and carbachol), by pancreatic GFs (insulin, insulin-like growth factor 1, hepatocyte growth factor, epidermal growth factor, basic fibroblast growth factor, and platelet-derived growth factor), and by postreceptor stimulants (12-O-tetradecanoylphobol-13-acetate and A23187 ). CCK-8 activation of PAK4 required both high- and low-affinity CCK1-receptor state activation. It was reduced by PKC-, Src-, p44/42-, or p38-inhibition but not with phosphatidylinositol 3-kinase-inhibitors and only minimally by thapsigargin. A protein kinase D (PKD)-inhibitor completely inhibited CCK-8-stimulated PKD-activation; however, stimulated PAK4 phosphorylation was only inhibited by 60%, demonstrating that it is both PKD-dependent and PKD-independent. PF-3758309 and LCH-7749944, inhibitors of PAK4, decreased CCK-8-stimulated PAK4 activation but not PAK2 activation. Each inhibited ERK1/2 activation and amylase release induced by CCK-8 or bombesin. These results show that PAK4 has an important role in modulating signal cascades activated by a number of GI hormones/neurotransmitters/GFs that have been shown to mediate both physiological/pathological responses in acinar cells. Therefore, in addition to the extensive studies on PAK4 in pancreatic cancer, PAK4 should also be considered an important signaling molecule for pancreatic acinar physiological responses and, in the future, should be investigated for a possible role in pancreatic acinar pathophysiological responses, such as in pancreatitis. NEW & NOTEWORTHY This study demonstrates that the only Group-II p21-activated kinase (PAK) in rat pancreatic acinar cells is PAK4, and thus differs from islets/pancreatic cancer. Both gastrointestinal hormones/neurotransmitters stimulating PLC and pancreatic growth factors activate PAK4. With cholecystokinin (CCK), activation is PKC-dependent/-independent, requires both CCK1-R affinity states, Src, p42/44, and p38 activation. PAK4 activation is required for CCK-mediated p42/44 activation/amylase release. These results show PAK4 plays an important role in mediating CCK physiological signal cascades and suggest it may be a target in pancreatic acinar diseases besides cancer.
Collapse
Affiliation(s)
- Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| | - R T Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| |
Collapse
|
16
|
Function and Regulation of Protein Kinase D in Oxidative Stress: A Tale of Isoforms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2138502. [PMID: 29854077 PMCID: PMC5944262 DOI: 10.1155/2018/2138502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/17/2022]
Abstract
Oxidative stress is a condition that arises when cells are faced with levels of reactive oxygen species (ROS) that destabilize the homeostatic redox balance. High levels of ROS can cause damage to macromolecules including DNA, lipids, and proteins, eventually resulting in cell death. Moderate levels of ROS however serve as signaling molecules that can drive and potentiate several cellular phenotypes. Increased levels of ROS are associated with a number of diseases including neurological disorders and cancer. In cancer, increased ROS levels can contribute to cancer cell survival and proliferation via the activation of several signaling pathways. One of the downstream effectors of increased ROS is the protein kinase D (PKD) family of kinases. In this review, we will discuss the regulation and function of this family of ROS-activated kinases and describe their unique isoform-specific features, in terms of both kinase regulation and signaling output.
Collapse
|
17
|
Protein kinase D1: gatekeeper of the epithelial phenotype and key regulator of cancer metastasis? Br J Cancer 2018; 118:459-461. [PMID: 29465085 PMCID: PMC5830601 DOI: 10.1038/bjc.2018.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
18
|
Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, Lohberger B, Wintersperger A, Zucol B, Graier WF, Kratky D, Malle E, Sattler W. Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. J Neuroinflammation 2017; 14:253. [PMID: 29258556 PMCID: PMC5735906 DOI: 10.1186/s12974-017-1024-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Extracellular lysophosphatidic acid (LPA) species transmit signals via six different G protein-coupled receptors (LPAR1-6) and are indispensible for brain development and function of the nervous system. However, under neuroinflammatory conditions or brain damage, LPA levels increase, thereby inducing signaling cascades that counteract brain function. We describe a critical role for 1-oleyl-2-hydroxy-sn-glycero-3-phosphate (termed "LPA" throughout our study) in mediating a motile and pro-inflammatory microglial phenotype via LPAR5 that couples to protein kinase D (PKD)-mediated pathways. METHODS Using the xCELLigence system and time-lapse microscopy, we investigated the migrational response of microglial cells. Different M1 and M2 markers were analyzed by confocal microscopy, flow cytometry, and immunoblotting. Using qPCR and ELISA, we studied the expression of migratory genes and quantitated the secretion of pro-inflammatory cytokines and chemokines, respectively. Different transcription factors that promote the regulation of pro-inflammatory genes were analyzed by western blot. Reactive oxygen species (ROS) and nitric oxide (NO) production, phagocytosis, and microglial cytotoxicity were determined using commercially available assay kits. RESULTS LPA induces MAPK family and AKT activation and pro-inflammatory transcription factors' phosphorylation (NF-κB, c-Jun, STAT1, and STAT3) that were inhibited by both LPAR5 and PKD family antagonists. LPA increases migratory capacity, induces secretion of pro-inflammatory cytokines and chemokines and expression of M1 markers, enhances production of ROS and NO by microglia, and augments cytotoxicity of microglial cell-conditioned medium towards neurons. The PKD family inhibitor blunted all of these effects. We propose that interference with this signaling axis could aid in the development of new therapeutic approaches to control neuroinflammation under conditions of overshooting LPA production. CONCLUSIONS In the present study, we show that inflammatory LPA levels increased the migratory response of microglia and promoted a pro-inflammatory phenotype via the LPAR5/PKD axis. Interference with this signaling axis reduced microglial migration, blunted microglial cytotoxicity, and abrogated the expression and secretion of pro-inflammatory mediators.
Collapse
Affiliation(s)
- I. Plastira
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - E. Bernhart
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - M. Goeritzer
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - T. DeVaney
- 0000 0000 8988 2476grid.11598.34Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - H. Reicher
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - A. Hammer
- 0000 0000 8988 2476grid.11598.34Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - B. Lohberger
- 0000 0000 8988 2476grid.11598.34Department of Orthopedic Surgery, Medical University of Graz, Graz, Austria
| | - A. Wintersperger
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - B. Zucol
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. F. Graier
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - D. Kratky
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - E. Malle
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. Sattler
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| |
Collapse
|
19
|
Durand N, Bastea LI, Döppler H, Eiseler T, Storz P. Src-mediated tyrosine phosphorylation of Protein Kinase D2 at focal adhesions regulates cell adhesion. Sci Rep 2017; 7:9524. [PMID: 28842658 PMCID: PMC5573332 DOI: 10.1038/s41598-017-10210-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/07/2017] [Indexed: 01/09/2023] Open
Abstract
Dependent on their cellular localization, Protein Kinase D (PKD) enzymes regulate different processes including Golgi transport, cell signaling and response to oxidative stress. The localization of PKD within cells is mediated by interaction with different lipid or protein binding partners. With the example of PKD2, we here show that phosphorylation events can also contribute to localization of subcellular pools of this kinase. Specifically, in the present study, we show that tyrosine phosphorylation of PKD2 at residue Y87 defines its localization to the focal adhesions and leads to activation. This phosphorylation occurs downstream of RhoA signaling and is mediated via Src. Moreover, mutation of this residue blocks PKD2's interaction with Focal Adhesion Kinase (FAK). The presence and regulation of PKD2 at focal adhesions identifies a novel function for this kinase as a modulator of cell adhesion and migration.
Collapse
Affiliation(s)
- Nisha Durand
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Ligia I Bastea
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Tim Eiseler
- Department of Internal Medicine I, Ulm University, D-89081, Ulm, Germany
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, 32224, USA.
| |
Collapse
|
20
|
Zhao M, Spiess M, Johansson HJ, Olofsson H, Hu J, Lehtiö J, Strömblad S. Identification of the PAK4 interactome reveals PAK4 phosphorylation of N-WASP and promotion of Arp2/3-dependent actin polymerization. Oncotarget 2017; 8:77061-77074. [PMID: 29100370 PMCID: PMC5652764 DOI: 10.18632/oncotarget.20352] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022] Open
Abstract
p21-activated kinase 4 (PAK4) regulates cell proliferation, apoptosis, cell motility and F-actin remodeling, but the PAK4 interactome has not been systematically analyzed. Here, we comprehensively characterized the human PAK4 interactome by iTRAQ quantitative mass spectrometry of PAK4-immunoprecipitations. Consistent with its multiple reported functions, the PAK4 interactome was enriched in diverse protein networks, including the 14-3-3, proteasome, replication fork, CCT and Arp2/3 complexes. Because PAK4 co-immunoprecipitated most subunits of the Arp2/3 complex, we hypothesized that PAK4 may play a role in Arp2/3 dependent actin regulation. Indeed, we found that PAK4 interacts with and phosphorylates the nucleation promoting factor N-WASP at Ser484/Ser485 and promotes Arp2/3-dependent actin polymerization in vitro. Also, PAK4 ablation in vivo reduced N-WASP Ser484/Ser485 phosphorylation and altered the cellular balance between G- and F-actin as well as the actin organization. By presenting the PAK4 interactome, we here provide a powerful resource for further investigations and as proof of principle, we also indicate a novel mechanism by which PAK4 regulates actin cytoskeleton remodeling.
Collapse
Affiliation(s)
- Miao Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Matthias Spiess
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Henrik J Johansson
- Cancer Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Helene Olofsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Jianjiang Hu
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Janne Lehtiö
- Cancer Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Strömblad
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Shaw AE, Bamburg JR. Peptide regulation of cofilin activity in the CNS: A novel therapeutic approach for treatment of multiple neurological disorders. Pharmacol Ther 2017; 175:17-27. [PMID: 28232023 PMCID: PMC5466456 DOI: 10.1016/j.pharmthera.2017.02.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cofilin is a ubiquitous protein which cooperates with many other actin-binding proteins in regulating actin dynamics. Cofilin has essential functions in nervous system development including neuritogenesis, neurite elongation, growth cone pathfinding, dendritic spine formation, and the regulation of neurotransmission and spine function, components of synaptic plasticity essential for learning and memory. Cofilin's phosphoregulation is a downstream target of many transmembrane signaling processes, and its misregulation in neurons has been linked in rodent models to many different neurodegenerative and neurological disorders including Alzheimer disease (AD), aggression due to neonatal isolation, autism, manic/bipolar disorder, and sleep deprivation. Cognitive and behavioral deficits of these rodent models have been largely abrogated by modulation of cofilin activity using viral-mediated, genetic, and/or small molecule or peptide therapeutic approaches. Neuropathic pain in rats from sciatic nerve compression has also been reduced by modulating the cofilin pathway within neurons of the dorsal root ganglia. Neuroinflammation, which occurs following cerebral ischemia/reperfusion, but which also accompanies many other neurodegenerative syndromes, is markedly reduced by peptides targeting specific chemokine receptors, which also modulate cofilin activity. Thus, peptide therapeutics offer potential for cost-effective treatment of a wide variety of neurological disorders. Here we discuss some recent results from rodent models using therapeutic peptides with a surprising ability to cross the rodent blood brain barrier and alter cofilin activity in brain. We also offer suggestions as to how neuronal-specific cofilin regulation might be achieved.
Collapse
Affiliation(s)
- Alisa E Shaw
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States
| | - James R Bamburg
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States.
| |
Collapse
|
22
|
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: 4.7] [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
|
23
|
Liu Y, Li J, Zhang J, Yu Z, Yu S, Wu L, Wang Y, Gong X, Wu C, Cai X, Mo L, Wang M, Gu J, Chen L. Oncogenic Protein Kinase D3 Regulating Networks in Invasive Breast Cancer. Int J Biol Sci 2017; 13:748-758. [PMID: 28656000 PMCID: PMC5485630 DOI: 10.7150/ijbs.18472] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/24/2017] [Indexed: 11/11/2022] Open
Abstract
Protein Kinase D3 (PRKD3) functions as an important oncogenic driver in invasive breast cancer, which is the leading cause of women mortality. However, PRKD3 regulating network is largely unknown. In this study, we systematically explored PRKD3 regulating networks via investigating phosphoproteome, interactome and transcriptome to uncover the molecular mechanism of PRKD3 in invasive breast cancer. Using iTRAQ, 270 proteins were identified as PRKD3 regulated phosphoproteins from 4619 phosphosites matching 3666 phosphopeptides from 2016 phosphoproteins with p-value <0.005. Transcriptome analysis using affymetrix microarray identified 45 PRKD3 regulated genes, in which 20 genes were upregulated and 25 genes were downregulated with p-value <0.005 upon silencing PRKD3. Using Co-IP in combination of MS identification, 606 proteins were identified to be PRKD3 interacting proteins from 2659 peptides. Further network analysis of PRKD3 regulated phosphoproteins, interacting proteins and regulated genes, reveals 19 hub nodes, including ELAVL1, UBC and BRCA1. UBC was recognized as the most common hub node in PRKD3 regulating networks. The enriched pathway analysis reveals that PRKD3 regulates pathways contributing to multiple cancer related events, including cell cycle, migration and others. Enrichment of cell cycle and cell mobility related pathways across PRKD3 networks, explained the observations that depletion of oncogenic PRKD3 led to alternation of cell cycle and decrease of cell migration ability. Taken together, our current study provided valuable information on the roles as well as the molecular mechanisms of PRKD3 in invasive breast cancer.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Jian Li
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China
| | - Jun Zhang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Zhenghong Yu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, NanJing 210002, P. R. China
| | - Shiyi Yu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Lele Wu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Yuzhi Wang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Xue Gong
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Chenxi Wu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Xiuxiu Cai
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Lin Mo
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, NanJing 210002, P. R. China
| | - Mingya Wang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China
| | - Jun Gu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, NanJing 210002, P. R. China
| | - Liming Chen
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing 210096, PR China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| |
Collapse
|
24
|
Bergeron V, Ghislain J, Poitout V. The P21-activated kinase PAK4 is implicated in fatty-acid potentiation of insulin secretion downstream of free fatty acid receptor 1. Islets 2016; 8:157-164. [PMID: 27700527 PMCID: PMC5161145 DOI: 10.1080/19382014.2016.1243191] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Free fatty acid receptor 1 (FFA1/GPR40) plays a key role in the potentiation of glucose-stimulated insulin secretion by fatty acids in pancreatic β cells. We previously demonstrated that GPR40 signaling leads to cortical actin remodeling and potentiates the second phase of insulin secretion. In this study, we examined the role of p21 activated kinase 4 (PAK4), a known regulator of cytoskeletal dynamics, in GPR40-dependent potentiation of insulin secretion. The fatty acid oleate induced PAK4 phosphorylation in human islets, in isolated mouse islets and in the insulin secreting cell line INS832/13. However, oleate-induced PAK4 phosphorylation was not observed in GPR40-null mouse islets. siRNA-mediated knockdown of PAK4 in INS832/13 cells abrogated the potentiation of insulin secretion by oleate, whereas PAK7 knockdown had no effect. Our results indicate that PAK4 plays an important role in the potentiation of insulin secretion by fatty acids downstream of GPR40.
Collapse
Affiliation(s)
- Valérie Bergeron
- Montreal Diabetes Research Center, CRCHUM, QC, Canada
- Department of Medicine, University of Montreal, QC, Canada
| | | | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, QC, Canada
- Department of Medicine, University of Montreal, QC, Canada
- CONTACT Vincent Poitout CRCHUM, 900 Saint-Denis Street, Montreal, QC, Canada, H2X 0A9
| |
Collapse
|
25
|
Chu E, Saini S, Liu T, Yoo J. Bradykinin stimulates protein kinase D-mediated colonic myofibroblast migration via cyclooxygenase-2 and heat shock protein 27. J Surg Res 2016; 209:191-198. [PMID: 28032559 DOI: 10.1016/j.jss.2016.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inflammatory bowel disease is characterized by episodic intestinal injury and repair. Myofibroblasts are gastrointestinal tract stromal cells that regulate the reparative process and are known targets of inflammatory mediators including bradykinin (BK). However, the mechanisms through which inflammation regulates myofibroblast-induced wound healing remain incompletely understood. Here, we demonstrate, for the first time, that BK stimulates myofibroblast migration through protein kinase D (PKD)-mediated activation of the cyclooxygenase-2 (COX-2) and heat shock protein 27 (Hsp27) pathways. MATERIALS AND METHODS CCD-18Co is a human colonic myofibroblast cell line used from passages 8 to 14. An in vitro scratch assay assessed the effect of BK (100 nM) on myofibroblast migration over 24 h in the presence or absence of several inhibitors (CID755673 [10 μM] and NS398 [10 μM]). Hsp27 small interfering RNA evaluated the effect of Hsp27 on colonic myofibroblast migration. Antibodies to pPKD, pHsp27, and COX-2 evaluated expression levels by Western blot. RESULTS BK stimulated myofibroblast migration over 24 h. BK also led to rapid and sustained phosphorylation of PKD at Ser-916, rapid phosphorylation of Hsp27 at Ser-82, and increased COX-2 expression over 4 h. BK-mediated COX-2 expression and Hsp27 phosphorylation were both inhibited by the PKD inhibitor CID755673. Similarly, BK-induced myofibroblast migration was significantly inhibited by CID755673 (P < 0.05), by the direct COX-2 inhibitor NS398 (P < 0.05), and by Hsp27 small interfering RNA (P < 0.05). CONCLUSIONS BK stimulates myofibroblast migration through PKD-mediated activation of COX-2 and Hsp27. PKD, COX-2, and Hsp27 all appear to regulate myofibroblast cell migration, a stromal population that may play an important role in mucosal healing in the setting of inflammation.
Collapse
Affiliation(s)
- Eric Chu
- Department of Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Shyla Saini
- Department of Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Tiegang Liu
- Department of Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - James Yoo
- Department of Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
26
|
Aicart-Ramos C, He SDQ, Land M, Rubin CS. A Novel Conserved Domain Mediates Dimerization of Protein Kinase D (PKD) Isoforms: DIMERIZATION IS ESSENTIAL FOR PKD-DEPENDENT REGULATION OF SECRETION AND INNATE IMMUNITY. J Biol Chem 2016; 291:23516-23531. [PMID: 27662904 DOI: 10.1074/jbc.m116.735399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Indexed: 01/22/2023] Open
Abstract
Protein kinase D (PKD) isoforms are protein kinase C effectors in signaling pathways regulated by diacylglycerol. Important physiological processes (including secretion, immune responses, motility, and transcription) are placed under diacylglycerol control by the distinctive substrate specificity and subcellular distribution of PKDs. Potentially, broadly co-expressed PKD polypeptides may interact to generate homo- or heteromultimeric regulatory complexes. However, the frequency, molecular basis, regulatory significance, and physiological relevance of stable PKD-PKD interactions are largely unknown. Here, we demonstrate that mammalian PKDs 1-3 and the prototypical Caenorhabditis elegans PKD, DKF-2A, are exclusively (homo- or hetero-) dimers in cell extracts and intact cells. We discovered and characterized a novel, highly conserved N-terminal domain, comprising 92 amino acids, which mediates dimerization of PKD1, PKD2, and PKD3 monomers. A similar domain directs DKF-2A homodimerization. Dimerization occurred independently of properties of the regulatory and kinase domains of PKDs. Disruption of PKD dimerization abrogates secretion of PAUF, a protein carried in small trans-Golgi network-derived vesicles. In addition, disruption of DKF-2A homodimerization in C. elegans intestine impaired and degraded the immune defense of the intact animal against an ingested bacterial pathogen. Finally, dimerization was indispensable for the strong, dominant negative effect of catalytically inactive PKDs. Overall, the structural integrity and function of the novel dimerization domain are essential for PKD-mediated regulation of a key aspect of cell physiology, secretion, and innate immunity in vivo.
Collapse
Affiliation(s)
- Clara Aicart-Ramos
- From the Department of Molecular Pharmacology, Atran Laboratories, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Sophia Dan Qing He
- From the Department of Molecular Pharmacology, Atran Laboratories, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Marianne Land
- From the Department of Molecular Pharmacology, Atran Laboratories, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Charles S Rubin
- From the Department of Molecular Pharmacology, Atran Laboratories, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
27
|
Döppler H, Bastea L, Borges S, Geiger X, Storz P. The phosphorylation status of VASP at serine 322 can be predictive for aggressiveness of invasive ductal carcinoma. Oncotarget 2016; 6:29740-52. [PMID: 26336132 PMCID: PMC4745759 DOI: 10.18632/oncotarget.4965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/31/2015] [Indexed: 11/25/2022] Open
Abstract
Vasodilator-stimulated phosphoprotein (VASP) signaling is critical for dynamic actin reorganization processes that define the motile phenotype of cells. Here we show that VASP is generally highly expressed in normal breast tissue and breast cancer. We also show that the phosphorylation status of VASP at S322 can be predictive for breast cancer progression to an aggressive phenotype. Our data indicate that phosphorylation at S322 is gradually decreased from normal breast to DCIS, luminal/ER+, HER2+ and basal-like/TN phenotypes. Similarly, the expression levels of PKD2, the kinase that phosphorylates VASP at this site, are decreased in invasive ductal carcinoma samples of all three groups. Overall, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.
Collapse
Affiliation(s)
- Heike Döppler
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
28
|
Hussein HAM, Walker LR, Akula SM. KSHV gB associated RGD interactions promote attachment of cells by inhibiting the potential migratory signals induced by the disintegrin-like domain. BMC Cancer 2016; 16:148. [PMID: 26912031 PMCID: PMC4766674 DOI: 10.1186/s12885-016-2173-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 02/14/2016] [Indexed: 12/18/2022] Open
Abstract
Background Kaposi’s sarcoma-associated herpesvirus (KSHV) glycoprotein B (gB) is not only expressed on the envelope of mature virions but also on the surfaces of cells undergoing lytic replication. Among herpesviruses, KSHV gB is the only glycoprotein known to possess the RGD (Arg-Gly-Asp) binding integrin domain critical to mediating cell attachment. Recent studies described gB to also possess a disintegrin-like domain (DLD) said to interact with non-RGD binding integrins. We wanted to decipher the roles of two individually distinct integrin binding domains (RGD versus DLD) within KSHV gB in regulating attachment of cells over cell migration. Methods We established HeLa cells expressing recombinant full length gB, gB lacking a functional RGD (gBΔR), and gB lacking a functionally intact DLD (gBΔD) on their cell surfaces. These cells were tested in wound healing assay, Transwell migration assay, and adhesion assay to monitor the ability of the RGD and DLD integrin recognition motifs in gB to mediate migration and attachment of cells. We also used soluble forms of the respective gB recombinant proteins to analyze and confirm their effect on migration and attachment of cells. The results from the above studies were authenticated by the use of imaging, and standard biochemical approaches as Western blotting and RNA silencing using small interfering RNA. Results The present report provides the following novel findings: (i) gB does not induce cell migration; (ii) RGD domain in KSHV gB is the switch that inhibits the ability of DLD to induce cellular migration thus promoting attachment of cells. Conclusions Independently, RGD interactions mediate attachment of cells while DLD interactions regulate migration of cells. However, when both RGD and DLD are functionally present in the same protein, gB, the RGD interaction-induced attachment of cells overshadows the ability of DLD mediated signaling to induce migration of cells. Furthering our understanding of the molecular mechanism of integrin engagement with RGD and DLD motifs within gB could identify promising new therapeutic avenues and research areas to explore. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2173-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hosni A M Hussein
- Department of Microbiology & Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA
| | - Lia R Walker
- Department of Microbiology & Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
29
|
Durand N, Borges S, Storz P. Protein Kinase D Enzymes as Regulators of EMT and Cancer Cell Invasion. J Clin Med 2016; 5:jcm5020020. [PMID: 26848698 PMCID: PMC4773776 DOI: 10.3390/jcm5020020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/15/2015] [Accepted: 01/18/2016] [Indexed: 12/20/2022] Open
Abstract
The Protein Kinase D (PKD) isoforms PKD1, PKD2, and PKD3 are effectors of the novel Protein Kinase Cs (nPKCs) and diacylglycerol (DAG). PKDs impact diverse biological processes like protein transport, cell migration, proliferation, epithelial to mesenchymal transition (EMT) and apoptosis. PKDs however, have distinct effects on these functions. While PKD1 blocks EMT and cell migration, PKD2 and PKD3 tend to drive both processes. Given the importance of EMT and cell migration to the initiation and progression of various malignancies, abnormal expression of PKDs has been reported in multiple types of cancers, including breast, pancreatic and prostate cancer. In this review, we discuss how EMT and cell migration are regulated by PKD isoforms and the significance of this regulation in the context of cancer development.
Collapse
Affiliation(s)
- Nisha Durand
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
30
|
Durand N, Borges S, Storz P. Functional and therapeutic significance of protein kinase D enzymes in invasive breast cancer. Cell Mol Life Sci 2015; 72:4369-82. [PMID: 26253275 DOI: 10.1007/s00018-015-2011-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 12/31/2022]
Abstract
The protein kinase D (PKD) family members, PKD1, PKD2 and PKD3 constitute a family of serine/threonine kinases that are essential regulators of cell migration, proliferation and protein transport. Multiple types of cancers are characterized by aberrant expression of PKD isoforms. In breast cancer PKD isoforms exhibit distinct expression patterns and regulate various oncogenic processes. In highly invasive breast cancer, the leading cause of cancer-associated deaths in females, the loss of PKD1 is thought to promote invasion and metastasis, while PKD2 and upregulated PKD3 have been shown to be positive regulators of proliferation, chemoresistance and metastasis. In this review, we examine the differential expression pattern, mechanisms of regulation and contributions made by each PKD isoform to the development and maintenance of invasive breast cancer. In addition, we discuss the potential therapeutic approaches for targeting PKD in this disease.
Collapse
Affiliation(s)
- Nisha Durand
- Department of Cancer Biology, Mayo Clinic, Griffin Room 306, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Sahra Borges
- Department of Cancer Biology, Mayo Clinic, Griffin Room 306, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Griffin Room 306, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
31
|
Borges S, Perez EA, Thompson EA, Radisky DC, Geiger XJ, Storz P. Effective Targeting of Estrogen Receptor-Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101. Mol Cancer Ther 2015; 14:1306-16. [PMID: 25852060 DOI: 10.1158/1535-7163.mct-14-0945] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/02/2015] [Indexed: 12/11/2022]
Abstract
Invasive ductal carcinomas (IDC) of the breast are associated with altered expression of hormone receptors (HR), amplification or overexpression of HER2, or a triple-negative phenotype. The most aggressive cases of IDC are characterized by a high proliferation rate, a great propensity to metastasize, and their ability to resist to standard chemotherapy, hormone therapy, or HER2-targeted therapy. Using progression tissue microarrays, we here demonstrate that the serine/threonine kinase protein kinase D3 (PKD3) is highly upregulated in estrogen receptor (ER)-negative (ER(-)) tumors. We identify direct binding of the ER to the PRKD3 gene promoter as a mechanism of inhibition of PKD3 expression. Loss of ER results in upregulation of PKD3, leading to all hallmarks of aggressive IDC, including increased cell proliferation, migration, and invasion. This identifies ER(-) breast cancers as ideal for treatment with the PKD inhibitor CRT0066101. We show that similar to a knockdown of PKD3, treatment with this inhibitor targets all tumorigenic processes in vitro and decreases growth of primary tumors and metastasis in vivo. Our data strongly support the development of PKD inhibitors for clinical use for ER(-) breast cancers, including the triple-negative phenotype.
Collapse
Affiliation(s)
- Sahra Borges
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Edith A Perez
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida. Department of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
32
|
Alpsoy A, Gündüz U. Protein kinase D2 silencing reduced motility of doxorubicin-resistant MCF7 cells. Tumour Biol 2015; 36:4417-26. [PMID: 25874490 DOI: 10.1007/s13277-015-3081-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/08/2015] [Indexed: 11/26/2022] Open
Abstract
Success of chemotherapy is generally impaired by multidrug resistance, intrinsic resistance, or acquired resistance to functionally and structurally irrelevant drugs. Multidrug resistance emerges via distinct mechanisms: increased drug export, decreased drug internalization, dysfunctional apoptotic machinery, increased DNA damage repair, altered cell cycle regulation, and increased drug detoxification. Several reports demonstrated that multidrug resistance is a multifaceted problem such that multidrug resistance correlates with increased aggressiveness and metastatic potential. Here, we tested the involvement of protein kinase D2, a serine/threonine kinase that was previously implicated in proliferation, drug resistance, and motility in doxorubicin-resistant MCF7 (MCF7/DOX) cell line, which served as an in vitro model for drug resistance and invasiveness. We showed that basal level activity of protein kinase D2 (PKD2) was higher in MCF7/DOX cells than parental MCF7 cells. To elucidate the roles of PKD2 MCF7/DOX, PKD2 expression was reduced via small interfering RNA (siRNA)-mediated knockdown. Results showed that acquired resistance of MCF7/DOX to doxorubicin was not affected by PKD2 silencing, while motility of MCF7/DOX cells was reduced. The results implied that PKD2 silencing might inhibit migration of MCF7/DOX cells without affecting chemoresistance significantly.
Collapse
Affiliation(s)
- Aktan Alpsoy
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | | |
Collapse
|