1
|
Kazanietz MG, Cooke M. Protein kinase C signaling "in" and "to" the nucleus: Master kinases in transcriptional regulation. J Biol Chem 2024; 300:105692. [PMID: 38301892 PMCID: PMC10907189 DOI: 10.1016/j.jbc.2024.105692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
PKC is a multifunctional family of Ser-Thr kinases widely implicated in the regulation of fundamental cellular functions, including proliferation, polarity, motility, and differentiation. Notwithstanding their primary cytoplasmic localization and stringent activation by cell surface receptors, PKC isozymes impel prominent nuclear signaling ultimately impacting gene expression. While transcriptional regulation may be wielded by nuclear PKCs, it most often relies on cytoplasmic phosphorylation events that result in nuclear shuttling of PKC downstream effectors, including transcription factors. As expected from the unique coupling of PKC isozymes to signaling effector pathways, glaring disparities in gene activation/repression are observed upon targeting individual PKC family members. Notably, specific PKCs control the expression and activation of transcription factors implicated in cell cycle/mitogenesis, epithelial-to-mesenchymal transition and immune function. Additionally, PKCs isozymes tightly regulate transcription factors involved in stepwise differentiation of pluripotent stem cells toward specific epithelial, mesenchymal, and hematopoietic cell lineages. Aberrant PKC expression and/or activation in pathological conditions, such as in cancer, leads to profound alterations in gene expression, leading to an extensive rewiring of transcriptional networks associated with mitogenesis, invasiveness, stemness, and tumor microenvironment dysregulation. In this review, we outline the current understanding of PKC signaling "in" and "to" the nucleus, with significant focus on established paradigms of PKC-mediated transcriptional control. Dissecting these complexities would allow the identification of relevant molecular targets implicated in a wide spectrum of diseases.
Collapse
Affiliation(s)
- Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
2
|
Tao H, Song SJ, Fan ZW, Li WT, Jin X, Jiang W, Bai J, Shi ZZ. PKCiota Inhibits the Ferroptosis of Esophageal Cancer Cells via Suppressing USP14-Mediated Autophagic Degradation of GPX4. Antioxidants (Basel) 2024; 13:114. [PMID: 38247539 PMCID: PMC10812620 DOI: 10.3390/antiox13010114] [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: 10/25/2023] [Revised: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most frequent malignant tumors, and the mechanisms underlying the anti-ferroptosis of esophageal cancer cells are still largely unclear. This study aims to explore the roles of amplified protein kinase C iota (PKCiota) in the ferroptosis of ESCC cells. Cell viability, colony formation, MDA assay, Western blotting, co-IP, PLA, and RNA-seq technologies are used to reveal the roles and mechanisms underlying the PKCiota-induced resistance of ESCC cells to ferroptosis. We showed here that PKCiota was amplified and overexpressed in ESCC and decreased during RSL3-induced ferroptosis of ESCC cells. PKCiota interacted with GPX4 and the deubiquitinase USP14 and improved the protein stability of GPX4 by suppressing the USP14-mediated autophagy-lysosomal degradation pathway. PKCiota was negatively regulated by miR-145-5p, which decreased in esophageal cancer, and also regulated by USP14 and GPX4 by a positive feedback loop. PKCiota silencing and miR-145-5p overexpression suppressed tumor growth of ESCC cells in vivo, respectively; even a combination of silencing PKCiota and RSL3 treatment showed more vital suppressive roles on tumor growth than silencing PKCiota alone. Both PKCiota silencing and miR-145-5p overexpression sensitized ESCC cells to RSL3-induced ferroptosis. These results unveiled that amplified and overexpressed PKCiota induced the resistance of ESCC cells to ferroptosis by suppressing the USP14-mediated autophagic degradation of GPX4. Patients with PKCiota/USP14/GPX4 pathway activation might be sensitive to GPX4-targeted ferroptosis-based therapy.
Collapse
Affiliation(s)
- Hao Tao
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Sheng-Jie Song
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Ze-Wen Fan
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Wen-Ting Li
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Xin Jin
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Wen Jiang
- Department of Thoracic Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650000, China;
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| | - Zhi-Zhou Shi
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; (H.T.); (S.-J.S.); (Z.-W.F.); (W.-T.L.); (X.J.); (J.B.)
| |
Collapse
|
3
|
Yin N, Liu Y, Weems C, Shreeder B, Lou Y, Knutson KL, Murray NR, Fields AP. Protein kinase Cι mediates immunosuppression in lung adenocarcinoma. Sci Transl Med 2022; 14:eabq5931. [PMID: 36383684 PMCID: PMC11457891 DOI: 10.1126/scitranslmed.abq5931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent form of non-small cell lung cancer (NSCLC) and a leading cause of cancer death. Immune checkpoint inhibitors (ICIs) of programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) signaling induce tumor regressions in a subset of LUAD, but many LUAD tumors exhibit resistance to ICI therapy. Here, we identified Prkci as a major determinant of response to ICI in a syngeneic mouse model of oncogenic mutant Kras/Trp53 loss (KP)-driven LUAD. Protein kinase Cι (PKCι)-dependent KP tumors exhibited resistance to anti-PD-1 antibody therapy (α-PD-1), whereas KP tumors in which Prkci was genetically deleted (KPI tumors) were highly responsive. Prkci-dependent resistance to α-PD-1 was characterized by enhanced infiltration of myeloid-derived suppressor cells (MDSCs) and decreased infiltration of CD8+ T cells in response to α-PD-1. Mechanistically, Prkci regulated YAP1-dependent expression of Cxcl5, which served to attract MDSCs to KP tumors. The PKCι inhibitor auranofin inhibited KP tumor growth and sensitized these tumors to α-PD-1, whereas expression of either Prkci or its downstream effector Cxcl5 in KPI tumors induced intratumoral infiltration of MDSCs and resistance to α-PD-1. PRKCI expression in tumors of patients with LUAD correlated with genomic signatures indicative of high YAP1-mediated transcription, elevated MDSC infiltration and low CD8+ T cell infiltration, and with elevated CXCL5/6 expression. Last, PKCι-YAP1 signaling was a biomarker associated with poor response to ICI in patients with LUAD. Our data indicate that immunosuppressive PKCι-YAP1-CXCL5 signaling is a key determinant of response to ICI, and pharmacologic inhibition of PKCι may improve therapeutic response to ICI in patients with LUAD.
Collapse
Affiliation(s)
- Ning Yin
- Department of Cancer Biology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Yi Liu
- Department of Cancer Biology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Capella Weems
- Department of Cancer Biology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Barath Shreeder
- Department of Immunology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Keith L. Knutson
- Department of Immunology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Nicole R. Murray
- Department of Cancer Biology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| | - Alan P. Fields
- Department of Cancer Biology, Mayo Clinic School of Medicine, Jacksonville, FL 32224, USA
| |
Collapse
|
4
|
Inman KS, Liu Y, Scotti Buzhardt ML, Leitges M, Krishna M, Crawford HC, Fields AP, Murray NR. Prkci Regulates Autophagy and Pancreatic Tumorigenesis in Mice. Cancers (Basel) 2022; 14:796. [PMID: 35159064 PMCID: PMC8834021 DOI: 10.3390/cancers14030796] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Protein kinase C iota (PKCι) functions as a bonafide human oncogene in lung and ovarian cancer and is required for KrasG12D-mediated lung cancer initiation and progression. PKCι expression is required for pancreatic cancer cell growth and maintenance of the transformed phenotype; however, nothing is known about the role of PKCι in pancreas development or pancreatic tumorigenesis. In this study, we investigated the effect of pancreas-specific ablation of PKCι expression on pancreatic cellular homeostasis, susceptibility to pancreatitis, and KrasG12D-mediated pancreatic cancer development. Knockout of pancreatic Prkci significantly increased pancreatic immune cell infiltration, acinar cell DNA damage, and apoptosis, but reduced sensitivity to caerulein-induced pancreatitis. Prkci-ablated pancreatic acinar cells exhibited P62 aggregation and a loss of autophagic vesicles. Loss of pancreatic Prkci promoted KrasG12D-mediated pancreatic intraepithelial neoplasia formation but blocked progression to adenocarcinoma, consistent with disruption of autophagy. Our results reveal a novel promotive role for PKCι in pancreatic epithelial cell autophagy and pancreatic cancer progression.
Collapse
Affiliation(s)
- Kristin S. Inman
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Environmental Health Perspectives/National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yi Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| | - Michele L. Scotti Buzhardt
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Neogenomics Laboratories, Clinical Division, Charlotte, NC 28104, USA
| | - Michael Leitges
- Department of BioMedical Sciences, Faculty of Medicine, Memorial University, St. John’s, NL A1M 2V7, Canada;
| | - Murli Krishna
- Department of Pathology/Lab Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Howard C. Crawford
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Detroit, MI 48202, USA
| | - Alan P. Fields
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| | - Nicole R. Murray
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| |
Collapse
|
5
|
Kenchappa RS, Liu Y, Argenziano MG, Banu MA, Mladek AC, West R, Luu A, Quiñones-Hinojosa A, Hambardzumyan D, Justilien V, Leitges M, Sarkaria JN, Sims PA, Canoll P, Murray NR, Fields AP, Rosenfeld SS. Protein kinase C ι and SRC signaling define reciprocally related subgroups of glioblastoma with distinct therapeutic vulnerabilities. Cell Rep 2021; 37:110054. [PMID: 34818553 PMCID: PMC9845019 DOI: 10.1016/j.celrep.2021.110054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/17/2021] [Accepted: 11/03/2021] [Indexed: 01/19/2023] Open
Abstract
We report that atypical protein kinase Cι (PKCι) is an oncogenic driver of glioblastoma (GBM). Deletion or inhibition of PKCι significantly impairs tumor growth and prolongs survival in murine GBM models. GBM cells expressing elevated PKCι signaling are sensitive to PKCι inhibitors, whereas those expressing low PKCι signaling exhibit active SRC signaling and sensitivity to SRC inhibitors. Resistance to the PKCι inhibitor auranofin is associated with activated SRC signaling and response to a SRC inhibitor, whereas resistance to a SRC inhibitor is associated with activated PKCι signaling and sensitivity to auranofin. Interestingly, PKCι- and SRC-dependent cells often co-exist in individual GBM tumors, and treatment of GBM-bearing mice with combined auranofin and SRC inhibitor prolongs survival beyond either drug alone. Thus, we identify PKCι and SRC signaling as distinct therapeutic vulnerabilities that are directly translatable into an improved treatment for GBM.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Michael G Argenziano
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Matei A Banu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA
| | - Rita West
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Amanda Luu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Dolores Hambardzumyan
- Departments of Neurosurgery and Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Verline Justilien
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Nicole R Murray
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Alan P Fields
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | |
Collapse
|