1
|
Whitford MKM, McCaffrey L. Polarity in breast development and cancer. Curr Top Dev Biol 2023; 154:245-283. [PMID: 37100520 DOI: 10.1016/bs.ctdb.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Mammary gland development and breast cancer progression are associated with extensive remodeling of epithelial tissue architecture. Apical-basal polarity is a key feature of epithelial cells that coordinates key elements of epithelial morphogenesis including cell organization, proliferation, survival, and migration. In this review we discuss advances in our understanding of how apical-basal polarity programs are used in breast development and cancer. We describe cell lines, organoids, and in vivo models commonly used for studying apical-basal polarity in breast development and disease and discuss advantages and limitations of each. We also provide examples of how core polarity proteins regulate branching morphogenesis and lactation during development. We describe alterations to core polarity genes in breast cancer and their associations with patient outcomes. The impact of up- or down-regulation of key polarity proteins in breast cancer initiation, growth, invasion, metastasis, and therapeutic resistance are discussed. We also introduce studies demonstrating that polarity programs are involved in regulating the stroma, either through epithelial-stroma crosstalk, or through signaling of polarity proteins in non-epithelial cell types. Overall, a key concept is that the function of individual polarity proteins is highly contextual, depending on developmental or cancer stage and cancer subtype.
Collapse
Affiliation(s)
- Mara K M Whitford
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Luke McCaffrey
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Velnati S, Centonze S, Girivetto F, Capello D, Biondi RM, Bertoni A, Cantello R, Ragnoli B, Malerba M, Graziani A, Baldanzi G. Identification of Key Phospholipids That Bind and Activate Atypical PKCs. Biomedicines 2021; 9:biomedicines9010045. [PMID: 33419210 PMCID: PMC7825596 DOI: 10.3390/biomedicines9010045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/30/2020] [Accepted: 01/01/2021] [Indexed: 12/02/2022] Open
Abstract
PKCζ and PKCι/λ form the atypical protein kinase C subgroup, characterised by a lack of regulation by calcium and the neutral lipid diacylglycerol. To better understand the regulation of these kinases, we systematically explored their interactions with various purified phospholipids using the lipid overlay assays, followed by kinase activity assays to evaluate the lipid effects on their enzymatic activity. We observed that both PKCζ and PKCι interact with phosphatidic acid and phosphatidylserine. Conversely, PKCι is unique in binding also to phosphatidylinositol-monophosphates (e.g., phosphatidylinositol 3-phosphate, 4-phosphate, and 5-phosphate). Moreover, we observed that phosphatidylinositol 4-phosphate specifically activates PKCι, while both isoforms are responsive to phosphatidic acid and phosphatidylserine. Overall, our results suggest that atypical Protein kinase C (PKC) localisation and activity are regulated by membrane lipids distinct from those involved in conventional PKCs and unveil a specific regulation of PKCι by phosphatidylinositol-monophosphates.
Collapse
Affiliation(s)
- Suresh Velnati
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
- Correspondence:
| | - Sara Centonze
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Federico Girivetto
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Capello
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- UPO Biobank, University of Piemonte Orientale, 28100 Novara, Italy
| | - Ricardo M. Biondi
- Department of Internal Medicine 1, Goethe University Hospital Frankfurt, 60590 Frankfurt, Germany;
- Biomedicine Research Institute of Buenos Aires—CONICET—Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Alessandra Bertoni
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
| | - Roberto Cantello
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
| | | | - Mario Malerba
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- Respiratory Unit, Sant’Andrea Hospital, 13100 Vercelli, Italy;
| | - Andrea Graziani
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy;
- Division of Oncology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (F.G.); (D.C.); (A.B.); (R.C.); (M.M.); (G.B.)
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
3
|
High PKCλ expression is required for ALDH1-positive cancer stem cell function and indicates a poor clinical outcome in late-stage breast cancer patients. PLoS One 2020; 15:e0235747. [PMID: 32658903 PMCID: PMC7357771 DOI: 10.1371/journal.pone.0235747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/22/2020] [Indexed: 12/19/2022] Open
Abstract
Despite development of markers for identification of cancer stem cells, the mechanism underlying the survival and division of cancer stem cells in breast cancer remains unclear. Here we report that PKCλ expression was enriched in basal-like breast cancer, among breast cancer subtypes, and was correlated with ALDH1A3 expression (p = 0.016, χ2-test). Late stage breast cancer patients expressing PKCλhigh and ALDH1A3high had poorer disease-specific survival than those expressing PKCλlow and ALDH1A3low (p = 0.018, log rank test for Kaplan-Meier survival curves: hazard ratio 2.58, 95% CI 1.24–5.37, p = 0.011, multivariate Cox regression analysis). Functional inhibition of PKCλ through siRNA-mediated knockdown or CRISPR-Cas9-mediated knockout in ALDH1high MDA-MB 157 and MDA-MB 468 basal-like breast cancer cells led to increases in the numbers of trypan blue-positive and active-caspase 3-positive cells, as well as suppression of tumor-sphere formation and cell migration. Furthermore, the amount of CASP3 and PARP mRNA and the level of cleaved caspase-3 protein were enhanced in PKCλ-deficient ALDH1high cells. An Apoptosis inhibitor (z-VAD-FMK) suppressed the enhancement of cell death as well as the levels of cleaved caspase-3 protein in PKCλ deficient ALDH1high cells. It also altered the asymmetric/symmetric distribution ratio of ALDH1A3 protein. In addition, PKCλ knockdown led to increases in cellular ROS levels in ALDH1high cells. These results suggest that PKCλ is essential for cancer cell survival and migration, tumorigenesis, the asymmetric distribution of ALDH1A3 protein among cancer cells, and the maintenance of low ROS levels in ALDH1-positive breast cancer stem cells. This makes it a key contributor to the poorer prognosis seen in late-stage breast cancer patients.
Collapse
|
4
|
A high-throughput screen identifies that CDK7 activates glucose consumption in lung cancer cells. Nat Commun 2019; 10:5444. [PMID: 31784510 PMCID: PMC6884612 DOI: 10.1038/s41467-019-13334-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Elevated glucose consumption is fundamental to cancer, but selectively targeting this pathway is challenging. We develop a high-throughput assay for measuring glucose consumption and use it to screen non-small-cell lung cancer cell lines against bioactive small molecules. We identify Milciclib that blocks glucose consumption in H460 and H1975, but not in HCC827 or A549 cells, by decreasing SLC2A1 (GLUT1) mRNA and protein levels and by inhibiting glucose transport. Milciclib blocks glucose consumption by targeting cyclin-dependent kinase 7 (CDK7) similar to other CDK7 inhibitors including THZ1 and LDC4297. Enhanced PIK3CA signaling leads to CDK7 phosphorylation, which promotes RNA Polymerase II phosphorylation and transcription. Milciclib, THZ1, and LDC4297 lead to a reduction in RNA Polymerase II phosphorylation on the SLC2A1 promoter. These data indicate that our high-throughput assay can identify compounds that regulate glucose consumption and that CDK7 is a key regulator of glucose consumption in cells with an activated PI3K pathway. Many cancer cells have increased glucose consumption compared to normal cells, a feature that can be exploited therapeutically. Here, the authors carry out a chemical screen and identify compounds that selectively blocks glucose metabolism in non-small-cell lung cancer cell lines.
Collapse
|
5
|
Aberrant Nuclear Localization of aPKCλ/ι is Associated With Poorer Prognosis in Uterine Cervical Cancer. Int J Gynecol Pathol 2019; 38:301-309. [PMID: 30059452 DOI: 10.1097/pgp.0000000000000539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We previously reported that aberrant expression of atypical protein kinase C λ/ι (aPKCλ/ι) in low-grade squamous intraepithelial uterine cervix lesions was associated with an increased risk of progression to higher grade. This study aimed to investigate aPKCλ/ι expression patterns in cervical squamous cell carcinoma (SCC) and its association with disease progression. We immunohistochemically assessed aPKCλ/ι expression in 168 SCC samples and 13 normal uterine cervix samples. In 69.0% of SCC cases, aPKCλ/ι was expressed more abundantly than in normal epithelium, but there was no significant association between aPKCλ/ι intensity and disease progression (P=0.087, Cochran-Mantel-Haenszel test). aPKCλ/ι in normal cervical epithelium was confined to the cytoplasm or intercellular junctions. In contrast, aPKCλ/ι was predominantly localized within the nucleus in 36.9% of SCC samples (P<0.001, χ test), and the prevalence was significantly increased relative to advanced tumor stage (P<0.001, Cochran-Mantel-Haenszel test). Moreover, patients with SCC with aPKCλ/ι nuclear localization had worse prognoses than those with cytoplasmic localization (P<0.001, log-rank test). aPKCλ/ι localization differed between the intraepithelial lesion and adjacent invasive cancer in 40% of cases, while the expression pattern was similar between primary and matched metastatic tumors. In conclusion, aPKCλ/ι nuclear localization in cervical cancer is associated with tumor progression and worse prognosis. This is the first report to show aberrant nuclear aPKCλ/ι localization in a subgroup of cervical cancer patients and its association with worse prognosis.
Collapse
|
6
|
Protein kinase Cε regulates nuclear translocation of extracellular signal-regulated kinase, which contributes to bradykinin-induced cyclooxygenase-2 expression. Sci Rep 2018; 8:8535. [PMID: 29867151 PMCID: PMC5986758 DOI: 10.1038/s41598-018-26473-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/04/2018] [Indexed: 01/18/2023] Open
Abstract
The proinflammatory mediator bradykinin stimulated cyclooxygenase-2 (COX-2) expression and subsequently prostaglandin E2 synthesis in dermal fibroblasts. The involvement of B2 receptors and Gαq in the role of bradykinin was suggested by using pharmacological inhibitors. The PKC activator PMA stimulated COX-2 mRNA expression. Bradykinin failed to induce COX-2 mRNA expression in the presence of PKC inhibitors, whereas the effect of bradykinin was observed in the absence of extracellular Ca2+. Bradykinin-induced COX-2 mRNA expression was inhibited in cells transfected with PKCε siRNA. These observations suggest that the novel PKCε is concerned with bradykinin-induced COX-2 expression. Bradykinin-induced PKCε phosphorylation and COX-2 mRNA expression were inhibited by an inhibitor of 3-phosphoinositide-dependent protein kinase-1 (PDK-1), and bradykinin-induced PDK-1 phosphorylation was inhibited by phospholipase D (PLD) inhibitors, suggesting that PLD/PDK-1 pathway contributes to bradykinin-induced PKCε activation. Pharmacological and knockdown studies suggest that the extracellular signal-regulated kinase 1 (ERK1) MAPK signaling is involved in bradykinin-induced COX-2 expression. Bradykinin-induced ERK phosphorylation was attenuated in the cells pretreated with PKC inhibitors or transfected with PKCε siRNA. We observed the interaction between PKCε and ERK by co-immunoprecipitation experiments. These observations suggest that PKCε activation contributes to the regulation of ERK1 activation. Bradykinin stimulated the accumulation of phosphorylated ERK in the nuclear fraction, that was inhibited in the cells treated with PKC inhibitors or transfected with PKCε siRNA. Consequently, we concluded that bradykinin activates PKCε via the PLD/PDK-1 pathway, which subsequently induces activation and translocation of ERK1 into the nucleus, and contributes to COX-2 expression for prostaglandin E2 synthesis in dermal fibroblasts.
Collapse
|
7
|
Murata M, Osanai M, Takasawa A, Takasawa K, Aoyama T, Kawada Y, Yamamoto A, Ono Y, Hiratsuka Y, Kojima T, Sawada N. Occludin induces microvillus formation via phosphorylation of ezrin in a mouse hepatic cell line. Exp Cell Res 2018; 366:172-180. [PMID: 29555369 DOI: 10.1016/j.yexcr.2018.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 11/30/2022]
Abstract
Apical and basolateral cell membranes are separated by tight junctions (TJs). Microvilli are limited to the apical cell membrane. TJs and microvilli are the landmarks for epithelial cell polarity. However, the direct relationship between TJ proteins (TJPs) and the components of microvilli remains unclear. In this study, we investigated whether occludin, which is considered to be a functional TJP, is involved in microvillus formation. In occludin knockout mouse hepatic cells (OcKO cells), the microvillus density was less than that in wild-type (WT) cells and the length of microvilli was short. Immunoreactivity of ezrin was decreased in OcKO cells compared with that in WT cells. Although there was no change in the expression level of ezrin, phosphorylation of ezrin was decreased in OcKO cells. The microvillus density and the length of microvilli were increased in OcKO cells by transfection of full-length mouse occludin and COOH-terminal domains of occludin. These results suggested that occludin induced microvillus formation via phosphorylation of ezrin and that the COOH-terminal domain of occludin, which is localized in non-TJ areas, might be able to induce microvilli formation. Our results provide new insights into the function of occludin.
Collapse
Affiliation(s)
- Masaki Murata
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan.
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Kumi Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Tomoyuki Aoyama
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yuka Kawada
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Akihiro Yamamoto
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yusuke Ono
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yutaro Hiratsuka
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute of Frontier Medicine, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Norimasa Sawada
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
8
|
Horikoshi Y, Kamizaki K, Hanaki T, Morimoto M, Kitagawa Y, Nakaso K, Kusumoto C, Matsura T. α-Tocopherol promotes HaCaT keratinocyte wound repair through the regulation of polarity proteins leading to the polarized cell migration. Biofactors 2018; 44:180-191. [PMID: 29399897 DOI: 10.1002/biof.1414] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022]
Abstract
In many developed countries including Japan, how to care the bedridden elderly people with chronic wounds such as decubitus becomes one of the most concerned issues. Although antioxidant micronutrients including vitamin E, especially α-tocopherol (α-Toc), are reported to shorten a period of wound closure, the promoting effect of α-Toc on wound healing independent of its antioxidant activity remains to be fully elucidated. The aim of this study was to examine whether α-Toc affects wound-mediated HaCaT keratinocyte polarization process including the recruitment of polarity regulating proteins, leading to wound repair independently of its antioxidant activity. We investigated the effects of α-Toc and other antioxidants such as Trolox, a cell-permeable α-Toc analog on the migration, proliferation, and cell polarization of HaCaT keratinocytes after wounding. We analyzed the localization and complex formation of polarity proteins, partitioning defective 3 (Par3), and atypical protein kinase C (aPKC), and aPKC activity by immunohistochemistry, immunoprecipitation analyses, and in vitro kinase assays, respectively. α-Toc but not other antioxidants enhanced the wound closure and cell polarization in HaCaT keratinocytes after wounding. α-Toc regulated the localization and complex formation of Par3 and aPKC during wound healing. Knockdown of aPKC or Par3 abrogated α-Toc-mediated promotion of the wound closure and cell polarization in HaCaT keratinocytes. Furthermore, aPKC kinase activity was significantly increased in α-Toc-treated cells through activation of phosphatidylinositol 3-kinase/Akt signaling pathway. These results suggest that α-Toc promotes HaCaT keratinocyte wound repair by regulating the aPKC kinase activity and the formation of aPKC-Par3 complex. © 2017 BioFactors, 44(2):180-191, 2018.
Collapse
Affiliation(s)
- Yosuke Horikoshi
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kouki Kamizaki
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
| | - Takehiko Hanaki
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
- Division of Surgical Oncology, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Masaki Morimoto
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
- Division of Surgical Oncology, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Yoshinori Kitagawa
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kazuhiro Nakaso
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Faculty of Health Sciences, Hiroshima International University, Higashihiroshima, Japan
| | - Tatsuya Matsura
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
9
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
10
|
A Review of Anti-Angiogenic Targets for Monoclonal Antibody Cancer Therapy. Int J Mol Sci 2017; 18:ijms18081786. [PMID: 28817103 PMCID: PMC5578174 DOI: 10.3390/ijms18081786] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is a key event that governs tumor progression and metastasis. It is controlled by the complicated and coordinated actions of pro-angiogenic factors and their receptors that become upregulated during tumorigenesis. Over the past several decades, vascular endothelial growth factor (VEGF) signaling has been identified as a central axis in tumor angiogenesis. The remarkable advent of recombinant antibody technology has led to the development of bevacizumab, a humanized antibody that targets VEGF and is a leading clinical therapy to suppress tumor angiogenesis. However, despite the clinical efficacy of bevacizumab, its significant side effects and drug resistance have raised concerns necessitating the identification of novel drug targets and development of novel therapeutics to combat tumor angiogenesis. This review will highlight the role and relevance of VEGF and other potential therapeutic targets and their receptors in angiogenesis. Simultaneously, we will also cover the current status of monoclonal antibodies being developed to target these candidates for cancer therapy.
Collapse
|
11
|
Aberrant Expression of the Cell Polarity Regulator aPKCλ/ι is Associated With Disease Progression in Cervical Intraepithelial Neoplasia (CIN): A Possible Marker for Predicting CIN Prognosis. Int J Gynecol Pathol 2016; 35:106-17. [PMID: 26535980 DOI: 10.1097/pgp.0000000000000228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Atypical protein kinase C λ/ι (aPKCλ/ι) is a regulator of epithelial cellular polarity. It is also overexpressed in several cancers and functions in cell proliferation and invasion. Therefore, we hypothesized that aPKCλ/ι may be involved in development and progression of cervical intraepithelial neoplasia (CIN), the precancerous disease of cervical cancer induced by human papillomavirus. To do this, we investigated the relationship between aPKCλ/ι expression and CIN. aPKCλ/ι expression level and subcellular localization were assessed in 192 CIN biopsy samples and 13 normal epithelial samples using immunohistochemistry. aPKCλ/ι overexpression (normal epithelium, 7.7%; CIN1, 41.7%; CIN2/3, 76.4%) and aPKCλ/ι nuclear localization (normal epithelium, 0.0%; CIN1, 36.9%; CIN2/3, 78.7%) were higher in CIN samples than normal samples (P<0.05), suggesting that CIN grade is related to aPKCλ/ι overexpression and nuclear localization. Then, 140 CIN cases were retrospectively analyzed for 4-yr cumulative disease progression and regression rates using the Cox proportional hazards model. CIN1 cases with aPKCλ/ι overexpression or aPKCλ/ι nuclear localization had a higher progression rate than CIN1 cases with normal aPKCλ/ι expression levels or cytoplasmic localization (62.5% vs. 9.7% and 63.1% vs. 9.4%, respectively; P<0.001). Multivariate analysis indicated that human papillomavirus types 16 and 18, aPKCλ/ι overexpression (hazard ratio=4.26; 95% confidence interval, 1.50-12.1; P=0.007), and aPKCλ/ι nuclear localization (hazard ratio=3.59; 95% confidence interval, 1.24-10.4; P=0.019) were independent risk factors for CIN1 progression. In conclusion, aPKCλ/ι could be useful for the therapeutic management of patients with CIN, particularly those with non-human papillomavirus 16/18 types.
Collapse
|
12
|
Nicotine enhances the malignant potential of human pancreatic cancer cells via activation of atypical protein kinase C. Biochim Biophys Acta Gen Subj 2016; 1860:2404-2415. [DOI: 10.1016/j.bbagen.2016.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/22/2016] [Accepted: 07/13/2016] [Indexed: 01/04/2023]
|
13
|
Banday AA. Chronic insulin treatment phosphorylates the renal Na-K-ATPase α1-subunit at serine 16/23 and reduces its activity involving PI3-kinase-dependent PKC activation. Am J Physiol Renal Physiol 2016; 311:F958-F966. [PMID: 27605582 DOI: 10.1152/ajprenal.00355.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022] Open
Abstract
The regulation of Na-K-ATPase in various tissues is under the control of a number of hormones and peptides that exert both short- and long-term control over its activity. The present study was performed to investigate the effect of chronic insulin treatment on Na-K-ATPase in renal proximal tubular cells. Incubation of opossum kidney (OK) cells, transfected with the rat Na-K-ATPase α1-subunit, with 1 nmol/l insulin for 48 h decreased Na-K-ATPase activity. Insulin decreased α1-protein content and increased α1-serine phosphorylation and α1-adaptor protein 2 (AP2) interaction. Removal of the 26 NH2-terminal (-NT) amino acid from the α1-subunit containing serine/threonine sites abolished the insulin-mediated serine phosphorylation and inhibition of Na-K-ATPase. Substitution of serine 16 and 23 with alanine showed a comparable effect on -NT. Insulin increased the activity of protein kinase C (PKC), which was blocked by the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin. Both PI3K and PKC inhibitors abolished the insulin-mediated inhibition of Na-K-ATPase. Insulin increased the expression of PKC-β1, -δ, -ξ, and-λ; however, only PKC-ξ/λ-specific inhibitors blocked insulin-induced phosphorylation and inhibition of Na-K-ATPase. Our data demonstrate that insulin activates the atypical PKC isoforms-ξ/λ via the PI3K pathway. PKC-ξ/λ-induced phosphorylation of the α1-subunit at serine 16 and 23 leads to AP2 recruitment, degradation, and a decrease in Na-K-ATPase activity.
Collapse
|
14
|
Ni S, Chen L, Li M, Zhao W, Shan X, Wu M, Cheng J, Liang L, Wang Y, Jiang W, Zhang J, Ni R. PKC iota promotes cellular proliferation by accelerated G1/S transition via interaction with CDK7 in esophageal squamous cell carcinoma. Tumour Biol 2016; 37:13799-13809. [PMID: 27481515 DOI: 10.1007/s13277-016-5193-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022] Open
Abstract
Protein kinase C iota (PKCι) has been shown to play an important role in tumorigenesis of many cancers. It was reported that frequent amplification and overexpression of PKCi were correlated with resistance to anoikis in primary esophageal squamous cell carcinomas (ESCC). In this study, we clarified a novel role of PKCι on the cell cycle progression and proliferation in ESCC. Western blot and immunohistochemistry (IHC) analysis showed that the expression of PKCι was higher in ESCC tumor tissues and cell lines. Meanwhile, IHC stain revealed that PKCι was positively correlated with clinical pathologic variables such as tumor size, tumor grade, and tumor invasion, as well as ki67. Immunoprecipitation and immunofluorescence assay revealed that PKCι/CDK7 has the physical interaction and were co-located in the cell nucleus. And this direct interaction could increase the phosphorylation level of CDK7. In vitro studies such as starvation and refeeding assay along with PKCι-shRNA transfection assay demonstrated that PKCι expression promoted proliferation of ESCC cells. And knocking PKCi down by silencing RNA (siRNA) significantly caused cell cycle arrest at G0/G1 phase, decreased rate of colony formation, and alleviated cellular apoptosis. This research provide new insights into PKCi signaling to more deeply understand its cancer-promoting function in ESCC.
Collapse
Affiliation(s)
- Sujie Ni
- Department of Medical Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Lingling Chen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Mei Li
- Department of Medical Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Weijuan Zhao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Xiaohang Shan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Miaomiao Wu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Jialin Cheng
- Department of Medical Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Li Liang
- Department of Medical Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Yayun Wang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Wenyan Jiang
- Department of Medical Oncology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China.
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
15
|
Kamishibahara Y, Kawaguchi H, Shimizu N. Rho kinase inhibitor Y-27632 promotes neuronal differentiation in mouse embryonic stem cells via phosphatidylinositol 3-kinase. Neurosci Lett 2016; 615:44-9. [PMID: 26797580 DOI: 10.1016/j.neulet.2016.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
Rho kinase (ROCK) regulates the functions of several target proteins via its kinase activity. Therefore, ROCK activity inhibition may provide new possibilities of controlling the in vitro neuronal differentiation of embryonic stem (ES) cells. When we investigated the effects of the ROCK inhibitor Y-27632 on ES cell differentiation, we found that this inhibitor promoted the differentiation of these cells into neurons. Furthermore, we found that ROCK inhibition may promote the neuronal differentiation of ES cells by activating extracellular signal-regulated kinase (ERK) involved in the ERK signaling pathway. In this study, we investigated the effects of specific inhibitors of several cellular signaling components on the promotion of neuronal differentiation in ES cells to clarify the roles of cellular signaling pathways in the ROCK inhibitor-mediated cell differentiation process. Our results suggest that ERK may be activated via the Ras/Raf/MEK, the PI3K/PKC, or the Cdc42/Rac signaling pathways in the ROCK inhibitor-mediated promotion of neuronal differentiation in ES cells.
Collapse
Affiliation(s)
- Yu Kamishibahara
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Hideo Kawaguchi
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Norio Shimizu
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan; Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama 350-8585, Japan.
| |
Collapse
|
16
|
Protein kinase Cζ exhibits constitutive phosphorylation and phosphatidylinositol-3,4,5-triphosphate-independent regulation. Biochem J 2015; 473:509-23. [PMID: 26635352 DOI: 10.1042/bj20151013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
Abstract
Atypical protein kinase C (aPKC) isoenzymes are key modulators of insulin signalling, and their dysfunction correlates with insulin-resistant states in both mice and humans. Despite the engaged interest in the importance of aPKCs to type 2 diabetes, much less is known about the molecular mechanisms that govern their cellular functions than for the conventional and novel PKC isoenzymes and the functionally-related protein kinase B (Akt) family of kinases. Here we show that aPKC is constitutively phosphorylated and, using a genetically-encoded reporter for PKC activity, basally active in cells. Specifically, we show that phosphorylation at two key regulatory sites, the activation loop and turn motif, of the aPKC PKCζ in multiple cultured cell types is constitutive and independently regulated by separate kinases: ribosome-associated mammalian target of rapamycin complex 2 (mTORC2) mediates co-translational phosphorylation of the turn motif, followed by phosphorylation at the activation loop by phosphoinositide-dependent kinase-1 (PDK1). Live cell imaging reveals that global aPKC activity is constitutive and insulin unresponsive, in marked contrast to the insulin-dependent activation of Akt monitored by an Akt-specific reporter. Nor does forced recruitment to phosphoinositides by fusing the pleckstrin homology (PH) domain of Akt to the kinase domain of PKCζ alter either the phosphorylation or activity of PKCζ. Thus, insulin stimulation does not activate PKCζ through the canonical phosphatidylinositol-3,4,5-triphosphate-mediated pathway that activates Akt, contrasting with previous literature on PKCζ activation. These studies support a model wherein an alternative mechanism regulates PKCζ-mediated insulin signalling that does not utilize conventional activation via agonist-evoked phosphorylation at the activation loop. Rather, we propose that scaffolding near substrates drives the function of PKCζ.
Collapse
|
17
|
Targeting Protein Kinase C Downstream of Growth Factor and Adhesion Signalling. Cancers (Basel) 2015; 7:1271-91. [PMID: 26184315 PMCID: PMC4586769 DOI: 10.3390/cancers7030836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/25/2015] [Accepted: 07/03/2015] [Indexed: 12/11/2022] Open
Abstract
The signaling outputs of Receptor Tyrosine Kinases, G-protein coupled receptors and integrins converge to mediate key cell process such as cell adhesion, cell migration, cell invasion and cell proliferation. Once activated by their ligands, these cell surface proteins recruit and direct a diverse range of proteins to disseminate the appropriate response downstream of the specific environmental cues. One of the key groups of proteins required to regulate these activities is the family of serine/threonine intracellular kinases called Protein Kinase Cs. The activity and subcellular location of PKCs are mediated by a series of tightly regulated events and is dependent on several posttranslational modifications and the availability of second messengers. Protein Kinase Cs exhibit both pro- and anti-tumorigenic effects making them an interesting target for anti-cancer treatment.
Collapse
|
18
|
Kusne Y, Carrera-Silva EA, Perry AS, Rushing EJ, Mandell EK, Dietrich JD, Errasti AE, Gibbs D, Berens ME, Loftus JC, Hulme C, Yang W, Lu Z, Aldape K, Sanai N, Rothlin CV, Ghosh S. Targeting aPKC disables oncogenic signaling by both the EGFR and the proinflammatory cytokine TNFα in glioblastoma. Sci Signal 2014; 7:ra75. [PMID: 25118327 PMCID: PMC4486020 DOI: 10.1126/scisignal.2005196] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Grade IV glioblastoma is characterized by increased kinase activity of epidermal growth factor receptor (EGFR); however, EGFR kinase inhibitors have failed to improve survival in individuals with this cancer because resistance to these drugs often develops. We showed that tumor necrosis factor-α (TNFα) produced in the glioblastoma microenvironment activated atypical protein kinase C (aPKC), thereby producing resistance to EGFR kinase inhibitors. Additionally, we identified that aPKC was required both for paracrine TNFα-dependent activation of the transcription factor nuclear factor κB (NF-κB) and for tumor cell-intrinsic receptor tyrosine kinase signaling. Targeting aPKC decreased tumor growth in mouse models of glioblastoma, including models of EGFR kinase inhibitor-resistant glioblastoma. Furthermore, aPKC abundance and activity were increased in human glioblastoma tumor cells, and high aPKC abundance correlated with poor prognosis. Thus, targeting aPKC might provide an improved molecular approach for glioblastoma therapy.
Collapse
Affiliation(s)
- Yael Kusne
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | - Anthony S. Perry
- Department of Pathology, Banner MD Anderson Cancer Center, Gilbert, AZ 85234, USA
| | | | - Edward K. Mandell
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Andrea E. Errasti
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniel Gibbs
- Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Michael E. Berens
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA
| | | | | | - Weiwei Yang
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhimin Lu
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Nader Sanai
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Carla V. Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sourav Ghosh
- Neuroscience Graduate Program, Arizona State University, Phoenix, AZ 85287, USA
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA
| |
Collapse
|
19
|
Satoh D, Hirose T, Harita Y, Daimon C, Harada T, Kurihara H, Yamashita A, Ohno S. aPKCλ maintains the integrity of the glomerular slit diaphragm through trafficking of nephrin to the cell surface. J Biochem 2014; 156:115-28. [PMID: 24700503 PMCID: PMC4112437 DOI: 10.1093/jb/mvu022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The slit diaphragm (SD), the specialized intercellular junction between renal glomerular epithelial cells (podocytes), provides a selective-filtration barrier in renal glomeruli. Dysfunction of the SD results in glomerular diseases that are characterized by disappearance of SD components, such as nephrin, from the cell surface. Although the importance of endocytosis and degradation of SD components for the maintenance of SD integrity has been suggested, the dynamic nature of the turnover of intact cell-surface SD components remained unclear. Using isolated rat glomeruli we show that the turnover rates of cell-surface SD components are relatively high; they almost completely disappear from the cell surface within minutes. The exocytosis, but not endocytosis, of heterologously expressed nephrin requires the kinase activity of the cell polarity regulator atypical protein kinase C (aPKC). Consistently, we demonstrate that podocyte-specific deletion of aPKCλ resulted in a decrease of cell-surface localization of SD components, causing massive proteinuria. In conclusion, the regulation of SD turnover by aPKC is crucial for the maintenance of SD integrity and defects in aPKC signalling can lead to proteinuria. These findings not only reveal the pivotal importance of the dynamic turnover of cell-surface SD components but also suggest a novel pathophysiological basis in glomerular disease.
Collapse
Affiliation(s)
- Daisuke Satoh
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Tomonori Hirose
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Yutaka Harita
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, JapanDepartment of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Chikara Daimon
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Tomonori Harada
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hidetake Kurihara
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, JapanDepartment of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| |
Collapse
|
20
|
Linch M, Sanz-Garcia M, Rosse C, Riou P, Peel N, Madsen CD, Sahai E, Downward J, Khwaja A, Dillon C, Roffey J, Cameron AJ, Parker PJ. Regulation of polarized morphogenesis by protein kinase C iota in oncogenic epithelial spheroids. Carcinogenesis 2014; 35:396-406. [PMID: 24072773 PMCID: PMC3908745 DOI: 10.1093/carcin/bgt313] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 08/07/2013] [Accepted: 08/24/2013] [Indexed: 12/20/2022] Open
Abstract
Protein kinase C iota (PKCι), a serine/threonine kinase required for cell polarity, proliferation and migration, is commonly up- or downregulated in cancer. PKCι is a human oncogene but whether this is related to its role in cell polarity and what repertoire of oncogenes acts in concert with PKCι is not known. We developed a panel of candidate oncogene expressing Madin-Darby canine kidney (MDCK) cells and demonstrated that H-Ras, ErbB2 and phosphatidylinositol 3-kinase transformation led to non-polar spheroid morphogenesis (dysplasia), whereas MDCK spheroids expressing c-Raf or v-Src were largely polarized. We show that small interfering RNA (siRNA)-targeting PKCι decreased the size of all spheroids tested and partially reversed the aberrant polarity phenotype in H-Ras and ErbB2 spheroids only. This indicates distinct requirements for PKCι and moreover that different thresholds of PKCι activity are required for these phenotypes. By manipulating PKCι function using mutant constructs, siRNA depletion or chemical inhibition, we have demonstrated that PKCι is required for polarization of parental MDCK epithelial cysts in a 3D matrix and that there is a threshold of PKCι activity above and below which, disorganized epithelial morphogenesis results. Furthermore, treatment with a novel PKCι inhibitor, CRT0066854, was able to restore polarized morphogenesis in the dysplastic H-Ras spheroids. These results show that tightly regulated PKCι is required for normal-polarized morphogenesis in mammalian cells and that H-Ras and ErbB2 cooperate with PKCι for loss of polarization and dysplasia. The identification of a PKCι inhibitor that can restore polarized morphogenesis has implications for the treatment of Ras and ErbB2 driven malignancies.
Collapse
Affiliation(s)
- Mark Linch
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
- Sarcoma Unit, Royal Marsden Hospital, London SW3 6JJ, UK
| | - Marta Sanz-Garcia
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Carine Rosse
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Philippe Riou
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Nick Peel
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | | | | | - Julian Downward
- Department of Signal Transduction Laboratories, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Asim Khwaja
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Christian Dillon
- Cancer Research Technology Discovery Laboratories, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK and
| | - Jon Roffey
- Cancer Research Technology Discovery Laboratories, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK and
| | - Angus J.M. Cameron
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Peter J. Parker
- Department of Protein Phosphorylation, Cancer Research UK London Research Institute, London WC2A 3LY, UK
- Division of Cancer Studies, King’s College London, London SE1 1UL, UK
| |
Collapse
|
21
|
Guan HP, Chen G. Factors affecting insulin-regulated hepatic gene expression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:165-215. [PMID: 24373238 DOI: 10.1016/b978-0-12-800101-1.00006-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity has become a major concern of public health. A common feature of obesity and related metabolic disorders such as noninsulin-dependent diabetes mellitus is insulin resistance, wherein a given amount of insulin produces less than normal physiological responses. Insulin controls hepatic glucose and fatty acid metabolism, at least in part, via the regulation of gene expression. When the liver is insulin-sensitive, insulin can stimulate the expression of genes for fatty acid synthesis and suppress those for gluconeogenesis. When the liver becomes insulin-resistant, the insulin-mediated suppression of gluconeogenic gene expression is lost, whereas the induction of fatty acid synthetic gene expression remains intact. In the past two decades, the mechanisms of insulin-regulated hepatic gene expression have been studied extensively and many components of insulin signal transduction pathways have been identified. Factors that alter these pathways, and the insulin-regulated hepatic gene expression, have been revealed and the underlying mechanisms have been proposed. This chapter summarizes the recent progresses in our understanding of the effects of dietary factors, drugs, bioactive compounds, hormones, and cytokines on insulin-regulated hepatic gene expression. Given the large amount of information and progresses regarding the roles of insulin, this chapter focuses on findings in the liver and hepatocytes and not those described for other tissues and cells. Typical insulin-regulated hepatic genes, such as insulin-induced glucokinase and sterol regulatory element-binding protein-1c and insulin-suppressed cytosolic phosphoenolpyruvate carboxyl kinase and insulin-like growth factor-binding protein 1, are used as examples to discuss the mechanisms such as insulin regulatory element-mediated transcriptional regulation. We also propose the potential mechanisms by which these factors affect insulin-regulated hepatic gene expression and discuss potential future directions of the area of research.
Collapse
Affiliation(s)
- Hong-Ping Guan
- Department of Diabetes, Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
22
|
Zou Q, Hou Y, Shen F, Wang Y. Polarized regulation of glycogen synthase kinase-3β is important for glioma cell invasion. PLoS One 2013; 8:e81814. [PMID: 24312592 PMCID: PMC3849364 DOI: 10.1371/journal.pone.0081814] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 10/16/2013] [Indexed: 11/24/2022] Open
Abstract
Glioma malignancy greatly depends on its aggressive invasion. The establishment of cell polarity is an important initial step for cell migration, which is essential for cell-directional translocation. However, our understanding of the molecular mechanisms underlying cell polarity formation in glioma cell invasion remains limited. Glycogen synthase kinase-3 (GSK-3) has a critical role in the formation of cell polarity. We therefore investigated whether localized GSK-3β, a subtype of GSK-3, is important for glioma cell invasion. We reported here that the localized phosphorylation of GSK-3β at the Ser9 (pSer9-GSK-3β) was critical for glioma cell invasion. Scratching glioma cell monolayer up-regulated pSer9-GSK-3β specifically at the wound edge. Inhibition of GSK-3 impaired the cell polarity and reduced the directional persistence of cell migration. Consistently, down-regulation of GSK-3α and 3β by specific small interfering RNAs inhibited glioma cell invasion. Over-expressing wild-type or constitutively active forms of GSK-3β also inhibited the cell invasion. These results indicated the polarized localization of GSK-3 regulation in cell migration might be also important for glioma cell migration. Further, EGF regulated both GSK-3α and 3β, but only pSer9-GSK-3β was enriched at the leading edge of scratched glioma cells. Up- or down-regulation of GSK-3β inhibited EGF-stimulated cell invasion. Moreover, EGF specifically regulated GSK-3β, but not GSK-3α, through atypical PKC pathways. Our results indicated that GSK-3 was important for glioma cell invasion and localized inhibition of GSK-3β was critical for cell polarity formation.
Collapse
Affiliation(s)
- Qifei Zou
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ying Hou
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Feng Shen
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
- * E-mail: (FS); (YZW)
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (FS); (YZW)
| |
Collapse
|
23
|
Xing B, Wang L, Guo D, Huang J, Espenel C, Kreitzer G, Zhang JJ, Guo L, Huang XY. Atypical protein kinase Cλ is critical for growth factor receptor-induced dorsal ruffle turnover and cell migration. J Biol Chem 2013; 288:32827-36. [PMID: 24092753 DOI: 10.1074/jbc.m113.489427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Gα13, a member of the heterotrimeric G proteins, is critical for actin cytoskeletal reorganization and cell migration. Previously we have shown that Gα13 is essential for both G protein-coupled receptor and receptor tyrosine kinase-induced actin cytoskeletal reorganization such as dynamic dorsal ruffle turnover and cell migration. Ric-8A, a non-receptor guanine nucleotide exchange factor for some heterotrimeric G proteins, is critical for coupling receptor tyrosine kinases to Gα13. Here, we show that PDGF can induce phosphorylation of Ric-8A. Atypical protein kinase Cλ (aPKCλ) is required for Ric-8A phosphorylation. Furthermore, aPKCλ is required for PDGF-induced dorsal ruffle turnover and cell migration as demonstrated by both down-regulation of aPKCλ protein levels in cells by RNA interference and by studies in aPKCλ knock-out cells. Moreover, phosphorylation of Ric-8A modulates its subcellular localization. Hence, aPKCλ is critical for PDGF-induced actin cytoskeletal reorganization and cell migration.
Collapse
Affiliation(s)
- Bowen Xing
- From the College of Life Sciences, Wuhan University, Wuhan 430072, China, and
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kato S, Akimoto K, Nagashima Y, Ishiguro H, Kubota K, Kobayashi N, Hosono K, Watanabe S, Sekino Y, Sato T, Sasaki K, Nakaigawa N, Kubota Y, Inayama Y, Endo I, Ohno S, Maeda S, Nakajima A. aPKCλ/ι is a beneficial prognostic marker for pancreatic neoplasms. Pancreatology 2013; 13:360-8. [PMID: 23890134 DOI: 10.1016/j.pan.2013.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a lethal disease. Overall survival is typically 6 months from diagnosis. Determination of prognostic factors in pancreatic cancer that would allow identification of patients who could potentially benefit from aggressive treatment is important. However, until date, there are no established reliable prognostic factors for pancreatic cancer patients. Herein, we propose a beneficial biomarker which is significantly correlated with the prognosis in pancreatic cancer patients. Atypical protein kinase C λ/ι (aPKCλ/ι) is overexpressed and has been implicated in the progression of several cancers. We tested the expression levels of aPKCλ/ι in two types of pancreatic neoplasm, pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasms (IPMNs), by immunohistochemistry. Examination of the aPKCλ/ι expression levels in surgically resected specimens of PDCA (n = 115) demonstrated that the expression levels of aPKCλ/ιin PDAC had prognostic implications, independent of the Tumor-Node-Metastasis classification and World Health Organization tumor grade. In the case of IPMNs (n = 46) also, the expression levels of aPKCλ/ιin IPMN were found to be of prognostic importance, independent of the World Health Organization histological grade or morphological type. Interestingly, high expression levels of aPKCλ/ι were significantly correlated with a worse histological grade (p = 0.010) and advanced stage of the tumor (p = 0.0050) in IPMN patients. These findings suggest that high expression levels of aPKCλ/ι could be involved in the malignant transformation of IPMNs. Based on these observations, we propose the expression level of aPKCλ/ι as a prognostic marker common to different types of pancreatic neoplasms.
Collapse
Affiliation(s)
- Shingo Kato
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Awadelkarim KD, Callens C, Rossé C, Susini A, Vacher S, Rouleau E, Lidereau R, Bièche I. Quantification of PKC family genes in sporadic breast cancer by qRT-PCR: evidence that PKCι/λ overexpression is an independent prognostic factor. Int J Cancer 2012; 131:2852-62. [PMID: 22511072 DOI: 10.1002/ijc.27600] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 04/05/2012] [Indexed: 11/11/2022]
Abstract
Drugs targeting protein kinase C (PKC) show promising therapeutic activity. However, little is known about the expression patterns of the 11 PKC genes in human tumors, and the clinical significance of most PKC genes is unknown. We used qRT-PCR assays to quantify mRNA levels of the 11 PKC genes in 458 breast tumors from patients with known clinical/pathological status and long-term outcome. The proportion of tumors in which the expression of the different genes was altered varied widely, from 9.6% for PKN2 to 40.2% for PKCι/λ. In breast tumors, overexpression was the main alteration observed for PKCι/λ (33.4%), PKCδ (29.5%) and PKCζ (9.6%), whereas underexpression was the main alteration observed for PKCα (27.3%), PKCε (11.6%), PKCη (8.7%) and PKN2 (8.1%). Both overexpression and underexpression were observed for PKCβ (underexpression 15.5%, overexpression 13.8%), PKCθ (underexpression 14.8%, overexpression 10.0%) and PKN1 (underexpression 6.6%, overexpression 7.4%). Several links were found between different PKC genes; and also between the expression patterns of PKC genes and several classical pathological and clinical parameters. PKCι/λ alone was found to have prognostic significance (p = 0.043), whereas PKCα showed a trend towards an influence on relapse-free survival (p = 0.052). PKCι/λ retained its prognostic significance in Cox multivariate regression analysis (p = 0.031). These results reveal very complex expression patterns of PKC genes in breast tumors, and suggest that their expression should be considered together when evaluating anti-tumoral drugs. PKCι/λ seems to be the most promising therapeutic target in breast cancer.
Collapse
|
26
|
Desai SR, Pillai PP, Patel RS, McCray AN, Win-Piazza HY, Acevedo-Duncan ME. Regulation of Cdk7 activity through a phosphatidylinositol (3)-kinase/PKC-ι-mediated signaling cascade in glioblastoma. Carcinogenesis 2011; 33:10-9. [PMID: 22021906 DOI: 10.1093/carcin/bgr231] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The objective of this research was to study the potential function of protein kinase C (PKC)-ι in cell cycle progression and proliferation in glioblastoma. PKC-ι is highly overexpressed in human glioma and benign and malignant meningioma; however, little is understood about its role in regulating cell proliferation of glioblastoma. Several upstream molecular aberrations and/or loss of PTEN have been implicated to constitutively activate the phosphatidylinositol (PI) (3)-kinase pathway. PKC-ι is a targeted mediator in the PI (3)-kinase signal transduction repertoire. Results showed that PKC-ι was highly activated and overexpressed in glioma cells. PKC-ι directly associated and phosphorylated Cdk7 at T170 in a cell cycle-dependent manner, phosphorylating its downstream target, cdk2 at T160. Cdk2 has a major role in inducing G(1)-S phase progression of cells. Purified PKC-ι phosphorylated both endogenous and exogenous Cdk7. PKC-ι downregulation reduced Cdk7 and cdk2 phosphorylation following PI (3)-kinase inhibition, phosphotidylinositol-dependent kinase 1 knockdown as well as PKC-ι silencing (by siRNA treatment). It also diminished cdk2 activity. PKC-ι knockdown inhibited overall proliferation rates and induced apoptosis in glioma cells. These findings suggest that glioma cells may be proliferating through a novel PI (3)-kinase-/PKC-ι/Cdk7/cdk2-mediated pathway.
Collapse
Affiliation(s)
- Shraddha R Desai
- James A. Haley Veteran's Hospital, 13000 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | | | | | | | | | | |
Collapse
|
27
|
Luna-Ulloa LB, Hernández-Maqueda JG, Santoyo-Ramos P, Castañeda-Patlán MC, Robles-Flores M. Protein kinase C ζ is a positive modulator of canonical Wnt signaling pathway in tumoral colon cell lines. Carcinogenesis 2011; 32:1615-24. [PMID: 21859831 DOI: 10.1093/carcin/bgr190] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The colonic epithelium is a continuously renewing tissue with a dynamic turnover of cells. Wnt pathway is a key regulator of its homeostasis and is altered in a large proportion of colon cancers. Protein kinase C (PKC) family of serine/threonine kinases are also involved in colon tumor formation and progression; however, the molecular role played by them in the Wnt pathway, is poorly understood. Reciprocal coimmunoprecipitation and immunofluorescence studies revealed that PKCζ interacts with β-catenin mainly in tumoral colon cells, which overexpressed this PKC isoform. The pharmacological inhibition, the small interference RNA-mediated knockdown of PKCζ or the expression of a dominant-negative form of it in tumoral SW480 cells, blocked in a dose-dependent manner the constitutive transcriptional activity mediated by β-catenin, the cell proliferation and the expression of the Wnt target gene c-myc. Remarkably, the PKCζ stably depleted cells exhibited diminished tumorigenic activity in grafted mice. We show that PKCζ functions in a mechanism that does not involve β-catenin degradation since the effects produced by PKCζ inhibition were also obtained in the presence of proteosome inhibitor and in cells expressing a β-catenin degradation-resistant mutant. It was found that PKCζ activity regulates the nuclear localization of β-catenin since PKCζ inhibition induces a rapid export of β-catenin from the nucleus to the cytoplasm in a Leptomycin B sensitive manner. Taken together, our results indicate that the atypical PKCζ plays an important role in the positive regulation of canonical Wnt pathway.
Collapse
Affiliation(s)
- Luis Bernardo Luna-Ulloa
- Department of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico, 04510, Mexico
| | | | | | | | | |
Collapse
|
28
|
Ishiguro H, Akimoto K, Nagashima Y, Kagawa E, Sasaki T, Sano JY, Takagawa R, Fujinami K, Sasaki K, Aoki I, Ohno S, Kubota Y, Uemura H. Coexpression of aPKCλ/ι and IL-6 in prostate cancer tissue correlates with biochemical recurrence. Cancer Sci 2011; 102:1576-81. [PMID: 21535317 DOI: 10.1111/j.1349-7006.2011.01972.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Atypical protein kinase C λ/ι (aPKCλ/ι) and interleukin-6 (IL-6) have been implicated in prostate cancer progression, the mechanisms of which have been demonstrated both in vitro and in vivo. However, the clinical significance of the correlation between the expressions of these factors remains to be clarified. In the present study, we report a significant correlation between aPKCλ/ι and IL-6 proteins in prostate cancer tissue by immunohistochemical staining. We evaluated the association of both proteins by analyzing clinicopathological parameters using chi-square test, Kaplan-Meier with log-rank test, and a Cox proportional hazard regression model in univariate and multivariate analyses. The results again showed that the expression of aPKCλ/ι and IL-6 correlates in prostate cancer tissue (P < 0.001). Atypical protein kinase C λ/ι was also found to correlate with the Gleason score (P < 0.001) and with biochemical recurrence after prostatectomy (P = 0.02). Furthermore, aPKCλ/ι correlated with biochemical recurrence in a Kaplan-Meier and log-rank test (P = 0.01) and Cox analysis (P = 0.02 in the univariate analysis, P = 0.02 in the multivariate analysis). The coexpression of aPKCλ/ι and IL-6 also correlated with biochemical recurrence by Kaplan-Meier and log-rank test (P = 0.005) and Cox analysis (P = 0.01 in the univariate analysis, P = 0.03 in the multivariate analysis). These results indicate a strong correlation between aPKCλ/ι and IL-6 in prostate tumors, and that the aPKCλ/ι-IL-6 axis is a reliable prognostic factor for the biochemical recurrence of this cancer.
Collapse
Affiliation(s)
- Hitoshi Ishiguro
- Department of Urology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
OBJECTIVE To determine whether the phosphoinositide 3-kinase (PI3K) catalytic subunits p110-α and p110-β play a role in liver steatosis induced by a high-fat diet (HFD). RESEARCH DESIGN AND METHODS Liver-specific p110-α and p110-β knockout mice and control animals for each group were fed an HFD or normal chow for 8 weeks. Biochemical assays and quantitative real-time PCR were used to measure triglyceride, expression of lipogenic and gluconeogenic genes, and activity of protein kinases downstream of PI3K in liver lysates. Fatty acid uptake and incorporation into triglycerides were assessed in isolated hepatocytes. RESULTS Hepatic triglyceride levels in HFD-fed p110-α(-/-) mice were 84 ± 3% lower than in p110-α(+/+) mice, whereas the loss of p110-β did not significantly alter liver lipid accumulation. p110-α(-/-) livers also showed a reduction in atypical protein kinase C activity and decreased mRNA and protein expression of several lipogenic genes. Hepatocytes isolated from p110-α(-/-) mice exhibited decreased palmitate uptake and reduced fatty acid incorporation into triglycerides as compared with p110-α(+/+) cells, and hepatic expression of liver fatty acid binding protein was lower in p110-α(-/-) mice fed the HFD as compared with controls. Ablation of neither p110-α nor p110-β ameliorated glucose intolerance induced by the HFD, and genes involved in gluconeogenesis were upregulated in the liver of both knockout animals. CONCLUSIONS PI3K p110-α, and not p110-β, promotes liver steatosis in mice fed an HFD. p110-α might exert this effect in part through activation of atypical protein kinase C, upregulation of lipogenesis, and increased uptake of fatty acids.
Collapse
Affiliation(s)
- Mohar Chattopadhyay
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York
| | | | - Lisa M. Ballou
- Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Richard Z. Lin
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York
- Department of Medicine, Stony Brook University, Stony Brook, New York
- Department of Veterans Affairs Medical Center, Northport, New York
- Corresponding author: Richard Z. Lin,
| |
Collapse
|
30
|
Desai S, Pillai P, Win-Piazza H, Acevedo-Duncan M. PKC-ι promotes glioblastoma cell survival by phosphorylating and inhibiting BAD through a phosphatidylinositol 3-kinase pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1190-7. [PMID: 21419810 DOI: 10.1016/j.bbamcr.2011.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 03/08/2011] [Accepted: 03/10/2011] [Indexed: 11/15/2022]
Abstract
The focus of this research was to investigate the role of protein kinase C-iota (PKC-ι) in regulation of Bad, a pro-apoptotic BH3-only molecule of the Bcl-2 family in glioblastoma. Robust expression of PKC-ι is a hallmark of human glioma and benign and malignant meningiomas. The results were obtained from the two human glial tumor derived cell lines, T98G and U87MG. In these cells, PKC-ι co-localized and directly associated with Bad, as shown by immunofluorescence, immunoprecipitation, and Western blotting. Furthermore, in-vitro kinase activity assay showed that PKC-ι directly phosphorylated Bad at phospho specific residues, Ser-112, Ser-136 and Ser-155 which in turn induced inactivation of Bad and disruption of Bad/Bcl-XL dimer. Knockdown of PKC-ι by siRNA exhibited a corresponding reduction in Bad phosphorylation suggesting that PKC-ι may be a Bad kinase. PKC-ι knockdown also induced apoptosis in both the cell lines. Since, PKC-ι is an essential downstream mediator of the PI (3)-kinase, we hypothesize that glioma cell survival is mediated via a PI (3)-kinase/PDK1/PKC-ι/Bad pathway. Treatment with PI (3)-kinase inhibitors Wortmannin and LY294002, as well as PDK1 siRNA, inhibited PKC-ι activity and subsequent phosphorylation of Bad suggesting that PKC-ι regulates the activity of Bad in a PI (3)-kinase dependent manner. Thus, our data suggest that glioma cell survival occurs through a novel PI (3)-kinase/PDK1/PKC-ι/BAD mediated pathway.
Collapse
Affiliation(s)
- S Desai
- James A. Haley Veteran's Hospital, Tampa, FL 33612, USA
| | | | | | | |
Collapse
|
31
|
Campa D, Hüsing A, Stein A, Dostal L, Boeing H, Pischon T, Tjønneland A, Roswall N, Overvad K, Østergaard JN, Rodríguez L, Sala N, Sánchez MJ, Larrañaga N, Huerta JM, Barricarte A, Khaw KT, Wareham N, Travis RC, Allen NE, Lagiou P, Trichopoulou A, Trichopoulos D, Palli D, Sieri S, Tumino R, Sacerdote C, van Kranen H, Bueno-de-Mesquita HB, Hallmans G, Johansson M, Romieu I, Jenab M, Cox DG, Siddiq A, Riboli E, Canzian F, Kaaks R. Genetic variability of the mTOR pathway and prostate cancer risk in the European Prospective Investigation on Cancer (EPIC). PLoS One 2011; 6:e16914. [PMID: 21373201 PMCID: PMC3044148 DOI: 10.1371/journal.pone.0016914] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 01/01/2011] [Indexed: 01/12/2023] Open
Abstract
The mTOR (mammalian target of rapamycin) signal transduction pathway integrates various signals, regulating ribosome biogenesis and protein synthesis as a function of available energy and amino acids, and assuring an appropriate coupling of cellular proliferation with increases in cell size. In addition, recent evidence has pointed to an interplay between the mTOR and p53 pathways. We investigated the genetic variability of 67 key genes in the mTOR pathway and in genes of the p53 pathway which interact with mTOR. We tested the association of 1,084 tagging SNPs with prostate cancer risk in a study of 815 prostate cancer cases and 1,266 controls nested within the European Prospective Investigation into Cancer and Nutrition (EPIC). We chose the SNPs (n = 11) with the strongest association with risk (p<0.01) and sought to replicate their association in an additional series of 838 prostate cancer cases and 943 controls from EPIC. In the joint analysis of first and second phase two SNPs of the PRKCI gene showed an association with risk of prostate cancer (ORallele = 0.85, 95% CI 0.78–0.94, p = 1.3×10−3 for rs546950 and ORallele = 0.84, 95% CI 0.76–0.93, p = 5.6×10−4 for rs4955720). We confirmed this in a meta-analysis using as replication set the data from the second phase of our study jointly with the first phase of the Cancer Genetic Markers of Susceptibility (CGEMS) project. In conclusion, we found an association with prostate cancer risk for two SNPs belonging to PRKCI, a gene which is frequently overexpressed in various neoplasms, including prostate cancer.
Collapse
Affiliation(s)
- Daniele Campa
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anika Hüsing
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angelika Stein
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lucie Dostal
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, Deutsches Institut für Ernährungsforschung, Potsdam-Rehbrücke, Germany
| | - Tobias Pischon
- Department of Epidemiology, Deutsches Institut für Ernährungsforschung, Potsdam-Rehbrücke, Germany
| | - Anne Tjønneland
- The Danish Cancer Society, Institute of Cancer Epidemiology, Copenhagen, Denmark
| | - Nina Roswall
- The Danish Cancer Society, Institute of Cancer Epidemiology, Copenhagen, Denmark
| | - Kim Overvad
- Department of Cardiology, Center for Cardiovascular Research, Aalborg Hospital, Aarhus University Hospital, Aalborg, Denmark
- Department of Epidemiology, School of Public Health, Aarhus University, Denmark
| | - Jane Nautrup Østergaard
- Department of Cardiology, Center for Cardiovascular Research, Aalborg Hospital, Aarhus University Hospital, Aalborg, Denmark
- Department of Epidemiology, School of Public Health, Aarhus University, Denmark
| | - Laudina Rodríguez
- Public Health and Participation Directorate, Health and Health Care Services Council, Asturias, Spain
| | - Núria Sala
- Catalan Institute of Oncology (ICO) - IDIBELL, Barcelona, Spain
| | - Maria-José Sánchez
- Andalusian School of Public Health, Granada, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Nerea Larrañaga
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Department of Gipuzkoa, Basque Government, Gipuzkoa, Spain
| | - José María Huerta
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Authority, Murcia, Spain
| | - Aurelio Barricarte
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarre Public Health Institute, Pamplona, Spain
| | - Kay-Tee Khaw
- University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Naomi E. Allen
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Pagona Lagiou
- WHO Collaborating Center for Food and Nutrition Policies, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston MA, USA
| | - Antonia Trichopoulou
- WHO Collaborating Center for Food and Nutrition Policies, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
- Hellenic Health Foundation, Athens, Greece
| | - Dimitrios Trichopoulos
- Department of Epidemiology, Harvard School of Public Health, Boston MA, USA
- Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Domenico Palli
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute – ISPO, Florence, Italy
| | - Sabina Sieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, “Civile - M.P.Arezzo” Hospital, ASP 7, Ragusa, Italy
| | - Carlotta Sacerdote
- Center for Cancer Prevention (CPO-Piemonte), Turin, Italy
- Human Genetic Foundation (HuGeF), Turin, Italy
| | - Henk van Kranen
- Centre for Nutrition and Health (CVG), National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - H. Bas Bueno-de-Mesquita
- Centre for Nutrition and Health (CVG), National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Göran Hallmans
- Dept of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Mattias Johansson
- Dept of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- International Agency for Research on Cancer, Lyon, France
| | | | - Mazda Jenab
- International Agency for Research on Cancer, Lyon, France
| | - David G. Cox
- Imperial College, London, United Kingdom
- INSERM U590, Centre Léon Bérard, Lyon France
| | | | | | | | - Rudolf Kaaks
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- * E-mail:
| |
Collapse
|
32
|
Sender V, Moulakakis C, Stamme C. Pulmonary surfactant protein A enhances endolysosomal trafficking in alveolar macrophages through regulation of Rab7. THE JOURNAL OF IMMUNOLOGY 2011; 186:2397-411. [PMID: 21248257 DOI: 10.4049/jimmunol.1002446] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Surfactant protein A (SP-A), the most abundant pulmonary soluble collectin, modulates innate and adaptive immunity of the lung, partially via its direct effects on alveolar macrophages (AM), the most predominant intra-alveolar cells under physiological conditions. Enhanced phagocytosis and endocytosis are key functional consequences of AM/SP-A interaction, suggesting a SP-A-mediated modulation of small Rab (Ras related in brain) GTPases that are pivotal membrane organizers in both processes. In this article, we show that SP-A specifically and transiently enhances the protein expression of endogenous Rab7 and Rab7b, but not Rab5 and Rab11, in primary AM from rats and mice. SP-A-enhanced GTPases are functionally active as determined by increased interaction of Rab7 with its downstream effector Rab7 interacting lysosomal protein (RILP) and enhanced maturation of cathepsin-D, a function of Rab7b. In AM and RAW264.7 macrophages, the SP-A-enhanced lysosomal delivery of GFP-Escherichia coli is abolished by the inhibition of Rab7 and Rab7 small interfering RNA transfection, respectively. The constitutive expression of Rab7 in AM from SP-A(-/-) mice is significantly reduced compared with SP-A(+/+) mice and is restored by SP-A. Rab7 blocking peptides antagonize SP-A-rescued lysosomal delivery of GFP-E. coli in AM from SP-A(-/-) mice. Activation of Rab7, but not Rab7b, by SP-A depends on the PI3K/Akt/protein kinase Cζ (PKCζ) signal transduction pathway in AM and RAW264.7 macrophages. SP-A induces a Rab7/PKCζ interaction in these cells, and the disruption of PKCζ by small interfering RNA knockdown abolishes the effect of SP-A on Rab7. The data demonstrate a novel role for SP-A in modulating endolysosomal trafficking via Rab7 in primary AM and define biochemical pathways involved.
Collapse
Affiliation(s)
- Vicky Sender
- Division of Cellular Pneumology, Department of Experimental Pneumology, Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23845 Borstel, Germany
| | | | | |
Collapse
|
33
|
Baldwin RM, Barrett GM, Parolin DAE, Gillies JK, Paget JA, Lavictoire SJ, Gray DA, Lorimer IAJ. Coordination of glioblastoma cell motility by PKCι. Mol Cancer 2010; 9:233. [PMID: 20815904 PMCID: PMC2941485 DOI: 10.1186/1476-4598-9-233] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 09/03/2010] [Indexed: 11/26/2022] Open
Abstract
Background Glioblastoma is one of the deadliest forms of cancer, in part because of its highly invasive nature. The tumor suppressor PTEN is frequently mutated in glioblastoma and is known to contribute to the invasive phenotype. However the downstream events that promote invasion are not fully understood. PTEN loss leads to activation of the atypical protein kinase C, PKCι. We have previously shown that PKCι is required for glioblastoma cell invasion, primarily by enhancing cell motility. Here we have used time-lapse videomicroscopy to more precisely define the role of PKCι in glioblastoma. Results Glioblastoma cells in which PKCι was either depleted by shRNA or inhibited pharmacologically were unable to coordinate the formation of a single leading edge lamellipod. Instead, some cells generated multiple small, short-lived protrusions while others generated a diffuse leading edge that formed around the entire circumference of the cell. Confocal microscopy showed that this behavior was associated with altered behavior of the cytoskeletal protein Lgl, which is known to be inactivated by PKCι phosphorylation. Lgl in control cells localized to the lamellipod leading edge and did not associate with its binding partner non-muscle myosin II, consistent with it being in an inactive state. In PKCι-depleted cells, Lgl was concentrated at multiple sites at the periphery of the cell and remained in association with non-muscle myosin II. Videomicroscopy also identified a novel role for PKCι in the cell cycle. Cells in which PKCι was either depleted by shRNA or inhibited pharmacologically entered mitosis normally, but showed marked delays in completing mitosis. Conclusions PKCι promotes glioblastoma motility by coordinating the formation of a single leading edge lamellipod and has a role in remodeling the cytoskeleton at the lamellipod leading edge, promoting the dissociation of Lgl from non-muscle myosin II. In addition PKCι is required for the transition of glioblastoma cells through mitosis. PKCι therefore has a role in both glioblastoma invasion and proliferation, two key aspects in the malignant nature of this disease.
Collapse
Affiliation(s)
- R Mitchell Baldwin
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa K1H 8L6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Tediose T, Kolev M, Sivasankar B, Brennan P, Morgan BP, Donev R. Interplay between REST and nucleolin transcription factors: a key mechanism in the overexpression of genes upon increased phosphorylation. Nucleic Acids Res 2010; 38:2799-812. [PMID: 20100803 PMCID: PMC2875004 DOI: 10.1093/nar/gkq013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Revised: 12/14/2009] [Accepted: 01/06/2010] [Indexed: 12/19/2022] Open
Abstract
Non-malignant cells can be transformed via the activation of kinases that control degradation of neural-restrictive silencer factor (REST). Here, we identify a mechanism that contributes to the activation of genes, expression of which is controlled by responsive elements containing overlapping binding sites for REST and nucleolin. We demonstrate that both phosphorylated and non-phosphorylated nucleolin-bound DNA; however, only phosphorylated nucleolin successfully competed with either full-length REST or a REST-derived DNA-binding peptide, REST68, for binding to the overlapping binding sites. We show that this interplay between the two transcription factors regulates the activation of cell survival and immunomodulatory genes in tumors and non-malignant cells with activated protein kinase C, which is accompanied with alterations in cell proliferation and apoptosis. We propose a model for the regulation of these genes, which brings a new insight into the molecular mechanisms that control cellular transformation driven by activation of protein kinases.
Collapse
Affiliation(s)
- Teeo Tediose
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Martin Kolev
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Baalasubramanian Sivasankar
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Paul Brennan
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - B. Paul Morgan
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Rossen Donev
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff CF14 4XN, and Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| |
Collapse
|
35
|
Angiogenesis inhibition in cancer therapy: platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) and their receptors: biological functions and role in malignancy. Recent Results Cancer Res 2010; 180:51-81. [PMID: 20033378 DOI: 10.1007/978-3-540-78281-0_5] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen in vitro and an angiogenic inducer in a variety of in vivo models. VEGF gene transcription is induced in particular in hypoxic cells. In developmental angiogenesis, the role of VEGF is demonstrated by the finding that the loss of a single VEGF allele results in defective vascularization and early embryonic lethality. Substantial evidence also implicates VEGF as a mediator of pathological angiogenesis. In situ hybridization studies demonstrate expression of VEGF mRNA in the majority of human tumors. Platelet-derived growth factor (PDGF) is mainly believed to be an important mitogen for connective tissue, and also has important roles during embryonal development. Its overexpression has been linked to different types of malignancies. Thus, it is important to understand the physiology of VEGF and PDGF and their receptors as well as their roles in malignancies in order to develop antiangiogenic strategies for the treatment of malignant disease.
Collapse
|
36
|
Sugiyama Y, Akimoto K, Robinson ML, Ohno S, Quinlan RA. A cell polarity protein aPKClambda is required for eye lens formation and growth. Dev Biol 2009; 336:246-56. [PMID: 19835853 PMCID: PMC2806522 DOI: 10.1016/j.ydbio.2009.10.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 10/03/2009] [Accepted: 10/05/2009] [Indexed: 11/28/2022]
Abstract
The organisation of individual cells into a functional three-dimensional tissue is still a major question in developmental biology. Modulation of epithelial cell shape is a critical driving force in forming tissues. This is well illustrated in the eye lens where epithelial cells elongate extensively during their differentiation into fibre cells. It is at the lens equator that epithelial cells elongate along their apical–basal axis. During this process the elongating epithelial cells and their earliest fibre cell derivatives remain anchored at their apical tips, forming a discrete region or modiolus, which we term the lens fulcrum. How this is achieved has received scant attention and is little understood. Here, we show that conditional depletion of aPKCλ, a central effector of the PAR polarity complex, disrupts the apical junctions in elongating epithelial cells so that the lens fulcrum fails to form. This results in disorganised fibre cell alignment that then causes cataract. Interestingly, aPKCλ depletion also promotes epithelial–mesenchymal transition of the lens epithelial cells, reducing their proliferation, leading ultimately to a small lens and microphthalmia. These observations indicate that aPKCλ, a regulator of polarity and apical junctions, is required for development of a lens that is the correct size and shape.
Collapse
Affiliation(s)
- Yuki Sugiyama
- School of Biological and Biomedical Sciences, University of Durham, Durham, UK
| | | | | | | | | |
Collapse
|
37
|
Takagawa R, Akimoto K, Ichikawa Y, Akiyama H, Kojima Y, Ishiguro H, Inayama Y, Aoki I, Kunisaki C, Endo I, Nagashima Y, Ohno S. High expression of atypical protein kinase C lambda/iota in gastric cancer as a prognostic factor for recurrence. Ann Surg Oncol 2009; 17:81-8. [PMID: 19774416 DOI: 10.1245/s10434-009-0708-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/21/2009] [Accepted: 08/22/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND The atypical protein kinase C lambda/iota (aPKClambda/iota) is involved in several signal transduction pathways that influence cell growth, apoptosis, and the establishment and maintenance of epithelial cell polarity. Overexpression of aPKClambda/iota has been reported in several cancers and been shown to be associated with oncogenesis. However, the expression and role of aPKClambda/iota in gastric cancer, one of the commonest cancers in Asia, have not so far been investigated. This study aimed to clarify the relationship between aPKClambda/iota expression and the clinicopathological features of gastric cancer. PATIENTS AND METHODS Gastric adenocarcinoma samples were obtained from 177 patients who underwent gastrectomy at the Yokohama City University Hospital between 1999 and 2004. Expression of aPKClambda/iota and E: -cadherin was examined immunohistochemically and compared with clinicopathological features of the tumors. Univariate and multivariate analyses were performed for both disease-specific and relapse-free survival. RESULTS Overexpression of aPKClambda/iota protein was detected in 126 of the 177 (71.2%) gastric cancers. Immunohistological staining for aPKClambda/iota was stronger in gastric adenocarcinoma of intestinal type than diffuse type (p = 0.036), but was not correlated with E: -cadherin expression. A multivariate analysis suggested that nodal metastasis and aPKClambda/iota overexpression were prognostic factors for disease recurrence. CONCLUSIONS Our results suggested that aPKClambda/iota overexpression was a strong prognostic factor for gastric adenocarcinoma recurrence. As well as being a new prognostic indicator, aPKClambda/iota is also likely to be a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Ryo Takagawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
aPKClambda/iota promotes growth of prostate cancer cells in an autocrine manner through transcriptional activation of interleukin-6. Proc Natl Acad Sci U S A 2009; 106:16369-74. [PMID: 19805306 DOI: 10.1073/pnas.0907044106] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Understanding the mechanism by which hormone refractory prostate cancer (HRPC) develops remains a major issue. Alterations in HRPC include androgen receptor (AR) changes. In addition, the AR is activated by cytokines such as interleukin-6 (IL-6). Atypical protein kinase C (aPKClambda/iota) has been implicated in the progression of several cancers. Herein, we provide evidence that aPKClambda/iota expression correlates with prostate cancer recurrence. Experiments in vitro and in vivo revealed aPKClambda/iota to be involved in prostate cancer cell growth through secretion of IL-6. Further, aPKClambda/iota activates transcription of the IL-6 gene through NFkappaB and AP-1. We conclude that aPKClambda/iota promotes the growth of hormone independent prostate cancer cells by stimulating IL-6 production in an autocrine manner. Our findings not only explain the link between aPKClambda/iota and IL-6, implicated in the progression a variety of cancers, but also establish a molecular change involved in the development of HRPC. Further, aPKClambda/iota expression might be a biomarker for prostate cancer progression.
Collapse
|
39
|
An essential role of the aPKC-Aurora A-NDEL1 pathway in neurite elongation by modulation of microtubule dynamics. Nat Cell Biol 2009; 11:1057-68. [PMID: 19668197 DOI: 10.1038/ncb1919] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 06/19/2009] [Indexed: 11/08/2022]
Abstract
Orchestrated remodelling of the cytoskeketon is prominent during neurite extension. In contrast with the extensive characterization of actin filament regulation, little is known about the dynamics of microtubules during neurite extension. Here we identify an atypical protein kinase C (aPKC)-Aurora A-NDEL1 pathway that is crucial for the regulation of microtubule organization during neurite extension. aPKC phosphorylates Aurora A at Thr 287 (T287), which augments interaction with TPX2 and facilitates activation of Aurora A at the neurite hillock, followed by phosphorylation of NDEL1 at S251 and recruitment. Suppression of aPKC, Aurora A or TPX2, or disruption of Ndel1, results in severe impairment of neurite extension. Analysis of microtubule dynamics with a microtubule plus-end marker revealed that suppression of the aPKC-Aurora A-NDEL1 pathway resulted in a significant decrease in the frequency of microtubule emanation from the microtubule organizing centre (MTOC), suggesting that Aurora A acts downstream of aPKC. These findings demonstrate a surprising role of aPKC-Aurora A-NDEL1 pathway in microtubule remodelling during neurite extension.
Collapse
|
40
|
Hengst U, Deglincerti A, Kim HJ, Jeon NL, Jaffrey SR. Axonal elongation triggered by stimulus-induced local translation of a polarity complex protein. Nat Cell Biol 2009; 11:1024-30. [PMID: 19620967 PMCID: PMC2724225 DOI: 10.1038/ncb1916] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 04/23/2009] [Indexed: 12/11/2022]
Affiliation(s)
- Ulrich Hengst
- Department of Pharmacology, Weill Medical College, Cornell University, NY 10065, USA
| | | | | | | | | |
Collapse
|
41
|
Wang G, Krishnamurthy K, Umapathy NS, Verin AD, Bieberich E. The carboxyl-terminal domain of atypical protein kinase Czeta binds to ceramide and regulates junction formation in epithelial cells. J Biol Chem 2009; 284:14469-75. [PMID: 19304661 DOI: 10.1074/jbc.m808909200] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Atypical protein kinase Cs (PKCs) (aPKCzeta and lambda/iota) have emerged as important binding partners for ceramide, a membrane-resident cell signaling lipid that is involved in the regulation of apoptosis as well as cell polarity. Using ceramide overlay assays with proteolytic fragments of PKCzeta and vesicle binding assays with ectopically expressed protein, we show that a protein fragment comprising the carboxyl-terminal 20-kDa sequence of PKCzeta (C20zeta, amino acids 405-592) bound to C16:0 ceramide. This sequence is not identical to the C1 domain (amino acids 131-180), which has been suggested to serve as a potential ceramide binding domain. Using immunocytochemistry, we found that a C20zeta protein fragment ectopically expressed in two epithelial cell types (neural progenitors and Madin-Darby canine kidney cells) co-distributed with ceramide. Stable expression of C20zeta-EGFP in Madin-Darby canine kidney cells disrupted the formation of adherens and tight junctions and impaired the epithelium integrity by reducing transepithelial electrical resistance. Disruption of cell adhesion and loss of transepithelial electrical resistance was prevented by incubation with C16:0 ceramide. Our results show, for the first time, that there is a novel ceramide binding domain (C20zeta) in the carboxyl terminus of aPKC. Our results also show that the interaction of ceramide with this binding domain is essential for cell-to-cell contacts in epithelia. Therefore, ceramide interaction with the C20zeta binding domain is a potential mechanism by which ceramide and aPKC regulate the formation of junctional complexes in epithelial cells.
Collapse
Affiliation(s)
- Guanghu Wang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
42
|
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) have served as prototypes for growth factor and receptor tyrosine kinase function for more than 25 years. Studies of PDGFs and PDGFRs in animal development have revealed roles for PDGFR-alpha signaling in gastrulation and in the development of the cranial and cardiac neural crest, gonads, lung, intestine, skin, CNS, and skeleton. Similarly, roles for PDGFR-beta signaling have been established in blood vessel formation and early hematopoiesis. PDGF signaling is implicated in a range of diseases. Autocrine activation of PDGF signaling pathways is involved in certain gliomas, sarcomas, and leukemias. Paracrine PDGF signaling is commonly observed in epithelial cancers, where it triggers stromal recruitment and may be involved in epithelial-mesenchymal transition, thereby affecting tumor growth, angiogenesis, invasion, and metastasis. PDGFs drive pathological mesenchymal responses in vascular disorders such as atherosclerosis, restenosis, pulmonary hypertension, and retinal diseases, as well as in fibrotic diseases, including pulmonary fibrosis, liver cirrhosis, scleroderma, glomerulosclerosis, and cardiac fibrosis. We review basic aspects of the PDGF ligands and receptors, their developmental and pathological functions, principles of their pharmacological inhibition, and results using PDGF pathway-inhibitory or stimulatory drugs in preclinical and clinical contexts.
Collapse
|
43
|
Kojima Y, Akimoto K, Nagashima Y, Ishiguro H, Shirai S, Chishima T, Ichikawa Y, Ishikawa T, Sasaki T, Kubota Y, Inayama Y, Aoki I, Ohno S, Shimada H. The overexpression and altered localization of the atypical protein kinase C lambda/iota in breast cancer correlates with the pathologic type of these tumors. Hum Pathol 2008; 39:824-31. [PMID: 18538170 DOI: 10.1016/j.humpath.2007.11.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 10/29/2007] [Accepted: 11/02/2007] [Indexed: 01/28/2023]
Abstract
Breast cancer is one of the common malignant diseases among women in Japan as well as in western countries, and its incidence continues to increase. Normal mammary duct epithelial cells exhibit a well-organized apicobasal polarity, which forms the basis for their specific structure and function. Although the loss of epithelial cell polarity is one of the major changes that occur during the progression of tumor cells, including breast cancer, the underlying molecular mechanisms for this, as well as their relationship to other changes such as increased proliferation and metastasis, remain to be elucidated. The atypical protein kinase C lambda/iota (aPKC lambda/iota) is involved in several signal transduction pathways, including the establishment of epithelial cell polarity. In this study we evaluated the expression and localization of aPKC lambda/iota in breast cancer by immunohistochemistry and compared our findings with the clinicopathologic factors associated with the tumor specimens. We detected aPK Clambda/iota protein overexpression in 88 of the 110 breast cancer cases (80.0%) under study, expect for decreased expression in a few cases. The immunoreactivity of aPK Clambda/iota was generally weak in ductal carcinoma in situ, but strong in invasive ductal carcinoma (IDC; P = .022). The correlation between apical or cytoplasmic aPKC lambda/iota localization and tumor pathologic type (ie, atypical ductal hyperplasia, ductal carcinoma in situ. or IDC) was also demonstrated (P < .001). These results thus indicate that the normal apicobasal polarity is lost upon the progression of a breast lesion to IDC. This is also the first evidence to show aPKC lambda/iota overexpression in breast cancer and demonstrates that its localization is associated with the trend of pathologic type of the tumor.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/enzymology
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/therapy
- Carcinoma, Intraductal, Noninfiltrating/enzymology
- Carcinoma, Intraductal, Noninfiltrating/secondary
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Cell Polarity
- Combined Modality Therapy
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Humans
- Immunoenzyme Techniques
- Isoenzymes/metabolism
- Middle Aged
- Neoplasm Staging
- Protein Kinase C/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Yasuyuki Kojima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Baldwin RM, Parolin DAE, Lorimer IAJ. Regulation of glioblastoma cell invasion by PKC iota and RhoB. Oncogene 2008; 27:3587-95. [PMID: 18212741 DOI: 10.1038/sj.onc.1211027] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glioblastoma multiforme is the most aggressive form of primary brain tumor and remains largely incurable, in large part, due to its highly invasive nature. The phosphoinositide (PI) 3-kinase pathway is often constitutively active in these tumors due to activating mutations in the epidermal growth factor receptor, or deletion/loss of function of the tumor suppressor PTEN. Protein kinase C type iota (PKC iota), a member of the atypical protein kinase C family, is activated by the PI 3-kinase pathway and is an important downstream mediator. Here, we have assessed the role of PKC iota in glioblastoma cell invasion. Depletion of PKC iota with RNA interference caused an increase in actin stress fibers and a decrease in cell motility and invasion. Gene expression microarray analysis of U87MG cells showed that PKC iota repressed expression of mRNA for RhoB, which has previously been shown to have a role in actin stress fiber formation. Western blot analysis showed that both PKC iota depletion and pharmacological inhibition of PKC iota caused an increase in the protein levels of RhoB, as did inhibition of PI 3-kinase. Expression of RhoB from a constitutive promoter caused changes in actin stress fibers and cell invasion that were similar to those seen with PKC iota depletion. These data show that PKC iota, activated as a consequence of aberrant upstream PI 3-kinase signaling, mediates glioblastoma cell motility and invasion, and that repression of RhoB is key downstream event in PKC iota signaling leading to enhanced cell motility. In addition, constitutive expression of RhoB repressed PKC iota activity, as assessed by its phosphorylation status on Thr555. PKC iota and RhoB are, therefore, mutually antagonistic, potentially creating a sensitive switch between invasive and non-invasive phenotypes.
Collapse
Affiliation(s)
- R M Baldwin
- Ottawa Health Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
45
|
Winbanks CE, Grimwood L, Gasser A, Darby IA, Hewitson TD, Becker GJ. Role of the phosphatidylinositol 3-kinase and mTOR pathways in the regulation of renal fibroblast function and differentiation. Int J Biochem Cell Biol 2007; 39:206-19. [PMID: 16973406 DOI: 10.1016/j.biocel.2006.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 07/31/2006] [Accepted: 08/08/2006] [Indexed: 11/19/2022]
Abstract
Tubulointerstitial fibrosis is largely mediated by (myo)fibroblasts present in the interstitium. In this study, we investigated the role of mTOR and phosphatidylinositol 3-kinase in the regulation of fibroblast kinetics, fibroblast differentiation, and collagen synthesis. Rat renal fibroblasts were propagated from kidneys 3 days post-ureteric obstruction and specific inhibitors of mTOR (RAD) and phosphatidylinositol 3-kinase (LY294002) were used to examine the regulation of fibrogenesis. LY294002 but not RAD completely inhibited phosphorylation of Akt, while both inhibitors decreased phosphorylation of the S6 ribosomal protein. RAD and LY decreased foetal calf serum stimulated proliferation and DNA synthesis. In addition to their individual effects, treatment with both RAD and LY294002 decreased serum-induced fibroblast proliferation and DNA synthesis significantly more than either drug alone. TUNEL positive cells (apoptosis) in RAD and LY294002 treated groups were not different from control groups. In addition to their effect on proliferation, both inhibitors also reduced total collagen synthesis. Differentiation studies indicated an increase in alpha-smooth muscle actin expression relative to beta-actin (western blotting), with cytochemistry confirming that all doses of RAD and LY294002 increased the proportion of alpha-smooth muscle actin positive cells, and hence myofibroblasts. Effects were independent of cell toxicity. These results highlight the potential significance of PI3K and mTOR, in the regulation of renal (myo)fibroblast activity. The synergistic effects of LY and RAD on proliferation suggest that mTOR signalling involves pathways other than phosphatidylinositol 3-kinase. These results provide a novel insight into the mechanisms of fibroblast regulation during fibrogenesis.
Collapse
Affiliation(s)
- Catherine E Winbanks
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Vic. 3050, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Merrick DT, Kittelson J, Winterhalder R, Kotantoulas G, Ingeberg S, Keith RL, Kennedy TC, Miller YE, Franklin WA, Hirsch FR. Analysis of c-ErbB1/epidermal growth factor receptor and c-ErbB2/HER-2 expression in bronchial dysplasia: evaluation of potential targets for chemoprevention of lung cancer. Clin Cancer Res 2006; 12:2281-8. [PMID: 16609045 DOI: 10.1158/1078-0432.ccr-05-2291] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Lung cancer is preceded by a premalignant phase during which intervention could decrease associated morbidity and mortality. Molecular characterization of factors involved in controlling progression of bronchial dysplasias will provide markers of premalignant change and identify targets for chemoprevention. EXPERIMENTAL DESIGN Immunohistochemical analysis of epidermal growth factor receptor (EGFR; c-ErbB1/EGFR), HER-2/neu (c-ErbB2/HER-2), Ki-67, and minichromosome maintenance protein 2 (MCM2) expression in bronchial dysplasia was undertaken to characterize molecular alterations associated with the progression of these lesions in 268 bronchoscopically obtained biopsies from 134 subjects. RESULTS Analysis of biopsies with the most severe diagnosis from each subject showed a linear relationship between increasing marker expression and severity of dysplastic change for EGFR (P < 0.001), Ki-67 (P < 0.001), and MCM2 (P = 0.001) but not HER-2 (P = 0.102). Increased expression of either EGFR or HER-2 was associated with increased levels of Ki-67 and MCM2 expression, and combined overexpression of these receptors was associated with the highest levels of proliferation, suggesting a synergistic effect. Finally, the lack of an associated trend toward increased EGFR expression when comparing the worst and best biopsies within each subject indicated a potential field effect in the induction of EGFR expression. CONCLUSIONS The results suggest a prominent role for EGFR overexpression in the development and progression of bronchial dysplasia and provide rationale for exploring inhibition of EGFR signaling in lung cancer chemoprevention.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biopsy
- Bronchial Neoplasms/genetics
- Bronchial Neoplasms/pathology
- Bronchial Neoplasms/prevention & control
- Cell Cycle Proteins/analysis
- Cell Cycle Proteins/drug effects
- Cell Cycle Proteins/genetics
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Chemoprevention
- Disease Progression
- ErbB Receptors/biosynthesis
- ErbB Receptors/drug effects
- ErbB Receptors/genetics
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Immunohistochemistry
- Ki-67 Antigen/analysis
- Ki-67 Antigen/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Male
- Middle Aged
- Minichromosome Maintenance Complex Component 2
- Nuclear Proteins/analysis
- Nuclear Proteins/drug effects
- Nuclear Proteins/genetics
- Precancerous Conditions/drug therapy
- Precancerous Conditions/genetics
- Precancerous Conditions/metabolism
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/drug effects
- Receptor, ErbB-2/genetics
- Sensitivity and Specificity
Collapse
Affiliation(s)
- Daniel T Merrick
- Department of Pathology, University of Colorado Cancer Center, Denver, Colorado, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Liao L, Zeng XM, Gao PJ, Zhu DL, Mei YA. PKC pathway associated with the expression of an A-type K+ channel induced by TGF-β1 in rat vascular myofibroblasts. Biochem Biophys Res Commun 2005; 336:854-9. [PMID: 16154536 DOI: 10.1016/j.bbrc.2005.08.180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Accepted: 08/23/2005] [Indexed: 01/14/2023]
Abstract
Our previous study indicated that TGF-beta1 induced the expression of a transient outward K+ channel (A-type) during the phenotypic transformation of vascular fibroblasts to myofibroblasts. Here, we studied the relevant signal transduction pathway using whole cell recording and a quantitative RT-PCR technique. Results indicate that the protein kinase C (PKC) agonist phorbol-12-myristate-13-acetate (PMA, 1 microM) could mimic the effect of TGF-beta1 (20 ng/ml) on the expression of an A-type K+ channel and induced a similar A-type K+ current. Moreover, a PKC inhibitor, bisindolylmaleimide I (1 microM), could abrogate the effect of TGF-beta1 on K(V)4.2 expression. This result suggests that a PKC pathway may be involved in the expression of an A-type K+ channel induced by TGF-beta1 in rat vascular myofibroblasts.
Collapse
Affiliation(s)
- Lei Liao
- Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | | | | | | | | |
Collapse
|
48
|
Hashimoto N, Kido Y, Uchida T, Matsuda T, Suzuki K, Inoue H, Matsumoto M, Ogawa W, Maeda S, Fujihara H, Ueta Y, Uchiyama Y, Akimoto K, Ohno S, Noda T, Kasuga M. PKClambda regulates glucose-induced insulin secretion through modulation of gene expression in pancreatic beta cells. J Clin Invest 2005; 115:138-45. [PMID: 15630453 PMCID: PMC539193 DOI: 10.1172/jci22232] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 10/26/2004] [Indexed: 12/15/2022] Open
Abstract
Altered regulation of insulin secretion by glucose is characteristic of individuals with type 2 diabetes mellitus, although the mechanisms that underlie this change remain unclear. We have now generated mice that lack the lambda isoform of PKC in pancreatic beta cells (betaPKClambda(-/-) mice) and show that these animals manifest impaired glucose tolerance and hypoinsulinemia. Furthermore, insulin secretion in response to high concentrations of glucose was impaired, whereas the basal rate of insulin release was increased, in islets isolated from betaPKClambda(-/-) mice. Neither the beta cell mass nor the islet insulin content of betaPKClambda(-/-) mice differed from that of control mice, however. The abundance of mRNAs for Glut2 and HNF3beta was reduced in islets of betaPKClambda(-/-) mice, and the expression of genes regulated by HNF3beta was also affected (that of Sur1 and Kir6.2 genes was reduced, whereas that of hexokinase 1 and hexokinase 2 genes was increased). Normalization of HNF3beta expression by infection of islets from betaPKClambda(-/-) mice with an adenoviral vector significantly reversed the defect in glucose-stimulated insulin secretion. These results indicate that PKClambda plays a prominent role in regulation of glucose-induced insulin secretion by modulating the expression of genes important for beta cell function.
Collapse
Affiliation(s)
- Naoko Hashimoto
- Department of Clinical Molecular Medicine, Division of Diabetes and Digestive and Kidney Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Plusa B, Frankenberg S, Chalmers A, Hadjantonakis AK, Moore CA, Papalopulu N, Papaioannou VE, Glover DM, Zernicka-Goetz M. Downregulation of Par3 and aPKC function directs cells towards the ICM in the preimplantation mouse embryo. J Cell Sci 2005; 118:505-15. [PMID: 15657073 DOI: 10.1242/jcs.01666] [Citation(s) in RCA: 204] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Generation of inside cells that develop into inner cell mass (ICM) and outside cells that develop into trophectoderm is central to the development of the early mouse embryo. Critical to this decision is the development of cell polarity and the associated asymmetric (differentiative) divisions of the 8-cell-stage blastomeres. The underlying molecular mechanisms for these events are not understood. As the Par3/aPKC complex has a role in establishing cellular polarity and division orientation in other systems, we explored its potential function in the developing mouse embryo. We show that both Par3 and aPKC adopt a polarized localization from the 8-cell stage onwards and that manipulating their function re-directs cell positioning and consequently influences cell fate. Injection of dsRNA against Par3 or mRNA for a dominant negative form of aPKC into a random blastomere at the 4-cell stage directs progeny of the injected cell into the inside part of the embryo. This appears to result from both an increased frequency by which such cells undertake differentiative divisions and their decreased probability of retaining outside positions. Thus, the natural spatial allocation of blastomere progeny can be over-ridden by downregulation of Par3 or aPKC, leading to a deceased tendency for them to remain outside and so develop into trophectoderm. In addition, this experimental approach illustrates a powerful means of manipulating gene expression in a specific clonal population of cells in the preimplantation embryo.
Collapse
Affiliation(s)
- Berenika Plusa
- Wellcome Trust and Cancer Research UK Gurdon Institute, Tennis Court Road, Cambridge CB2 1QR, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Harhaj NS, Antonetti DA. Regulation of tight junctions and loss of barrier function in pathophysiology. Int J Biochem Cell Biol 2004; 36:1206-37. [PMID: 15109567 DOI: 10.1016/j.biocel.2003.08.007] [Citation(s) in RCA: 404] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2003] [Accepted: 08/21/2003] [Indexed: 12/16/2022]
Abstract
The mechanism by which epithelial and endothelial cells interact to form polarized tissue is of fundamental importance to multicellular organisms. Dysregulation of these barriers occurs in a variety of diseases, destroying the normal cellular environments and leading to organ failure. Increased levels of growth factors are a common characteristic of diseases exhibiting tissue permeability, suggesting that growth factors play a direct role in elevating permeability. Of particular concern for this laboratory, increased expression of vascular endothelial growth factor may enhance vascular permeability in diabetic retinopathy, leading to vision impairment and blindness. However, the mechanism by which growth factors increase permeability is unclear. Polarized cells form strong barriers through the development of tight junctions, which are specialized regions of the junctional complex. Tight junctions are composed of three types of transmembrane proteins, a number of peripheral membrane structural proteins, and are associated with a variety of regulatory proteins. Recent data suggest that growth factor-stimulated alterations in tight junctions contribute to permeability in a variety of disease states. The goal of this review was to elucidate potential mechanisms by which elevated growth factors elicit deregulated paracellular permeability via altered regulation of tight junctions, with particular emphasis on the tight junction proteins occludin and ZO-1, protein kinase C signaling, and endocytosis of junctional proteins. Understanding the molecular mechanisms underlying growth factor-mediated regulation of tight junctions will facilitate the development of novel treatments for diseases such as brain tumors, diabetic retinopathy and other diseases with compromised tight junction barriers.
Collapse
Affiliation(s)
- Nicole S Harhaj
- Penn State Retina Research Group, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|