1
|
Christoforou M, Charalambous A, Sfakianakis D, Skourides PA. Targeting the hydrophobic pockets of FAK/PYK2 FAT domain: a highly effective inhibitory strategy suppressing tumor growth and eliminating metastasis. Cell Commun Signal 2025; 23:231. [PMID: 40390029 PMCID: PMC12087225 DOI: 10.1186/s12964-025-02203-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 04/15/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND FAK is a non-receptor tyrosine kinase and an adaptor protein commonly overexpressed in cancer. It regulates multiple tumorigenic pathways through both kinase-dependent and kinase-independent scaffolding functions and thus represents a promising therapeutic target for various cancers. Several FAK kinase inhibitors shown to be effective in preclinical studies advanced to clinical trials, however none produced objective clinical responses. These results are in part attributed to drug resistance and the inability to simultaneously target kinase-dependent and kinase-independent functions of the protein, both of which have been shown to promote tumorigenesis. This has led to the development of scaffold inhibitors that could be used as adjuvants, none of which have so far reached the clinical stage. Importantly, FAK's closely related paralogue, PYK2, compensates for the loss of FAK thus it is also important to target both kinases. In the present study, we evaluate a novel strategy for the inhibition of kinase-dependent and kinase-independent functions of FAK and PYK2 through the expression of the FAT HP-site-specific LD2-LD4 peptide that leads to their displacement from focal adhesions. METHODS The impact of LD2-LD4 expression on FAK and PYK2 was assessed through co-immunoprecipitation experiments, Western Blot analysis and quantitative immunofluorescence. In vitro investigation of the effects of LD2-LD4 expression on tumor cell migration and proliferation was carried out using 2D migration, 3D invasion and proliferation assays. The preclinical experiments of this study were carried out using an orthotopic xenograft model, followed by immunohistochemical analysis. RESULTS We show that LD2-LD4 expression leads to the displacement of FAK and PYK2 from focal adhesions, blocking both enzymatic and non-enzymatic activities. It also dramatically inhibits 2D cell migration, as well as invasion in vitro. Importantly, LD2-LD4 exerts promising anti-tumor effects and nearly abolishes the appearance of metastatic foci. Finally, we show that an LD monomer can also displace both FAK and PYK2 from FAs suggesting that organic molecules with high affinity for the FAT HPs could mimic the LD2-LD4 activity. CONCLUSIONS Targeting the FAT domain hydrophobic patches of FAK/PYK2 is a highly effective inhibitory strategy that can overcome the limitations of existing ATP competitive inhibitors and lead to the development of novel inhibitors with strong antitumor and antimetastatic activity.
Collapse
Affiliation(s)
- Maria Christoforou
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 1678, Nicosia, Cyprus
| | - Anna Charalambous
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 1678, Nicosia, Cyprus.
| | - Dimitrios Sfakianakis
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 1678, Nicosia, Cyprus
| | | |
Collapse
|
2
|
Lopes AL, Paulino AC, Thaumaturgo MAS, Araújo WM, Caloba P, Kawanishi K, Willert K, De Oliveira RP, Machado JC, Lemos F, Varki N, Morgado-Diaz JA, Läubli H, Todeschini AR, Varki A, Alisson-Silva F. Dietary intake of the red meat-derived glycan Neu5Gc fuels colorectal cancer through up-regulation of Wnt signaling pathway. Cancer Lett 2025; 616:217598. [PMID: 40023392 DOI: 10.1016/j.canlet.2025.217598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Colorectal cancer (CRC) is a significant health concern, often linked to western diets, particularly red meat consumption. Several mechanisms, such as the high heme iron content, the formation of N-nitroso compounds (NOCs), heterocyclic amines (HCAs), and polycyclic aromatic hydrocarbons (PAHs), have been suggested to explain red meat's cancer-promoting effects. However, these factors are also found in fish and poultry, which are not linked to CRC risk. A new hypothesis attributes red meat's impact on CRC to its high content of a nonhuman glycan. While most mammals express N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), humans express only Neu5Ac due to the loss of the CMAH enzyme. A red meat-rich diet leads to the incorporation of Neu5Gc into human cells, triggering an antibody-mediated inflammatory process known as xenosialitis. This study shows that Neu5Gc incorporation into CRC cells activates the Wnt/β-catenin signaling pathway, promoting cell proliferation. In a murine model lacking CMAH, a Neu5Gc-enriched diet induced intestinal polyp growth, with more malignant characteristics. Additionally, Neu5Gc incorporation in intestinal cells increased the expression of genes downstream of Wnt signaling. These findings reveal, for the first time in an in vivo model, a mechanism independent of immune response, where red meat consumption accelerates tumor progression through Neu5Gc incorporation. This activation of the Wnt/β-catenin signaling pathway provides new insight into how red meat consumption may influence CRC progression.
Collapse
Affiliation(s)
- Ana Luiza Lopes
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Amanda Carlos Paulino
- Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Mariana A S Thaumaturgo
- Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | | | - Philippe Caloba
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Kunio Kawanishi
- Department of Anatomy, Showa Medical University, Tokyo, Japan
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California San Diego, UCSD, USA
| | | | - João C Machado
- Biomedical Engineering Program, COPPE/UFRJ Brazil, Brazil; Post-Graduation Program on Surgical Sciences, School of Medicine/UFRJ, Brazil
| | - Felipe Lemos
- Immunopharmacology Laboratory, Fiocruz Brazil, Brazil
| | - Nissi Varki
- Departments of Medicine and Cellular & Molecular Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Glycobiology Research and Training Center (GRTC), University of California San Diego, UCSD, USA
| | | | - Heinz Läubli
- Division of Oncology and Department of Biomedicine, University Hospital and University of Basel, 4031, Basel, Switzerland
| | | | - Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Glycobiology Research and Training Center (GRTC), University of California San Diego, UCSD, USA
| | - Frederico Alisson-Silva
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil.
| |
Collapse
|
3
|
Chocarro-Calvo A, Jociles-Ortega M, García-Martinez JM, Louphrasitthiphol P, Carvalho-Marques S, Vivas-García Y, Ramírez-Sánchez A, Chauhan J, Fiuza MC, Duran M, Sánchez-Danés A, Goding CR, García-Jiménez C. Fatty acid uptake activates an AXL-CAV1-β-catenin axis to drive melanoma progression. Genes Dev 2025; 39:463-489. [PMID: 40015991 PMCID: PMC11960706 DOI: 10.1101/gad.351985.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Abstract
Interaction between the tumor microenvironment and cancer cell plasticity drives intratumor phenotypic heterogeneity and underpins disease progression and nongenetic therapy resistance. Phenotype-specific expression of the AXL receptor tyrosine kinase is a pivotal player in dormancy, invasion, and resistance to treatment. However, although the AXL ligand GAS6 is present within tumors, how AXL is activated in metastasizing cells remains unclear. Here, using melanoma as a model, we reveal that AXL is activated by exposure to human adipocytes and to oleic acid, a monounsaturated fatty acid abundant in lymph and in adipocytes. AXL activation triggers SRC-dependent formation and nuclear translocation of a β-catenin-CAV1 complex required for melanoma invasiveness. Remarkably, only undifferentiated AXLHigh melanoma cells engage in symbiosis with human adipocytes, in part by triggering WNT5a-mediated lipolysis, leading to AXL-dependent, but FATP-independent, fatty acid uptake and nuclear localization of the β-catenin-CAV1 complex. Significantly, human melanomas in the vicinity of adipocytes exhibit high levels of nuclear CAV1. The results unveil an AXL- and CAV1-dependent mechanism through which a nutritional input drives phenotype-specific activation of a prometastasis program. Given the key role of AXL in a broad range of cancers, the results offer major insights into the mechanisms of cancer cell dormancy and therapy resistance.
Collapse
Affiliation(s)
- Ana Chocarro-Calvo
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
- Area of Physiology, Faculty Health Sciences, University Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Miguel Jociles-Ortega
- Area of Physiology, Faculty Health Sciences, University Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | | | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | | | - Yurena Vivas-García
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Ana Ramírez-Sánchez
- Area of Physiology, Faculty Health Sciences, University Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - M Carmen Fiuza
- Department of Surgery, University Hospital Fundación Alcorcón, Alcorcón, Madrid 28922, Spain
| | - Manuel Duran
- Department of General Surgery, University Hospital Rey Juan Carlos, Móstoles, Madrid 28933, Spain
| | - Adriana Sánchez-Danés
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom;
| | - Custodia García-Jiménez
- Area of Physiology, Faculty Health Sciences, University Rey Juan Carlos, Alcorcón, Madrid 28922, Spain;
| |
Collapse
|
4
|
Liu Y, Nishiura M, Fujii M, Sandhu S, Yawaka Y, Yamazaki Y, Hasebe A, Iimura T, Kong SW, Lee JW. Selective Pyk2 inhibition enhances bone restoration through SCARA5-mediated bone marrow remodeling in ovariectomized mice. Cell Commun Signal 2024; 22:561. [PMID: 39578816 PMCID: PMC11583405 DOI: 10.1186/s12964-024-01945-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
Understanding the intricate cellular interactions involved in bone restoration is crucial for developing effective strategies to promote bone healing and mitigate conditions such as osteoporosis and fractures. Here, we provide compelling evidence supporting the anabolic effects of a pharmacological Pyk2 inhibitor (Pyk2-Inh) in promoting bone restoration. In vitro, Pyk2 signaling inhibition markedly enhances alkaline phosphatase (ALP) activity, a hallmark of osteoblast differentiation, through activation of canonical Wnt/β-catenin signaling. Notably, analysis of human mesenchymal stem cells through RNA-seq revealed a novel candidate, SCARA5, identified through Pyk2-Inh treatment. We demonstrate that Scara5 plays a crucial role in suppressing the differentiation from stromal cells into adipocytes, and accelerates lineage commitment to osteoblasts, establishing Scara5 as a negative regulator of bone formation. Additionally, Pyk2 inhibition significantly impedes osteoclast differentiation and bone resorption. In a co-culture system comprising osteoblasts and osteoclasts, Pyk2-Inh effectively suppressed osteoclast differentiation, accompanied by a substantial increase in the transcriptional expression of Tnfrsf11b and Csf1 in osteoblasts, highlighting a dual regulatory role in osteoblast-osteoclast crosstalk. In an ovariectomized mouse model of osteoporosis, oral administration of Pyk2-Inh significantly increased bone mass by simultaneously reducing bone resorption, promoting bone formation and decreasing bone marrow fat. These results suggest Pyk2 as a potential therapeutic target for both adipogenesis and osteogenesis in bone marrow. Our findings underscore the importance of Pyk2 signaling inhibition as a key regulator of bone remodeling, offering promising prospects for the development of novel osteoporosis therapies.
Collapse
Affiliation(s)
- Yunqing Liu
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
- Department of Stomatology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
- Dentistry for Children and Disabled Persons, Department of Oral Functional Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Mika Fujii
- Department of Oral Health Science, Gerodontology, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Sumiti Sandhu
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, MA, 02215, USA
| | - Yasutaka Yawaka
- Dentistry for Children and Disabled Persons, Department of Oral Functional Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Yutaka Yamazaki
- Department of Oral Health Science, Gerodontology, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Akira Hasebe
- Department of Oral Pathobiological Science, Microbiology, Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Sek Won Kong
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, MA, 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ji-Won Lee
- Department of Oral Pathobiological Science, Microbiology, Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan.
| |
Collapse
|
5
|
Wu F, Zhang K, Song Z, Zhou Q, Sun H, Tan Z, Huang Z, Wang F, Wang Z, Yang R, Huang Y. Reduced Proline-Rich Tyrosine Kinase 2 Promotes Tumor Metastasis by Activating Epithelial-Mesenchymal Transition in Colorectal Cancer. Dig Dis Sci 2024; 69:4098-4107. [PMID: 39414740 DOI: 10.1007/s10620-024-08643-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/05/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND Proline-rich tyrosine kinase 2 (PYK2) is involved in the occurrence, proliferation, migration, and invasion of various tumors. However, few studies have reported the role of PYK2 in colorectal cancer (CRC). AIM To explore the effects of PYK2 on CRC metastasis and elucidate the detailed molecular mechanisms involved. METHODS The expression and prognosis value of PYK2 in CRC prognosis were analyzed using data from The Cancer Genome Atlas (TCGA). PYK2 was knocked down or overexpressed in human CRC cell line, HCT116. Cell proliferation, migration, invasion, and cycle changes were analyzed using CCK-8, Transwell, and flow cytometry assays. Western blotting and quantitative real-time PCR were performed to detect the mRNA and protein levels of cell proliferation and epithelial-mesenchymal transition (EMT) indicators. Fluorescence staining was performed to examine the cytoskeleton. RESULTS Lower expression of PYK2 was observed in CRC tissues and associated with poor prognosis and metastasis in patients with CRC in TCGA database. PYK2 knockdown significantly induced the migration and invasion of CRC cells but did not affect cell proliferation or cycle. Immunofluorescence staining of phalloidin showed that the downregulation of PYK2 increased the cytoskeleton in CRC cells. Moreover, low expression of PYK2 induced the downregulation of E-cadherin and upregulation of snail and vimentin by activating Wnt/β-catenin signaling, thus promoting EMT in CRC cells. CONCLUSIONS Low PYK2 expression was found in tumor tissues, especially metastases, and significantly correlated with patient prognosis. Moreover, decreased PYK2 induces EMT by activating Wnt/β-catenin signaling, which is the potential mechanism of CRC metastasis. Regulating the expression of PYK2 to suppress tumor cell metastasis may represent a promising therapeutic strategy for metastatic CRC.
Collapse
Affiliation(s)
- Fangquan Wu
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ke Zhang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhengyang Song
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qishuo Zhou
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hongxia Sun
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zenglin Tan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhenxuan Huang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhonglin Wang
- Department of Anorectal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Riwei Yang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yingpeng Huang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
6
|
Geijerman E, Terrana F, Peters GJ, Deng D, Diana P, Giovannetti E, Xu G. Targeting a key FAK-tor: the therapeutic potential of combining focal adhesion kinase (FAK) inhibitors and chemotherapy for chemoresistant non-small cell lung cancer. Expert Opin Investig Drugs 2024; 33:1103-1118. [PMID: 39435477 DOI: 10.1080/13543784.2024.2417762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION NSCLC is the leading cause of cancer-related deaths globally, with a low survival rate primarily due to NSCLC frequently becoming chemoresistant. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase involved in pathways regulating multiple processes in the cell, including survival, migration, and the TME, that contribute to both tumor progression and drug resistance. Recently, FAK inhibitors (FAKi) have shown promising potential for the treatment of NSCLC. AREAS COVERED This narrative review aims to summarize key signaling pathways involving FAK that contribute to tumor progression and drug resistance. It will further provide an overview of FAKi currently in pre- and early-phase clinical trials for solid tumors, as well as the therapeutic potential of combining FAKi with chemotherapy, as this has emerged as a promising strategy to overcome chemoresistance in NSCLC. EXPERT OPINION It is becoming increasingly clear that FAK is not an oncogenic driver but rather contributes to tumor progression and drug resistance. Hence, while FAKi have only demonstrated modest results in clinical trials when given by themselves, treatment regimens combining other therapies with FAKi have shown promising potential to overcome drug resistance. Lastly, of particular novelty are FAK-PROTACs (proteolysis-targeting chimaeras), which uniquely target both cytosolic and nuclear FAK.
Collapse
Affiliation(s)
- Emma Geijerman
- Amsterdam University College, Amsterdam, The Netherlands
| | - Francesca Terrana
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| | - Geng Xu
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Stoyell-Conti FF, Suresh Kumar M, Zigmond ZM, Rojas MG, Santos Falcon N, Martinez L, Vazquez-Padron RI. Gene inactivation of lysyl oxidase in smooth muscle cells reduces atherosclerosis burden and plaque calcification in hyperlipidemic mice. Atherosclerosis 2024; 397:118582. [PMID: 39260002 PMCID: PMC11465391 DOI: 10.1016/j.atherosclerosis.2024.118582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS Lysyl oxidase (LOX) catalyzes the crosslinking of collagen and elastin to maintain tensile strength and structural integrity of the vasculature. Excessive LOX activity increases vascular stiffness and the severity of occlusive diseases. Herein, we investigated the mechanisms by which LOX controls atherogenesis and osteogenic differentiation of vascular smooth muscle cells (SMC) in hyperlipidemic mice. METHODS Gene inactivation of Lox in SMC was achieved in conditional knockout mice after tamoxifen injections. Atherosclerosis burden and vascular calcification were assessed in hyperlipidemic conditional [Loxf/fMyh11-CreERT2ApoE-/-] and sibling control mice [Loxwt/wtMyh11-CreERT2ApoE-/-]. Mechanistic studies were performed with primary aortic SMC from Lox mutant and wild type mice. RESULTS Inactivation of Lox in SMCs decreased > 70 % its RNA expression and protein level in the aortic wall and significantly reduced LOX activity without compromising vascular structure and function. Moreover, LOX deficiency protected mice against atherosclerotic burden (13 ± 2 versus 23 ± 1 %, p < 0.01) and plaque calcification (5 ± 0.4 versus 11.8 ± 3 %, p < 0.05) compared to sibling controls. Interestingly, gene inactivation of Lox in SMCs preserved the contractile phenotype of vascular SMC under hyperlipidemic conditions as demonstrated by single-cell RNA sequencing and immunofluorescence. Mechanistically, the absence of LOX in SMC prevented excessive collagen crosslinking and the subsequent activation of the pro-osteogenic FAK/β-catenin signaling axis. CONCLUSIONS Lox inactivation in SMC protects mice against atherosclerosis and plaque calcification by reducing SMC modulation and FAK/β-catenin signaling.
Collapse
MESH Headings
- Animals
- Protein-Lysine 6-Oxidase/metabolism
- Protein-Lysine 6-Oxidase/genetics
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/enzymology
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/pathology
- Vascular Calcification/enzymology
- Vascular Calcification/prevention & control
- Vascular Calcification/metabolism
- Plaque, Atherosclerotic
- Hyperlipidemias/genetics
- Hyperlipidemias/enzymology
- Hyperlipidemias/complications
- Hyperlipidemias/metabolism
- Disease Models, Animal
- Mice, Knockout
- Mice
- Osteogenesis
- Cells, Cultured
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/enzymology
- Aortic Diseases/prevention & control
- Aortic Diseases/metabolism
- Aorta/pathology
- Aorta/enzymology
- Aorta/metabolism
- Male
- Mice, Inbred C57BL
- beta Catenin/metabolism
- Signal Transduction
- Extracellular Matrix Proteins
Collapse
Affiliation(s)
- Filipe F Stoyell-Conti
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maya Suresh Kumar
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Miguel G Rojas
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nieves Santos Falcon
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, USA.
| |
Collapse
|
8
|
Schlaepfer DD, Ojalill M, Stupack DG. Focal adhesion kinase signaling - tumor vulnerabilities and clinical opportunities. J Cell Sci 2024; 137:jcs261723. [PMID: 39034922 PMCID: PMC11298715 DOI: 10.1242/jcs.261723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Focal adhesion kinase (FAK; encoded by PTK2) was discovered over 30 years ago as a cytoplasmic protein tyrosine kinase that is localized to cell adhesion sites, where it is activated by integrin receptor binding to extracellular matrix proteins. FAK is ubiquitously expressed and functions as a signaling scaffold for a variety of proteins at adhesions and in the cell cytoplasm, and with transcription factors in the nucleus. FAK expression and intrinsic activity are essential for mouse development, with molecular connections to cell motility, cell survival and gene expression. Notably, elevated FAK tyrosine phosphorylation is common in tumors, including pancreatic and ovarian cancers, where it is associated with decreased survival. Small molecule and orally available FAK inhibitors show on-target inhibition in tumor and stromal cells with effects on chemotherapy resistance, stromal fibrosis and tumor microenvironment immune function. Herein, we discuss recent insights regarding mechanisms of FAK activation and signaling, its roles as a cytoplasmic and nuclear scaffold, and the tumor-intrinsic and -extrinsic effects of FAK inhibitors. We also discuss results from ongoing and advanced clinical trials targeting FAK in low- and high-grade serous ovarian cancers, where FAK acts as a master regulator of drug resistance. Although FAK is not known to be mutationally activated, preventing FAK activity has revealed multiple tumor vulnerabilities that support expanding clinical combinatorial targeting possibilities.
Collapse
Affiliation(s)
- David D. Schlaepfer
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, Moores Cancer Center, Division of Gynecologic Oncology, 3855 Health Sciences Dr., La Jolla, CA 92098, USA
| | - Marjaana Ojalill
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, Moores Cancer Center, Division of Gynecologic Oncology, 3855 Health Sciences Dr., La Jolla, CA 92098, USA
| | - Dwayne G. Stupack
- University of California, San Diego, Department of Obstetrics, Gynecology, and Reproductive Sciences, Moores Cancer Center, Division of Gynecologic Oncology, 3855 Health Sciences Dr., La Jolla, CA 92098, USA
| |
Collapse
|
9
|
Hu B, Chen S. The role of UBR5 in tumor proliferation and oncotherapy. Gene 2024; 906:148258. [PMID: 38331119 DOI: 10.1016/j.gene.2024.148258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Ubiquitin (Ub) protein ligase E3 component n-recognin 5 (UBR5), as a crucial Ub ligase, plays a pivotal role in the field of cell biology, attracting significant attention for its functions in regulating protein degradation and signaling pathways. This review delves into the fundamental characteristics and structure of UBR5. UBR5, through ubiquitination, regulates various key proteins, directly or indirectly participating in cell cycle control, thereby exerting a direct impact on the proliferation of tumor cells. Meanwhile, we comprehensively review the expression levels of UBR5 in different types of tumors and its relationship with tumor development, providing key clues for the role of UBR5 in cancer. Furthermore, we summarize the current research status of UBR5 in cancer treatment. Through literature review, we find that UBR5 may play a crucial role in the sensitivity of tumor cells to radiotherapy chemotherapy, and other anti-tumor treatment, providing new insights for optimizing cancer treatment strategies. Finally, we discuss the challenges faced by UBR5 in cancer treatment, and looks forward to the future research directions. With the continuous breakthroughs in technology and in-depth research, we hope to further study the biological functions of UBR5 and lay the foundation for its anti-tumor treatment.
Collapse
Affiliation(s)
- Bin Hu
- Department of Geriatrics, Beilun District People's Hospital, Ningbo 315800, China
| | - Shiyuan Chen
- Department of Geriatrics, Beilun District People's Hospital, Ningbo 315800, China.
| |
Collapse
|
10
|
Dahiya S, Saleh M, Rodriguez UA, Rajasundaram D, R Arbujas J, Hajihassani A, Yang K, Sehrawat A, Kalsi R, Yoshida S, Prasadan K, Lickert H, Hu J, Piganelli JD, Gittes GK, Esni F. Acinar to β-like cell conversion through inhibition of focal adhesion kinase. Nat Commun 2024; 15:3740. [PMID: 38702347 PMCID: PMC11068907 DOI: 10.1038/s41467-024-47972-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
Insufficient functional β-cell mass causes diabetes; however, an effective cell replacement therapy for curing diabetes is currently not available. Reprogramming of acinar cells toward functional insulin-producing cells would offer an abundant and autologous source of insulin-producing cells. Our lineage tracing studies along with transcriptomic characterization demonstrate that treatment of adult mice with a small molecule that specifically inhibits kinase activity of focal adhesion kinase results in trans-differentiation of a subset of peri-islet acinar cells into insulin producing β-like cells. The acinar-derived insulin-producing cells infiltrate the pre-existing endocrine islets, partially restore β-cell mass, and significantly improve glucose homeostasis in diabetic mice. These findings provide evidence that inhibition of the kinase activity of focal adhesion kinase can convert acinar cells into insulin-producing cells and could offer a promising strategy for treating diabetes.
Collapse
Affiliation(s)
- Shakti Dahiya
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Mohamed Saleh
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Uylissa A Rodriguez
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jorge R Arbujas
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Arian Hajihassani
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Kaiyuan Yang
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anuradha Sehrawat
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ranjeet Kalsi
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Shiho Yoshida
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Krishna Prasadan
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Jing Hu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jon D Piganelli
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - George K Gittes
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- School of Medicine, Technical University of Munich, Munich, Germany.
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Fang L, Tao Y, Che G, Yun Y, Ren M, Liu Y. WSB1, as an E3 ligase, restrains myocardial ischemia-reperfusion injury by activating β-catenin signaling via promoting GSK3β ubiquitination. Mol Med 2024; 30:31. [PMID: 38395742 PMCID: PMC10893653 DOI: 10.1186/s10020-024-00800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Reperfusion is the most effective strategy for myocardial infarct, but induces additional injury. WD repeat and SOCS box containing protein 1 (WSB1) plays a protective role in ischemic cells. This study aims to investigate the effects of WSB1 on myocardial ischemia-reperfusion (IR) injury. METHODS The myocardial IR was induced by left anterior descending (LAD) ligation for 45 min and subsequent reperfusion. The overexpression of WSB1 was mediated by tail vein injection of AAV9 loaded with WSB1 encoding sequence two weeks before IR surgery. H9c2 myocardial cells underwent oxygen-sugar deprivation/reperfusion (OGD/R) to mimic IR, and transfected with WSB1 overexpression or silencing plasmid to alter the expression of WSB1. RESULTS WSB1 was found highly expressed in penumbra of myocardial IR rats, and the WSB1 overexpression relieved IR-induced cardio dysfunction, myocardial infarct and pathological damage, and cardiomyocyte death in penumbra. The ectopic expression of WSB1 in H9c2 myocardial cells mitigated OGD/R-caused apoptosis, and silencing of WSB1 exacerbated the apoptosis. In addition, WSB1 activated β-catenin signaling, which was deactivated under the ischemic condition. The co-immunoprecipitation results revealed that WSB1 mediated ubiquitination and degradation of glycogen synthase kinase 3 beta (GSK3β) as an E3 ligase in myocardial cells. The effects of WSB1 on myocardial cells under ischemic conditions were abolished by an inhibitor of β-catenin signaling. CONCLUSION WSB1 activated β-catenin pathway by promoting the ubiquitination of GSK3β, and restrained IR-induced myocardial injury. These findings might provide novel insights for clinical treatment of myocardial ischemic patients.
Collapse
Affiliation(s)
- Lini Fang
- Department of Function, Sanya Central Hospital (Hainan Third People's Hospital), 1154# Jiefang Fourth Road, Sanya, Hainan Province, China
| | - Yang Tao
- Department of Function, Sanya Central Hospital (Hainan Third People's Hospital), 1154# Jiefang Fourth Road, Sanya, Hainan Province, China
| | - Guoying Che
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yongzi Yun
- Department of Function, Sanya Central Hospital (Hainan Third People's Hospital), 1154# Jiefang Fourth Road, Sanya, Hainan Province, China
| | - Min Ren
- Ultrasound Department, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 536# Changle Road, Shanghai, China.
| | - Yujie Liu
- Department of Function, Sanya Central Hospital (Hainan Third People's Hospital), 1154# Jiefang Fourth Road, Sanya, Hainan Province, China.
| |
Collapse
|
12
|
Chocarro-Calvo A, Jociles-Ortega M, García-Martinez JM, Louphrasitthiphol P, Garcia YV, Ramírez-Sánchez A, Chauhan J, Fiuza MC, Duran M, García-Jiménez C, Goding CR. Phenotype-specific melanoma uptake of fatty acid from human adipocytes activates AXL and CAV1-dependent β-catenin nuclear accumulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576568. [PMID: 38328032 PMCID: PMC10849526 DOI: 10.1101/2024.01.21.576568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Phenotypic diversity of cancer cells within tumors generated through bi-directional interactions with the tumor microenvironment has emerged as a major driver of disease progression and therapy resistance. Nutrient availability plays a critical role in determining phenotype, but whether specific nutrients elicit different responses on distinct phenotypes is poorly understood. Here we show, using melanoma as a model, that only MITF Low undifferentiated cells, but not MITF High cells, are competent to drive lipolysis in human adipocytes. In contrast to MITF High melanomas, adipocyte-derived free fatty acids are taken up by undifferentiated MITF Low cells via a fatty acid transporter (FATP)-independent mechanism. Importantly, oleic acid (OA), a monounsaturated long chain fatty acid abundant in adipose tissue and lymph, reprograms MITF Low undifferentiated melanoma cells to a highly invasive state by ligand-independent activation of AXL, a receptor tyrosine kinase associated with therapy resistance in a wide range of cancers. AXL activation by OA then drives SRC-dependent formation and nuclear translocation of a β-catenin-CAV1 complex. The results highlight how a specific nutritional input drives phenotype-specific activation of a pro-metastasis program with implications for FATP-targeted therapies.
Collapse
|
13
|
Mubtasim N, Gollahon L. The Effect of Adipocyte-Secreted Factors in Activating Focal Adhesion Kinase-Mediated Cell Signaling Pathway towards Metastasis in Breast Cancer Cells. Int J Mol Sci 2023; 24:16605. [PMID: 38068928 PMCID: PMC10706115 DOI: 10.3390/ijms242316605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Obesity-associated perturbations in the normal secretion of adipocytokines from white adipocytes can drive the metastatic progression of cancer. However, the association between obesity-induced changes in secretory factors of white adipocytes and subsequent transactivation of the downstream effector proteins impacting metastasis in breast cancer cells remains unclear. Focal adhesion kinase, a cytoplasmic signal transducer, regulates the biological phenomenon of metastasis by activating downstream targets such as beta-catenin and MMP9. Thus, the possible role of phosphorylated FAK in potentiating cancer cell migration was investigated. To elucidate this potential relationship, MCF7 (ER+), MDA-MB-231 (Triple Negative) breast cancer cells, and MCF-10A non-tumorigenic breast cells were exposed to in vitro murine adipocyte-conditioned medium derived from 3T3-L1 MBX cells differentiated to obesity with fatty acid supplementation. Our results show that the conditioned medium derived from these obese adipocytes enhanced motility and invasiveness of breast cancer cells. Importantly, no such changes were observed in the non-tumorigenic breast cells. Our results also show that increased FAK autophosphorylation was followed by increased expression of beta-catenin and MMP9 in the breast cancer cells when exposed to obese adipocyte-conditioned medium, but not in the MCF10A cells. These results indicate that adipocyte-derived secretory factors induced FAK activation through phosphorylation. This in turn increased breast cancer cell migration and invasion by activating its downstream effector proteins beta-catenin and MMP9.
Collapse
Affiliation(s)
- Noshin Mubtasim
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
14
|
Wu YB, Li SY, Liu JY, Xue JJ, Xu JF, Chen T, Cao TY, Zhou H, Wu TT, Dong CL, Qian WF, Qiao LW, Hou SY, Wang T, Shen C. Long non-coding RNA NRSN2-AS1 promotes ovarian cancer progression through targeting PTK2/β-catenin pathway. Cell Death Dis 2023; 14:696. [PMID: 37875515 PMCID: PMC10598275 DOI: 10.1038/s41419-023-06214-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/17/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023]
Abstract
As a common malignant tumor among women, ovarian cancer poses a serious threat to their health. This study demonstrates that long non-coding RNA NRSN2-AS1 is over-expressed in ovarian cancer tissues using patient sample and tissue microarrays. In addition, NRSN2-AS1 is shown to promote ovarian cancer cell proliferation and metastasis both in vitro and in vivo. Mechanistically, NRSN2-AS1 stabilizes protein tyrosine kinase 2 (PTK2) to activate the β-catenin pathway via repressing MG-53-mediated ubiquitinated degradation of PTK2, thereby facilitating ovarian cancer progression. Rescue experiments verify the function of the NRSN2-AS1/PTK2/β-catenin axis and the effects of MG53 on this axis in ovarian cancer cells. In conclusion, this study demonstrates the key role of the NRSN2-AS1/PTK2/β-catenin axis for the first time and explores its potential clinical applications in ovarian cancer.
Collapse
Affiliation(s)
- Yi-Bo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Shen-Yi Li
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Department of Obstetrics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Jin-Yan Liu
- Department of Breast and Thyroid Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Jia-Jia Xue
- Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, 215124, China
| | - Jin-Fu Xu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Ting Chen
- Department of Gynaecology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Tian-Yue Cao
- Department of Gynaecology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Hui Zhou
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Tian-Tian Wu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Chun-Lin Dong
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Wei-Feng Qian
- Department of Breast and Thyroid Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Long-Wei Qiao
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| | - Shun-Yu Hou
- Department of Gynaecology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| | - Ting Wang
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
15
|
Tejeda-Muñoz N, Azbazdar Y, Monka J, Binder G, Dayrit A, Ayala R, O’Brien N, De Robertis EM. The PMA Phorbol Ester Tumor Promoter Increases Canonical Wnt Signaling Via Macropinocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543509. [PMID: 37333286 PMCID: PMC10274750 DOI: 10.1101/2023.06.02.543509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Activation of the Wnt pathway lies at the core of many human cancers. Wnt and macropinocytosis are often active in the same processes, and understanding how Wnt signaling and membrane trafficking cooperate should improve our understanding of embryonic development and cancer. Here we show that a macropinocytosis activator, the tumor promoter Phorbol 12-myristate 13-acetate (PMA), enhances Wnt signaling. Experiments using the Xenopus embryo as an in vivo model showed marked cooperation between the PMA phorbol ester and Wnt signaling, which was blocked by inhibitors of macropinocytosis, Rac1 activity, and lysosome acidification. Human colorectal cancer tissue arrays and xenografts in mice showed a correlation of cancer progression with increased macropinocytosis/multivesicular body/lysosome markers and decreased GSK3 levels. The crosstalk between canonical Wnt, focal adhesions, lysosomes, and macropinocytosis suggests possible therapeutic targets for cancer progression in Wnt-driven cancers.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
- Department of Oncology Science, Health Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- These authors contributed equally
| | - Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
- These authors contributed equally
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Grace Binder
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Alex Dayrit
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Raul Ayala
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Neil O’Brien
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Edward M. De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| |
Collapse
|
16
|
Kim DJ, Yi YW, Seong YS. Beta-Transducin Repeats-Containing Proteins as an Anticancer Target. Cancers (Basel) 2023; 15:4248. [PMID: 37686524 PMCID: PMC10487276 DOI: 10.3390/cancers15174248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Beta-transducin repeat-containing proteins (β-TrCPs) are E3-ubiquitin-ligase-recognizing substrates and regulate proteasomal degradation. The degradation of β-TrCPs' substrates is tightly controlled by various external and internal signaling and confers diverse cellular processes, including cell cycle progression, apoptosis, and DNA damage response. In addition, β-TrCPs function to regulate transcriptional activity and stabilize a set of substrates by distinct mechanisms. Despite the association of β-TrCPs with tumorigenesis and tumor progression, studies on the mechanisms of the regulation of β-TrCPs' activity have been limited. In this review, we studied publications on the regulation of β-TrCPs themselves and analyzed the knowledge gaps to understand and modulate β-TrCPs' activity in the future.
Collapse
Affiliation(s)
- Dong Joon Kim
- Department of Microbiology, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea;
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou 450008, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China
| | - Yong Weon Yi
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Yeon-Sun Seong
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan-si 31116, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
17
|
Qin R, Huang Y, Yao Y, Wang L, Zhang Z, Huang W, Su Y, Zhang Y, Guan A, Wang H. The role and molecular mechanism of metabolic reprogramming of colorectal cancer by UBR5 through PYK2 regulation of OXPHOS expression study. J Biochem Mol Toxicol 2023; 37:e23376. [PMID: 37098808 DOI: 10.1002/jbt.23376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/21/2023] [Accepted: 04/14/2023] [Indexed: 04/27/2023]
Abstract
Colorectal carcinoma (CRC) is the third most malignant tumor in the world, but the key mechanisms of CRC progression have not been confirmed. UBR5 and PYK2 expression levels were detected by RT-qPCR. The levels of UBR5, PYK2, and mitochondrial oxidative phosphorylation (OXPHOS) complexes were detected by western blot analysis. Flow cytometry was used to detect ROS activity. The CCK-8 assay was used to assess cell proliferation and viability. The interaction between UBR5 and PYK2 was detected by immunoprecipitation. A clone formation assay was used to determine the cell clone formation rate. The ATP level and lactate production of each group of cells were detected by the kit. EdU staining was performed for cell proliferation.Transwell assay was performed for cell migration ability. For the CRC nude mouse model, we also observed and recorded the volume and mass of tumor-forming tumors. The expression of UBR5 and PYK2 was elevated in both CRC and human colonic mucosal epithelial cell lines, and knockdown of UBR5 had inhibitory effects on cancer cell proliferation and cloning and other behaviors in the CRC process by knockdown of UBR5 to downregulate the expression of PYK2, thus inhibiting the OXPHOS process in CRC; rotenone (OXPHOS inhibitor) treatment enhanced all these inhibitory effects. Knockdown of UBR5 can reduce the expression level of PYK2, thus downregulating the OXPHOS process in CRC cell lines and inhibiting the CRC metabolic reprogramming process.
Collapse
Affiliation(s)
- Rong Qin
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Yun Huang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming City, Yunnan, China
| | - Ying Yao
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Likun Wang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Zhibo Zhang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Weikang Huang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Yu Su
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Yulu Zhang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Aoran Guan
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
| | - Hui Wang
- Department of Gastroenterology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming City, Yunnan, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming City, Yunnan, China
| |
Collapse
|
18
|
Das S, Kundu M, Hassan A, Parekh A, Jena BC, Mundre S, Banerjee I, Yetirajam R, Das CK, Pradhan AK, Das SK, Emdad L, Mitra P, Fisher PB, Mandal M. A novel computational predictive biological approach distinguishes Integrin β1 as a salient biomarker for breast cancer chemoresistance. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166702. [PMID: 37044238 DOI: 10.1016/j.bbadis.2023.166702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/11/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Chemoresistance is a primary cause of breast cancer treatment failure, and protein-protein interactions significantly contribute to chemoresistance during different stages of breast cancer progression. In pursuit of novel biomarkers and relevant protein-protein interactions occurring during the emergence of breast cancer chemoresistance, we used a computational predictive biological (CPB) approach. CPB identified associations of adhesion molecules with proteins connected with different breast cancer proteins associated with chemoresistance. This approach identified an association of Integrin β1 (ITGB1) with chemoresistance and breast cancer stem cell markers. ITGB1 activated the Focal Adhesion Kinase (FAK) pathway promoting invasion, migration, and chemoresistance in breast cancer by upregulating Erk phosphorylation. FAK also activated Wnt/Sox2 signaling, which enhanced self-renewal in breast cancer. Activation of the FAK pathway by ITGB1 represents a novel mechanism linked to breast cancer chemoresistance, which may lead to novel therapies capable of blocking breast cancer progression by intervening in ITGB1-regulated signaling pathways.
Collapse
Affiliation(s)
- Subhayan Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Moumita Kundu
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Atif Hassan
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Aditya Parekh
- Anant National University, Ahmedabad, Gujarat, India
| | - Bikash Ch Jena
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Swati Mundre
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Indranil Banerjee
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; School of Pharmacy, Sister Nivedita University (Techno India Group), Kolkata, West Bengal, India
| | - Rajesh Yetirajam
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chandan K Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Pralay Mitra
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
19
|
3,3'-Diindolylmethane Augments 5-Fluorouracil-InducedGrowth Suppression in Gastric Cancer Cells through Suppression of the Akt/GSK-3 β and WNT/Beta-Catenin. JOURNAL OF ONCOLOGY 2023; 2023:8268955. [PMID: 36785670 PMCID: PMC9922186 DOI: 10.1155/2023/8268955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 02/09/2023]
Abstract
Gastric cancer (GC) is one of the most lethal cancers in South Korea, and it is a cancer of concern worldwide. 5-fluorouracil (5-Fu) is commonly used as the first-line therapy for advanced GC; however, its side effects often limit the dosage range and impair patients' quality of life. Due to the limitations of current chemotherapy, new anticancer therapies are urgently needed. 3,3'-diindolylmethane (DIM) has been reported to have the ability to protect against various types of cancer. Our study aimed to elucidate the anticancer effect of DIM in GC when treated with the chemotherapeutic agent 5-Fu. In our results, combined treatment with DIM and 5-Fu resulted in higher apoptosis and lower cell proliferation than treatment with 5-Fu in SNU484 and SNU638 cell lines. Furthermore, when DIM and 5-Fu were administered together, cell invasion was diminished by mediated E-cadherin, MMP-9, and uPA; p-Akt and p-GSK-3β levels were reduced more significantly than when 5-Fu was administered alone. Moreover, in the Wnt signaling pathway, combined treatment of DIM and 5-Fu diminished β-catenin levels in the nucleus and inhibited cyclin D1and c-Myc protein levels. The Akt inhibitor, wortmannin, further inhibited the levels of β-catenin and c-Myc that were inhibited by DIM and 5-Fu. Furthermore, an animal xenograft model demonstrated that DIM combined with 5-Fu considerably reduced tumor growth without any toxic effects by regulating the Akt/GSK-3β and β-catenin levels. Our findings suggest that DIM significantly potentiates the anticancer effects of 5-Fu by targeting the Akt/GSK-3β and WNT/β-catenin because the combination therapy is more effective than 5-Fu alone, thereby offering an innovative potential therapy for patients with GC.
Collapse
|
20
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
21
|
Lee D, Hong JH. Activated PyK2 and Its Associated Molecules Transduce Cellular Signaling from the Cancerous Milieu for Cancer Metastasis. Int J Mol Sci 2022; 23:ijms232415475. [PMID: 36555115 PMCID: PMC9779422 DOI: 10.3390/ijms232415475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
PyK2 is a member of the proline-rich tyrosine kinase and focal adhesion kinase families and is ubiquitously expressed. PyK2 is mainly activated by stimuli, such as activated Src kinases and intracellular acidic pH. The mechanism of PyK2 activation in cancer cells has been addressed extensively. The up-regulation of PyK2 through overexpression and enhanced phosphorylation is a key feature of tumorigenesis and cancer migration. In this review, we summarized the cancer milieu, including acidification and cancer-associated molecules, such as chemical reagents, interactive proteins, chemokine-related molecules, calcium channels/transporters, and oxidative molecules that affect the fate of PyK2. The inhibition of PyK2 leads to a beneficial strategy to attenuate cancer cell development, including metastasis. Thus, we highlighted the effect of PyK2 on various cancer cell types and the distribution of molecules that affect PyK2 activation. In particular, we underlined the relationship between PyK2 and cancer metastasis and its potential to treat cancer cells.
Collapse
|
22
|
Ventura E, Xie C, Buraschi S, Belfiore A, Iozzo RV, Giordano A, Morrione A. Complexity of progranulin mechanisms of action in mesothelioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:333. [PMID: 36471440 PMCID: PMC9720952 DOI: 10.1186/s13046-022-02546-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mesothelioma is an aggressive disease with limited therapeutic options. The growth factor progranulin plays a critical role in several cancer models, where it regulates tumor initiation and progression. Recent data from our laboratories have demonstrated that progranulin and its receptor, EphA2, constitute an oncogenic pathway in bladder cancer by promoting motility, invasion and in vivo tumor formation. Progranulin and EphA2 are expressed in mesothelioma cells but their mechanisms of action are not well defined. In addition, there are no data establishing whether the progranulin/EphA2 axis is tumorigenic for mesothelioma cells. METHODS The expression of progranulin in various mesothelioma cell lines derived from all major mesothelioma subtypes was examined by western blots on cell lysates, conditioned media and ELISA assays. The biological roles of progranulin, EphA2, EGFR, RYK and FAK were assessed in vitro by immunoblots, human phospho-RTK antibody arrays, pharmacological (specific inhibitors) and genetic (siRNAs, shRNAs, CRISPR/Cas9) approaches, motility, invasion and adhesion assays. In vivo tumorigenesis was determined by xenograft models. Focal adhesion turnover was evaluated biochemically using focal adhesion assembly/disassembly assays and immunofluorescence analysis with focal adhesion-specific markers. RESULTS In the present study we show that progranulin is upregulated in various mesothelioma cell lines covering all mesothelioma subtypes and is an important regulator of motility, invasion, adhesion and in vivo tumor formation. However, our results indicate that EphA2 is not the major functional receptor for progranulin in mesothelioma cells, where progranulin activates a complex signaling network including EGFR and RYK. We further characterized progranulin mechanisms of action and demonstrated that progranulin, by modulating FAK activity, regulates the kinetic of focal adhesion disassembly, a critical step for cell motility. CONCLUSION Collectively, our results highlight the complexity of progranulin oncogenic signaling in mesothelioma, where progranulin modulate functional cross-talks between multiple RTKs, thereby suggesting the need for combinatorial therapeutic approaches to improve treatments of this aggressive disease.
Collapse
Affiliation(s)
- Elisa Ventura
- grid.264727.20000 0001 2248 3398Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122 USA
| | - Christopher Xie
- grid.412726.40000 0004 0442 8581Department of Pathology, Anatomy and Cell Biology, Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Simone Buraschi
- grid.412726.40000 0004 0442 8581Department of Pathology, Anatomy and Cell Biology, Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Antonino Belfiore
- grid.8158.40000 0004 1757 1969Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Renato V. Iozzo
- grid.412726.40000 0004 0442 8581Department of Pathology, Anatomy and Cell Biology, Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Antonio Giordano
- grid.264727.20000 0001 2248 3398Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122 USA ,grid.9024.f0000 0004 1757 4641Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Andrea Morrione
- grid.264727.20000 0001 2248 3398Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122 USA
| |
Collapse
|
23
|
Jiang L, Sun YJ, Song XH, Sun YY, Yang WY, Li J, Wu YJ. Ivermectin inhibits tumor metastasis by regulating the Wnt/β-catenin/integrin β1/FAK signaling pathway. Am J Cancer Res 2022; 12:4502-4519. [PMID: 36381328 PMCID: PMC9641399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/01/2022] [Indexed: 06/16/2023] Open
Abstract
Tumor metastasis is the major cause of cancer mortality; therefore, it is imperative to discover effective therapeutic drugs for anti-metastasis therapy. In the current study, we investigated whether ivermectin (IVM), an FDA-approved antiparasitic drug, could prevent cancer metastasis. Colorectal and breast cancer cell lines and a cancer cell-derived xenograft tumor metastasis model were used to investigate the anti-metastasis effect of IVM. Our results showed that IVM significantly inhibited the motility of cancer cells in vitro and tumor metastasis in vivo. Mechanistically, IVM suppressed the expressions of the migration-related proteins via inhibiting the activation of Wnt/β-catenin/integrin β1/FAK and the downstream signaling cascades. Our findings indicated that IVM was capable of suppressing tumor metastasis, which provided the rationale on exploring the potential clinical application of IVM in the prevention and treatment of cancer metastasis.
Collapse
Affiliation(s)
- Lu Jiang
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
- Henan University of Chinese MedicineZhengzhou 450046, Henan, China
| | - Ying-Jian Sun
- Department of Veterinary Medicine, Beijing University of AgricultureBeijing 102206, China
| | - Xiao-Hua Song
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
| | - Yan-Yan Sun
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
| | - Wen-Yao Yang
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
| | - Jing Li
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
| | - Yi-Jun Wu
- Laboratory of Molecular Toxicology, Institute of Zoology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of SciencesBeijing 100101, China
| |
Collapse
|
24
|
Zhang C, Zhu X, Li Y, Shao J, Xu H, Chen L, Dan Y, Jin H, He A. High expression of PYK2 is associated with poor prognosis and cancer progression in early-stage cervical carcinoma. Medicine (Baltimore) 2022; 101:e31178. [PMID: 36253980 PMCID: PMC9575807 DOI: 10.1097/md.0000000000031178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Proline-rich tyrosine kinase-2 (PYK2), also known as calcium dependent tyrosine kinase, regulates different signal transduction cascades that control cell proliferation, migration, and invasion. However, the role of PYK2 in cervical cancer remains to be elucidated. The current study retrospectively included 134 patients with cervical cancer from December 2007 to September 2014. PYK2 expression was detected in tissue microarray and cervical cancer cell lines. Statistical analysis was performed to evaluate its clinicopathological significance. Small interfering RNA (siRNA) was employed to suppress endogenous PYK2 expression in cervical cancer cells to observe the biological function. PYK2 expression was up-regulated in cervical cancer specimens compared with paired adjacent normal cervical tissue samples. Statistical analyses indicated that PYK2 expression might be an independent prognostic indicator for patients with early-stage cervical cancer. A nomogram model was constructed based on PYK2 expression and other clinicopathological risk factors, and it performed well in predicting patients survival. In cellular studies, down-regulation of PYK2 remarkably inhibited cellular proliferation, migration and invasion. PYK2 expression possessed the potential to serve as a novel prognostic marker in cervical cancer patients.
Collapse
Affiliation(s)
- Can Zhang
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Xinghua Zhu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, Nantong, China
| | - Yong Li
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Jia Shao
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Haibo Xu
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Lei Chen
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Youli Dan
- Medical College of Nantong University, Nantong, China
| | - Hua Jin
- Cancer Research Centre Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Aiqin He
- Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
- *Correspondence: Aiqin He, Department of Gynecology Oncology, Affiliated Tumor Hospital of Nantong University, 30 Tongyang North Road, Nantong, 226300, China (e-mail: )
| |
Collapse
|
25
|
Pancreatic ductal adenocarcinoma: tumor microenvironment and problems in the development of novel therapeutic strategies. Clin Exp Med 2022:10.1007/s10238-022-00886-1. [DOI: 10.1007/s10238-022-00886-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/30/2022] [Indexed: 12/19/2022]
|
26
|
Li J, Hou W, Yang Y, Deng Q, Fu H, Yin Y, Duan K, Feng B, Guo T, Weng J. Micro/nano-topography promotes osteogenic differentiation of bone marrow stem cells by regulating periostin expression. Colloids Surf B Biointerfaces 2022; 218:112700. [PMID: 35907353 DOI: 10.1016/j.colsurfb.2022.112700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022]
Abstract
Micro/nano-topography (MNT) is an important factor affecting cell response. Earlier studies using titania (TiO2) nanotube as a model of MNT found that they mediated the differentiation of BMSCs into osteoblasts, but the mechanisms are not fully understood. Surprisingly, Periostin (Postn), a secreted protein involved in extracellular matrix (ECM) construction and promoting osteogenic differentiation of bone marrow stem cells (BMSCs), was previously observed to significantly up-regulated on TiO2 nanotube. We proposed that Postn may act as a MNT signal transduction role. In this study, we investigated the effect of MNT on Postn, and the influence of Postn on osteogenic differentiation-related genes through focal adhesion and downstream signals. It was found that, titanium (Ti) plates carrying TiO2 nanotubes with diameters of ∼100 nm (TNT-100) significantly up-regulated the expression of Postn compared with flat Ti. Furthermore, Postn activated the downstream focal adhesion kinase (FAK) signal pathway and β-catenin into the nucleus by interacting with integrin αV. Surprisingly, TNT-100 up-regulated the transcription level of Wnt3a, which was independent of the up-regulation of Postn. This new Postn signaling pathway may provide more insights into the signal transduction mechanism of MNT and development of biomaterials with improved osteogenic properties.
Collapse
Affiliation(s)
- Jinsheng Li
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenqing Hou
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qing Deng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Hong Fu
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yiran Yin
- Sichuan Provincial Lab of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ke Duan
- Sichuan Provincial Lab of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Bo Feng
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tailin Guo
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Jie Weng
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China; College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
27
|
FAK in Cancer: From Mechanisms to Therapeutic Strategies. Int J Mol Sci 2022; 23:ijms23031726. [PMID: 35163650 PMCID: PMC8836199 DOI: 10.3390/ijms23031726] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 01/25/2023] Open
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is overexpressed and activated in many cancer types. FAK regulates diverse cellular processes, including growth factor signaling, cell cycle progression, cell survival, cell motility, angiogenesis, and the establishment of immunosuppressive tumor microenvironments through kinase-dependent and kinase-independent scaffolding functions in the cytoplasm and nucleus. Mounting evidence has indicated that targeting FAK, either alone or in combination with other agents, may represent a promising therapeutic strategy for various cancers. In this review, we summarize the mechanisms underlying FAK-mediated signaling networks during tumor development. We also summarize the recent progress of FAK-targeted small-molecule compounds for anticancer activity from preclinical and clinical evidence.
Collapse
|
28
|
Juhl OJ, Merife A, Zhang Y, Donahue HJ. Inhibition of focal adhesion turnover prevents osteoblastic differentiation through β‐catenin mediated transduction of pro‐osteogenic substrate. J Biomed Mater Res B Appl Biomater 2022; 110:1573-1586. [PMID: 35099117 PMCID: PMC9306686 DOI: 10.1002/jbm.b.35018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022]
Abstract
The mechanism by which substrate surface characteristics are transduced by osteoblastic cells and their progenitors is not fully known. Data from previous studies by our group suggest the involvement of β‐catenin in the mechanism by which substrate surface characteristics are transduced. This focal adhesion and β‐catenin mediated mechanism functions through the liberation of β‐catenin from focal adhesion complexes in response to pro‐osteogenic substrate (POS) characteristics. After liberation, β‐catenin translocates and facilitates upregulation of genes associated with osteogenesis. It is not known whether the observed correlation between focal adhesion turnover and β‐catenin translocation directly results from focal adhesion turnover. In this study we inhibited focal adhesion turnover using a focal adhesion kinase inhibitor PF‐573228. We found that inhibition of focal adhesion turnover resulted in an abrogation of the more rapid translocation and increased transcriptional activity of β‐catenin induced by POS. In addition, inhibition of focal adhesion turnover mitigated the increase in osteoblastic differentiation induced by a POS as measured by alkaline phosphatase enzymatic activity and osteogenic gene and protein expression. Together, these data, coupled with previous findings, suggest that the observed β‐catenin translocation is a result of focal adhesion turnover, providing evidence for a focal adhesion initiated, β‐catenin mediated mechanism of substrate surface signal transduction.
Collapse
Affiliation(s)
- Otto J. Juhl
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Anna‐Blessing Merife
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Yue Zhang
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| | - Henry J. Donahue
- Department of Biomedical Engineering and Institute for Engineering and Medicine Virginia Commonwealth University Richmond Virginia USA
| |
Collapse
|
29
|
Cancer-associated fibroblasts promote oral squamous cell carcinoma progression through LOX-mediated matrix stiffness. J Transl Med 2021; 19:513. [PMID: 34930321 PMCID: PMC8686394 DOI: 10.1186/s12967-021-03181-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer-associated fibroblasts (CAFs), the most abundant cells in the tumor microenvironment, have prominent roles in the development of solid tumors as stromal targets. However, the underlying mechanism of CAFs’ function in oral squamous cell carcinoma (OSCC) development remains unclear. Here, we investigated the role of lysyl oxidase (LOX) expression in CAFs in tumor stromal remodeling and the mechanism of its effect on OSCC progression. Methods Multiple immunohistochemistry (IHC) staining was performed to detect the correlation of CAFs and LOX in the stroma of OSCC specimens, as well as the correlation with clinicopathological parameters and prognosis. The expression of LOX in CAFs were detected by RT-qPCR and western blot. The effects of LOX in CAFs on the biological characteristics of OSCC cell line were investigated using CCK-8, wound-healing and transwell assay. CAFs were co-cultured with type I collagen in vitro, and collagen contraction test, microstructure observation and rheometer were used to detect the effect of CAFs on remodeling collagen matrix. Then, collagen with different stiffness were established to investigate the effect of matrix stiffness on the progression of OSCC. Moreover, we used focal adhesion kinase (FAK) phosphorylation inhibitors to explored whether the increase in matrix stiffness promote the progression of OSCC through activating FAK phosphorylation pathway. Results LOX was colocalized with CAFs in the stroma of OSCC tissues, and its expression was significantly related to the degree of malignant differentiation and poor prognosis in OSCC. LOX was highly expressed in CAFs, and its knockdown impaired the proliferation, migration, invasion and EMT process of OSCC cells. The expression of LOX in CAFs can catalyze collagen crosslinking and increase matrix stiffness. Furthermore, CAFs-derived LOX-mediated increase in collagen stiffness induced morphological changes and promoted invasion and EMT process in OSCC cells by activating FAK phosphorylation pathway. Conclusions Our findings suggest that CAFs highly express LOX in the stroma of OSCC and can remodel the matrix collagen microenvironment, and the increase in matrix stiffness mediated by CAFs-derived LOX promotes OSCC development through FAK phosphorylation pathway. Thus, LOX may be a potential target for the early diagnosis and therapeutic treatment of OSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03181-x.
Collapse
|
30
|
Kinases of the Focal Adhesion Complex Contribute to Cardiomyocyte Specification. Int J Mol Sci 2021; 22:ijms221910430. [PMID: 34638793 PMCID: PMC8508671 DOI: 10.3390/ijms221910430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022] Open
Abstract
Differentiation of pluripotent stem cells to cardiomyocytes is influenced by culture conditions including the extracellular matrices or similar synthetic scaffolds on which they are grown. However, the molecular mechanisms that link the scaffold with differentiation outcomes are not fully known. Here, we determined by immunofluorescence staining and mass spectrometry approaches that extracellular matrix (ECM) engagement by mouse pluripotent stem cells activates critical components of canonical wingless/integrated (Wnt) signaling pathways via kinases of the focal adhesion to drive cardiomyogenesis. These kinases were found to be differentially activated depending on type of ECM engaged. These outcomes begin to explain how varied ECM composition of in vivo tissues with development and in vitro model systems gives rise to different mature cell types, having broad practical applicability for the design of engineered tissues.
Collapse
|
31
|
Shen J, Yang J, Sang L, Sun R, Bai W, Wang C, Sun Y, Sun J. PYK2 mediates the BRAF inhibitor (vermurafenib)-induced invadopodia formation and metastasis in melanomas. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0294. [PMID: 34570440 PMCID: PMC9425182 DOI: 10.20892/j.issn.2095-3941.2020.0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Objective: The BRAF inhibitor, vemurafenib, has been widely used in the treatment of patients with melanoma-bearing BRAFV600E mutations. While the initial response to vemurafenib is usually excellent, the majority of patients eventually develop resistance and metastatic disease. However, the underlying molecular mechanism remains elusive. The objective of this study was therefore to identify additional molecular targets responsible for vemurafenib resistance. Methods: Western blots and immunohistochemistry analyses were used to evaluate expressions of PYK2 and p-PYK2 in cultured cells and melanoma tissue microarrays. The relationships of p-PYK2 with clinicopathological parameters were statistically analyzed. Invadopodia cell invasion, and a Ca2+ assay were used to determine the effect of vemurafenib resistance-induced p-PYK2 on melanoma progression. A mouse model was used to assess the effects of PYK2 on melanoma metastasis. Results: Elevated p-PYK2 levels were detected in vemurafenib-resistant melanoma cells, and PYK2 was shown to regulate invadopodia formation in melanoma cells. Vemurafenib triggered invadopodia formation by activation of PYK2. Inhibition of PYK2 with either shRNA or the small molecule inhibitor, PF562711, dramatically reduced vemurafenib-induced invadopodia formation. Furthermore, knockdown of PYK2 significantly reduced melanoma lung metastasis in vivo. Increased expressions of p-PYK2 in melanoma patients were positively correlated with advanced stage (P = 0.002), metastasis (P < 0.001), and Clark grade (P < 0.001), and were also associated with short overall survival [hazard ratio (HR) = 3.304, P = 0.007] and progression-free survival (HR = 2.930, P = 0.001). Conclusions: PYK2 mediated vemurafenib-induced melanoma cell migration and invasion. Inhibition of PYK2 resensitized melanoma cells to vemurafenib. Phospho-PYK2 was a prognostic biomarker in melanoma patients.
Collapse
Affiliation(s)
- Junling Shen
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Jilong Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Sang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Rui Sun
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Weiyu Bai
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Chao Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yan Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jianwei Sun
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| |
Collapse
|
32
|
Benwell CJ, Taylor JAGE, Robinson SD. Endothelial neuropilin-2 influences angiogenesis by regulating actin pattern development and α5-integrin-p-FAK complex recruitment to assembling adhesion sites. FASEB J 2021; 35:e21679. [PMID: 34314542 DOI: 10.1096/fj.202100286r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
The ability to form a variety of cell-matrix connections is crucial for angiogenesis to take place. Without stable anchorage to the extracellular matrix (ECM), endothelial cells (ECs) are unable to sense, integrate and disseminate growth factor stimulated responses that drive growth of a vascular bed. Neuropilin-2 (NRP2) is a widely expressed membrane-bound multifunctional non-tyrosine kinase receptor, which has previously been implicated in influencing cell adhesion and migration by interacting with α5-integrin and regulating adhesion turnover. α5-integrin, and its ECM ligand fibronectin (FN) are both known to be upregulated during the formation of neo-vasculature. Despite being descriptively annotated as a candidate biomarker for aggressive cancer phenotypes, the EC-specific roles for NRP2 during developmental and pathological angiogenesis remain unexplored. The data reported here support a model whereby NRP2 actively promotes EC adhesion and migration by regulating dynamic cytoskeletal remodeling and by stimulating Rab11-dependent recycling of α5-integrin-p-FAK complexes to newly assembling adhesion sites. Furthermore, temporal depletion of EC-NRP2 in vivo impairs primary tumor growth by disrupting vessel formation. We also demonstrate that EC-NRP2 is required for normal postnatal retinal vascular development, specifically by regulating cell-matrix adhesion. Upon loss of endothelial NRP2, vascular outgrowth from the optic nerve during superficial plexus formation is disrupted, likely due to reduced FAK phosphorylation within sprouting tip cells.
Collapse
Affiliation(s)
- Christopher J Benwell
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - James A G E Taylor
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D Robinson
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| |
Collapse
|
33
|
Zhu T, Yang Q, Shao J, Chen Z, Cai B, Mao G. Pyk2 level is a novel prognostic marker for patients with esophageal squamous cell carcinoma after radical surgery. Virchows Arch 2021; 479:905-917. [PMID: 34313839 DOI: 10.1007/s00428-021-03153-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/24/2021] [Accepted: 07/03/2021] [Indexed: 12/09/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors in East Asia. Surgical resection is currently the typical treatment. However, due to the highly invasive and metastatic characteristic of the disease, the mortality rate is still high. A search for potential prognostic biomarkers and therapeutic targets is very necessary. Here, we studied the expression of proline-rich tyrosine kinase 2 (Pyk2), a non-receptor tyrosine protein kinase, in ESCC and its influence on prognosis. A total of 112 cases of ESCC and paired adjacent normal tissues (NT) were organized in tissue microarray (TMA) from the Nantong First People's Hospital. Our analysis of TMA revealed that Pyk2 levels were higher in ESCC than in paired adjacent NT by immunohistochemistry (p<0.001). Western blot and real-time quantitative PCR analysis (p=0.0359) also reached similar conclusions. To further explore the significance of Pyk2 in ESCC, another set of tissue microarrays was collected from the Affiliated Hospital of Nantong University, which includes 241 consecutive patients undergoing radical surgery for ESCC, to perform IHC scores. We demonstrated that the expression level of Pyk2 was positively correlated with N stage (node negative versus node positive, p=0.02) and clinical stage (I + II versus III + IV, p=0.042). Univariate and multivariate analyses suggested that high Pyk2 expression was an independent prognostic factor for overall survival with ESCC. Cell function studies found that Pyk2 promoted tumor proliferation and migration and reduced apoptosis. Pyk2 knockdown enhanced the sensitivity to cisplatin in ESCC cells. Western blot analysis confirmed that Pyk2 may promote tumor progression by activating the Akt signaling pathway.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Oncology, The First People's Hospital of Yancheng, Yancheng, China
| | - Qiuxing Yang
- Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Jingjing Shao
- Cancer Research Center Nantong, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Zhuolin Chen
- Department of Pathology, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Bo Cai
- Nantong Center for Disease Control and Prevention Institute of Chronic Noncommunicable Diseases Prevention and Control, Nantong, China.
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
34
|
Peng DY, Reed-Maldonado AB, Lin GT, Xia SJ, Lue TF. Low-intensity pulsed ultrasound for regenerating peripheral nerves: potential for penile nerve. Asian J Androl 2021; 22:335-341. [PMID: 31535626 PMCID: PMC7406088 DOI: 10.4103/aja.aja_95_19] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Peripheral nerve damage, such as that found after surgery or trauma, is a substantial clinical challenge. Much research continues in attempts to improve outcomes after peripheral nerve damage and to promote nerve repair after injury. In recent years, low-intensity pulsed ultrasound (LIPUS) has been studied as a potential method of stimulating peripheral nerve regeneration. In this review, the physiology of peripheral nerve regeneration is reviewed, and the experiments employing LIPUS to improve peripheral nerve regeneration are discussed. Application of LIPUS following nerve surgery may promote nerve regeneration and improve functional outcomes through a variety of proposed mechanisms. These include an increase of neurotrophic factors, Schwann cell (SC) activation, cellular signaling activations, and induction of mitosis. We searched PubMed for articles related to these topics in both in vitro and in vivo animal research models. We found numerous studies, suggesting that LIPUS following nerve surgery promotes nerve regeneration and improves functional outcomes. Based on these findings, LIPUS could be a novel and valuable treatment for nerve injury-induced erectile dysfunction.
Collapse
Affiliation(s)
- Dong-Yi Peng
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA 94143, USA.,Department of Urology, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA 94143, USA
| | - Gui-Ting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA 94143, USA
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
35
|
Quintanal-Villalonga A, Taniguchi H, Zhan YA, Hasan MM, Chavan SS, Meng F, Uddin F, Manoj P, Donoghue MTA, Won HH, Chan JM, Ciampricotti M, Chow A, Offin M, Chang JC, Ray-Kirton J, Tischfield SE, Egger J, Bhanot UK, Linkov I, Asher M, Sinha S, Silber J, Iacobuzio-Donahue CA, Roehrl MH, Hollmann TJ, Yu HA, Qiu J, de Stanchina E, Baine MK, Rekhtman N, Poirier JT, Loomis B, Koche RP, Rudin CM, Sen T. Multi-omic analysis of lung tumors defines pathways activated in neuroendocrine transformation. Cancer Discov 2021; 11:3028-3047. [PMID: 34155000 DOI: 10.1158/2159-8290.cd-20-1863] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/30/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022]
Abstract
Lineage plasticity is implicated in treatment resistance in multiple cancers. In lung adenocarcinomas (LUADs) amenable to targeted therapy, transformation to small cell lung cancer (SCLC) is a recognized resistance mechanism. Defining molecular mechanisms of neuroendocrine (NE) transformation in lung cancer has been limited by a paucity of pre-/post-transformation clinical samples. Detailed genomic, epigenomic, transcriptomic, and protein characterization of combined LUAD/SCLC tumors, as well as pre-/post-transformation samples, support that NE transformation is primarily driven by transcriptional reprogramming rather than mutational events. We identify genomic contexts in which NE transformation is favored, including frequent loss of the 3p chromosome arm. We observed enhanced expression of genes involved in PRC2 complex and PI3K/AKT and NOTCH pathways. Pharmacological inhibition of the PI3K/AKT pathway delayed tumor growth and NE transformation in an EGFR-mutant patient-derived xenograft model. Our findings define a novel landscape of potential drivers and therapeutic vulnerabilities of neuroendocrine transformation in lung cancer.
Collapse
Affiliation(s)
| | | | - Yingqian A Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center
| | - Maysun M Hasan
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | | | - Fanli Meng
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | | | | | - Mark T A Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | - Helen H Won
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | | | | | - Andrew Chow
- Medicine, Memorial Sloan Kettering Cancer Center
| | | | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | - Sam E Tischfield
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | | | - Umesh K Bhanot
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center
| | | | - Marina Asher
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | | | - Helena A Yu
- Medicine, Memorial Sloan Kettering Cancer Center
| | - Juan Qiu
- Memorial Sloan Kettering Cancer Center
| | | | | | | | - John T Poirier
- Perlmutter Cancer Center, New York University Langone Health
| | - Brian Loomis
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center
| | - Charles M Rudin
- Druckenmiller Center for Lung Cancer Research and Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center
| | | |
Collapse
|
36
|
Dawson JC, Serrels A, Stupack DG, Schlaepfer DD, Frame MC. Targeting FAK in anticancer combination therapies. Nat Rev Cancer 2021; 21:313-324. [PMID: 33731845 PMCID: PMC8276817 DOI: 10.1038/s41568-021-00340-6] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2021] [Indexed: 01/31/2023]
Abstract
Focal adhesion kinase (FAK) is both a non-receptor tyrosine kinase and an adaptor protein that primarily regulates adhesion signalling and cell migration, but FAK can also promote cell survival in response to stress. FAK is commonly overexpressed in cancer and is considered a high-value druggable target, with multiple FAK inhibitors currently in development. Evidence suggests that in the clinical setting, FAK targeting will be most effective in combination with other agents so as to reverse failure of chemotherapies or targeted therapies and enhance efficacy of immune-based treatments of solid tumours. Here, we discuss the recent preclinical evidence that implicates FAK in anticancer therapeutic resistance, leading to the view that FAK inhibitors will have their greatest utility as combination therapies in selected patient populations.
Collapse
Affiliation(s)
- John C Dawson
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| | - Alan Serrels
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Dwayne G Stupack
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Centre, La Jolla, CA, USA
| | - David D Schlaepfer
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Centre, La Jolla, CA, USA
| | - Margaret C Frame
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
37
|
Development of Pigmentation-Regulating Agents by Drug Repositioning. Int J Mol Sci 2021; 22:ijms22083894. [PMID: 33918792 PMCID: PMC8069425 DOI: 10.3390/ijms22083894] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
Skin color is determined by the processes of melanin synthesis and distribution. Problems in various molecules or signaling pathways involved in melanin synthesis contribute to skin pigmentation defects. Several trials have been conducted on the production of pigmentation-regulating agents, and drug repositioning has emerged as a modern technique to identify new uses for existing drugs. Our research team has researched substances or drugs associated with pigmentation control and, as a result, nilotinib, sorafenib, and ICG-001 have been found to promote pigmentation, while 5-iodotubercidin inhibits pigmentation. Therefore, these substances or medications were suggested as potential therapeutics for pigmentation disorders by drug repositioning.
Collapse
|
38
|
Trajectory modeling of endothelial-to-mesenchymal transition reveals galectin-3 as a mediator in pulmonary fibrosis. Cell Death Dis 2021; 12:327. [PMID: 33771973 PMCID: PMC7998015 DOI: 10.1038/s41419-021-03603-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022]
Abstract
The endothelial-to-mesenchymal transition (EndMT) is an important source of fibrotic cells in idiopathic pulmonary fibrosis (IPF). However, how endothelial cells (ECs) are activated and how EndMT impact IPF remain largely elusive. Here, we use unsupervised pseudotemporal analysis to recognize the heterogeneity of ECs and reconstruct EndMT trajectory of bleomycin (BLM)-treated Tie2creER/+;Rosa26tdTomato/+ IPF mice. Genes like C3ar1 and Lgals3 (protein name galectin-3) are highly correlated with the transitional pseudotime, whose expression is gradually upregulated during the fate switch of ECs from quiescence to activation in fibrosis. Inhibition of galectin-3 via siRNA or protein antagonists in mice could alleviate the pathogenesis of IPF and the transition of ECs. With the stimulation of human pulmonary microvascular endothelial cells (HPMECs) by recombinant proteins and/or siRNAs for galectin-3 in vitro, β-catenin/GSK3β signaling and its upstream regulator AKT are perturbed, which indicates they mediate the EndMT progress. These results suggest that EndMT is essential to IPF process and provide potential therapeutic targets for vascular remodeling.
Collapse
|
39
|
Griffith BGC, Upstill-Goddard R, Brunton H, Grimes GR, Biankin AV, Serrels B, Byron A, Frame MC. FAK regulates IL-33 expression by controlling chromatin accessibility at c-Jun motifs. Sci Rep 2021; 11:229. [PMID: 33420223 PMCID: PMC7794255 DOI: 10.1038/s41598-020-80111-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/10/2020] [Indexed: 01/29/2023] Open
Abstract
Focal adhesion kinase (FAK) localizes to focal adhesions and is overexpressed in many cancers. FAK can also translocate to the nucleus, where it binds to, and regulates, several transcription factors, including MBD2, p53 and IL-33, to control gene expression by unknown mechanisms. We have used ATAC-seq to reveal that FAK controls chromatin accessibility at a subset of regulated genes. Integration of ATAC-seq and RNA-seq data showed that FAK-dependent chromatin accessibility is linked to differential gene expression, including of the FAK-regulated cytokine and transcriptional regulator interleukin-33 (Il33), which controls anti-tumor immunity. Analysis of the accessibility peaks on the Il33 gene promoter/enhancer regions revealed sequences for several transcription factors, including ETS and AP-1 motifs, and we show that c-Jun, a component of AP-1, regulates Il33 gene expression by binding to its enhancer in a FAK kinase-dependent manner. This work provides the first demonstration that FAK controls transcription via chromatin accessibility, identifying a novel mechanism by which nuclear FAK regulates biologically important gene expression.
Collapse
Affiliation(s)
- Billie G C Griffith
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Rosanna Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Holly Brunton
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Graeme R Grimes
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Bryan Serrels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
- NanoString Technologies, Inc, Seattle, WA, 98109, USA.
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK.
| | - Margaret C Frame
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK.
| |
Collapse
|
40
|
Dominijanni A, Devarasetty M, Soker S. Manipulating the Tumor Microenvironment in Tumor Organoids Induces Phenotypic Changes and Chemoresistance. iScience 2020; 23:101851. [PMID: 33319176 PMCID: PMC7724203 DOI: 10.1016/j.isci.2020.101851] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Tumors comprised a tightly surrounded tumor microenvironment, made up of non-cellular extracellular matrix (ECM) and stromal cells. Although treatment response is often attributed to tumor heterogeneity, progression and malignancy are profoundly influenced by tumor cell interactions with the surrounding ECM. Here, we used a tumor organoid model, consisting of hepatic stellate cells (HSCs) embedded in collagen type 1 (Col1) and colorectal cancer cell (HCT-116) spheroids, to determine the relationship between the ECM architecture, cancer cell malignancy, and chemoresistance. Exogenous transforming growth factor beta (TGF-β) used to activate the HSCs increased the remodeling and bundling of Col1 in the ECM around the cancer spheroid. A dense ECM architecture inhibited tumor cell growth, reversed their mesenchymal phenotype, preserved stem cell population, and reduced chemotherapy response. Overall, our results demonstrate that controlled biofabrication and manipulation of the ECM in tumor organoids results enables studying tumor cell-ECM interactions and better understand tumor cell response to chemotherapies.
Collapse
Affiliation(s)
- Anthony Dominijanni
- Wake Forest University School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| | - Mahesh Devarasetty
- Wake Forest University School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| | - Shay Soker
- Wake Forest University School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
41
|
van der Wal T, van Amerongen R. Walking the tight wire between cell adhesion and WNT signalling: a balancing act for β-catenin. Open Biol 2020; 10:200267. [PMID: 33292105 PMCID: PMC7776579 DOI: 10.1098/rsob.200267] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
CTNNB1 (catenin β-1, also known as β-catenin) plays a dual role in the cell. It is the key effector of WNT/CTNNB1 signalling, acting as a transcriptional co-activator of TCF/LEF target genes. It is also crucial for cell adhesion and a critical component of cadherin-based adherens junctions. Two functional pools of CTNNB1, a transcriptionally active and an adhesive pool, can therefore be distinguished. Whether cells merely balance the distribution of available CTNNB1 between these functional pools or whether interplay occurs between them has long been studied and debated. While interplay has been indicated upon artificial modulation of cadherin expression levels and during epithelial-mesenchymal transition, it is unclear to what extent CTNNB1 exchange occurs under physiological conditions and in response to WNT stimulation. Here, we review the available evidence for both of these models, discuss how CTNNB1 binding to its many interaction partners is controlled and propose avenues for future studies.
Collapse
Affiliation(s)
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
42
|
The Crosstalk between FAK and Wnt Signaling Pathways in Cancer and Its Therapeutic Implication. Int J Mol Sci 2020; 21:ijms21239107. [PMID: 33266025 PMCID: PMC7730291 DOI: 10.3390/ijms21239107] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Focal adhesion kinase (FAK) and Wnt signaling pathways are important contributors to tumorigenesis in several cancers. While most results come from studies investigating these pathways individually, there is increasing evidence of a functional crosstalk between both signaling pathways during development and tumor progression. A number of FAK-Wnt interactions are described, suggesting an intricate, context-specific, and cell type-dependent relationship. During development for instance, FAK acts mainly upstream of Wnt signaling; and although in intestinal homeostasis and mucosal regeneration Wnt seems to function upstream of FAK signaling, FAK activates the Wnt/β-catenin signaling pathway during APC-driven intestinal tumorigenesis. In breast, lung, and pancreatic cancers, FAK is reported to modulate the Wnt signaling pathway, while in prostate cancer, FAK is downstream of Wnt. In malignant mesothelioma, FAK and Wnt show an antagonistic relationship: Inhibiting FAK signaling activates the Wnt pathway and vice versa. As the identification of effective Wnt inhibitors to translate in the clinical setting remains an outstanding challenge, further understanding of the functional interaction between Wnt and FAK could reveal new therapeutic opportunities and approaches greatly needed in clinical oncology. In this review, we summarize some of the most relevant interactions between FAK and Wnt in different cancers, address the current landscape of Wnt- and FAK-targeted therapies in different clinical trials, and discuss the rationale for targeting the FAK-Wnt crosstalk, along with the possible translational implications.
Collapse
|
43
|
Liu WL, Chiang FT, Kao JTW, Chiou SH, Lin HL. GSK3 modulation in acute lung injury, myocarditis and polycystic kidney disease-related aneurysm. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118798. [PMID: 32693109 PMCID: PMC7368652 DOI: 10.1016/j.bbamcr.2020.118798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/17/2022]
Abstract
GSK3 are involved in different physical and pathological conditions and inflammatory regulated by macrophages contribute to significant mechanism. Infection stimuli may modulate GSK3 activity and influence host cell adaption, immune cells infiltration or cytokine expressions. To further address the role of GSK3 modulation in macrophages, the signal transduction of three major organs challenged by endotoxin, virus and genetic inherited factors are briefly introduced (lung injury, myocarditis and autosomal dominant polycystic kidney disease). As a result of pro-inflammatory and anti-inflammatory functions of GSK3 in different microenvironments and stages of macrophages (M1/M2), the rational resolution should be considered by adequately GSK3.
Collapse
Affiliation(s)
- Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Critical Care Medicine, Department of Emergency and Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Fu-Tien Chiang
- Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan,Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan,Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Heng-Liang Lin
- Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan; Division of Fund Managing, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
44
|
Lu Z, Liu H, Fu W, Wang Y, Geng J, Wang Y, Yu X, Wang Q, Xu H, Sui D. 20(S)-Protopanaxadiol inhibits epithelial-mesenchymal transition by promoting retinoid X receptor alpha in human colorectal carcinoma cells. J Cell Mol Med 2020; 24:14349-14365. [PMID: 33128348 PMCID: PMC7754066 DOI: 10.1111/jcmm.16054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/27/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Colorectal carcinoma (CRC) recurrence is often accompanied by metastasis. Most metastasis undergo through epithelial‐mesenchymal transition (EMT). Studies showed that retinol X receptor alpha (RXRα) and 20(S)‐Protopanaxadiol (PPD) have anti‐tumour effects. However, the anti‐metastasis effect of 20(S)‐PPD and the effect of RXRα on EMT‐induced metastasis are few studies on. Therefore, the role of RXRα and 20(S)‐PPD in CRC cell metastasis remains to be fully elucidated. RXRα with clinicopathological characteristics and EMT‐related expression in clinical samples were examined. Then, RXRα and EMT level in SW480 and SW620 cells, overexpressed and silenced RXRα in SW620 cells and SW480 cells, respectively, were evaluated. Finally, 20(S)‐PPD effect on SW620 and SW480 cells was evaluated. The results showed that a lower RXRα expression in cancer tissues, and a moderate negative correlation between RXRα and N stage, and tended to higher level of EMT. SW480 and SW620 cells had the highest and lowest RXRα expression among four CRC cell lines. SW480 had lower EMT level than SW620. Furthermore, 20(S)‐PPD increased RXRα and inhibited EMT level in SW620 cell. Finally, 20(S)‐PPD cannot restore SW480 cells EMT level to normal when RXRα silencing. These findings suggest that 20(S)‐PPD may inhibit EMT process in CRC cells by regulating RXRα expression.
Collapse
Affiliation(s)
- Zeyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongyan Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yuchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jianan Geng
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yaozhen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Quan Wang
- Department of Gastrocolorectal Surgery, First Affiliated Hospital of Jilin University, Changchun, China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
45
|
Liu Q, Garcia M, Wang S, Chen CW. Therapeutic Target Discovery Using High-Throughput Genetic Screens in Acute Myeloid Leukemia. Cells 2020; 9:cells9081888. [PMID: 32806592 PMCID: PMC7465943 DOI: 10.3390/cells9081888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
The development of high-throughput gene manipulating tools such as short hairpin RNA (shRNA) and CRISPR/Cas9 libraries has enabled robust characterization of novel functional genes contributing to the pathological states of the diseases. In acute myeloid leukemia (AML), these genetic screen approaches have been used to identify effector genes with previously unknown roles in AML. These AML-related genes centralize alongside the cellular pathways mediating epigenetics, signaling transduction, transcriptional regulation, and energy metabolism. The shRNA/CRISPR genetic screens also realized an array of candidate genes amenable to pharmaceutical targeting. This review aims to summarize genes, mechanisms, and potential therapeutic strategies found via high-throughput genetic screens in AML. We also discuss the potential of these findings to instruct novel AML therapies for combating drug resistance in this genetically heterogeneous disease.
Collapse
Affiliation(s)
- Qiao Liu
- Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350108, China; (Q.L.); (S.W.)
- Union Clinical Medical College, Fujian Medical University, Fuzhou 350108, China
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
| | - Michelle Garcia
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
- Pomona College, Claremont, CA 91711, USA
| | - Shaoyuan Wang
- Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350108, China; (Q.L.); (S.W.)
- Union Clinical Medical College, Fujian Medical University, Fuzhou 350108, China
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
- Correspondence:
| |
Collapse
|
46
|
Pancho A, Aerts T, Mitsogiannis MD, Seuntjens E. Protocadherins at the Crossroad of Signaling Pathways. Front Mol Neurosci 2020; 13:117. [PMID: 32694982 PMCID: PMC7339444 DOI: 10.3389/fnmol.2020.00117] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/08/2020] [Indexed: 12/25/2022] Open
Abstract
Protocadherins (Pcdhs) are cell adhesion molecules that belong to the cadherin superfamily, and are subdivided into clustered (cPcdhs) and non-clustered Pcdhs (ncPcdhs) in vertebrates. In this review, we summarize their discovery, expression mechanisms, and roles in neuronal development and cancer, thereby highlighting the context-dependent nature of their actions. We furthermore provide an extensive overview of current structural knowledge, and its implications concerning extracellular interactions between cPcdhs, ncPcdhs, and classical cadherins. Next, we survey the known molecular action mechanisms of Pcdhs, emphasizing the regulatory functions of proteolytic processing and domain shedding. In addition, we outline the importance of Pcdh intracellular domains in the regulation of downstream signaling cascades, and we describe putative Pcdh interactions with intracellular molecules including components of the WAVE complex, the Wnt pathway, and apoptotic cascades. Our overview combines molecular interaction data from different contexts, such as neural development and cancer. This comprehensive approach reveals potential common Pcdh signaling hubs, and points out future directions for research. Functional studies of such key factors within the context of neural development might yield innovative insights into the molecular etiology of Pcdh-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Anna Pancho
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tania Aerts
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Manuela D Mitsogiannis
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Eve Seuntjens
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Aghabozorgi AS, Ebrahimi R, Bahiraee A, Tehrani SS, Nabizadeh F, Setayesh L, Jafarzadeh-Esfehani R, Ferns GA, Avan A, Rashidi Z. The genetic factors associated with Wnt signaling pathway in colorectal cancer. Life Sci 2020; 256:118006. [PMID: 32593708 DOI: 10.1016/j.lfs.2020.118006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is a common cancer with poor prognosis and high mortality. There is growing information about the factors involved in the pathogenesis of CRC. However, the knowledge of the predisposing factors is limited. The development of CRC is strongly associated with the Wingless/Integrated (Wnt) signaling pathway. This pathway comprises several major target proteins, including LRP5/6, GSK3β, adenomatous polyposis coli (APC), axis inhibition protein (Axin), and β-catenin. Genetic variations in these components of the Wnt signaling pathway may lead to the activation of β-catenin, potentially increasing the proliferation of colorectal cells. Because of the potentially important role of the Wnt signaling pathway in CRC, we aimed to review the involvement of different mutations in the main downstream proteins of this pathway, including LRP5/6, APC, GSK3β, Axin, and β-catenin. Determination of the genetic risk factors involved in the progression of CRC may lead to novel approaches for the early diagnosis of CRC and the identification of potential therapeutic targets in the treatment of CRC.
Collapse
Affiliation(s)
- Amirsaeed Sabeti Aghabozorgi
- Medical Genetics Research Center, Basic Medical Sciences Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabizadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Zahra Rashidi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
48
|
Glycogen Synthase Kinase 3β in Cancer Biology and Treatment. Cells 2020; 9:cells9061388. [PMID: 32503133 PMCID: PMC7349761 DOI: 10.3390/cells9061388] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase (GSK)3β is a multifunctional serine/threonine protein kinase with more than 100 substrates and interacting molecules. GSK3β is normally active in cells and negative regulation of GSK3β activity via phosphorylation of its serine 9 residue is required for most normal cells to maintain homeostasis. Aberrant expression and activity of GSK3β contributes to the pathogenesis and progression of common recalcitrant diseases such as glucose intolerance, neurodegenerative disorders and cancer. Despite recognized roles against several proto-oncoproteins and mediators of the epithelial–mesenchymal transition, deregulated GSK3β also participates in tumor cell survival, evasion of apoptosis, proliferation and invasion, as well as sustaining cancer stemness and inducing therapy resistance. A therapeutic effect from GSK3β inhibition has been demonstrated in 25 different cancer types. Moreover, there is increasing evidence that GSK3β inhibition protects normal cells and tissues from the harmful effects associated with conventional cancer therapies. Here, we review the evidence supporting aberrant GSK3β as a hallmark property of cancer and highlight the beneficial effects of GSK3β inhibition on normal cells and tissues during cancer therapy. The biological rationale for targeting GSK3β in the treatment of cancer is also discussed at length.
Collapse
|
49
|
Paul R, Luo M, Mo X, Lu J, Yeo SK, Guan JL. FAK activates AKT-mTOR signaling to promote the growth and progression of MMTV-Wnt1-driven basal-like mammary tumors. Breast Cancer Res 2020; 22:59. [PMID: 32493400 PMCID: PMC7268629 DOI: 10.1186/s13058-020-01298-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/20/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease. Hence, stratification of patients based on the subtype of breast cancer is key to its successful treatment. Among all the breast cancer subtypes, basal-like breast cancer is the most aggressive subtype with limited treatment options. Interestingly, we found focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase, is highly overexpressed and activated in basal-like breast cancer. METHODS To understand the role of FAK in this subtype, we generated mice with conditional deletion of FAK and a knock-in mutation in its kinase domain in MMTV-Wnt1-driven basal-like mammary tumors. Tumor initiation, growth, and metastasis were characterized for these mice cohorts. Immunohistochemical and transcriptomic analysis of Wnt1-driven tumors were also performed to elucidate the mechanisms underlying FAK-dependent phenotypes. Pharmacological inhibition of FAK and mTOR in human basal-like breast cancer cell lines was also tested. RESULTS We found that in the absence of FAK or its kinase function, growth and metastasis of the tumors were significantly suppressed. Furthermore, immunohistochemical analyses of cleaved caspase 3 revealed that loss of FAK results in increased tumor cell apoptosis. To further investigate the mechanism by which FAK regulates survival of the Wnt1-driven tumor cells, we prepared an isogenic pair of mammary tumor cells with and without FAK and found that FAK ablation increased their sensitivity to ER stress-induced cell death, as well as reduced tumor cell migration and tumor sphere formation. Comparative transcriptomic profiling of the pair of tumor cells and gene set enrichment analysis suggested mTOR pathway to be downregulated upon loss of FAK. Immunoblot analyses further confirmed that absence of FAK results in reduction of AKT and downstream mTOR pathways. We also found that inhibition of FAK and mTOR pathways both induces apoptosis, indicating the importance of these pathways in regulating cell survival. CONCLUSIONS In summary, our studies show that in a basal-like tumor model, FAK is required for survival of the tumor cells and can serve as a potential therapeutic target.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Basal Cell/genetics
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Cell Movement/physiology
- Cell Proliferation/physiology
- Cell Transformation, Neoplastic
- Disease Models, Animal
- Disease Progression
- Female
- Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors
- Focal Adhesion Protein-Tyrosine Kinases/genetics
- Focal Adhesion Protein-Tyrosine Kinases/metabolism
- Humans
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice, Transgenic
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Tumor Cells, Cultured
- Wnt1 Protein/genetics
- Wnt1 Protein/metabolism
Collapse
Affiliation(s)
- Ritama Paul
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ming Luo
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xueying Mo
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, 45229, USA
| | - Jason Lu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, 45229, USA
| | - Syn Kok Yeo
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
50
|
Ou D, Chen L, He J, Rong Z, Gao J, Li Z, Liu L, Tang F, Li J, Deng Y, Sun L. CDK11 negatively regulates Wnt/β-catenin signaling in the endosomal compartment by affecting microtubule stability. Cancer Biol Med 2020; 17:328-342. [PMID: 32587772 PMCID: PMC7309457 DOI: 10.20892/j.issn.2095-3941.2019.0229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
Objectives: Improper activation of Wnt/β-catenin signaling has been implicated in human diseases. Beyond the well-studied glycogen synthase kinase 3β (GSK3β) and casein kinase 1 (CK1), other kinases affecting Wnt/β-catenin signaling remain to be defined. Methods:To identify the kinases that modulate Wnt/β-catenin signaling, we applied a kinase small interfering RNA (siRNA) library screen approach. Luciferase assays, immunoblotting, and real-time polymerase chain reaction (PCR) were performed to confirm the regulation of the Wnt/β-catenin signaling pathway by cyclin-dependent kinase 11 (CDK11) and to investigate the underlying mechanism. Confocal immunofluorescence, coimmunoprecipitation (co-IP), and scratch wound assays were used to demonstrate colocalization, detect protein interactions, and explore the function of CDK11. Results: CDK11 was found to be a significant candidate kinase participating in the negative control of Wnt/β-catenin signaling. Down-regulation of CDK11 led to the accumulation of Wnt/β-catenin signaling receptor complexes, in a manner dependent on intact adenomatosis polyposis coli (APC) protein. Further analysis showed that CDK11 modulation of Wnt/β-catenin signaling engaged the endolysosomal machinery, and CDK11 knockdown enhanced the colocalization of Wnt/β-catenin signaling receptor complexes with early endosomes and decreased colocalization with lysosomes. Mechanistically, CDK11 was found to function in Wnt/β-catenin signaling by regulating microtubule stability. Depletion of CDK11 down-regulated acetyl-α-tubulin. Moreover, co-IP assays demonstrated that CDK11 interacts with the α-tubulin deacetylase SIRT2, whereas SIRT2 down-regulation in CDK11-depleted cells reversed the accumulation of Wnt/β-catenin signaling receptor complexes. CDK11 was found to suppress cell migration through altered Wnt/β-catenin signaling. Conclusions: CDK11 is a negative modulator of Wnt/β-catenin signaling that stabilizes microtubules, thus resulting in the dysregulation of receptor complex trafficking from early endosomes to lysosomes.
Collapse
Affiliation(s)
- Danmin Ou
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lin Chen
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang He
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhuoxian Rong
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Gao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi Li
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Liyu Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Feiyu Tang
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang Li
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuezhen Deng
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Lunquan Sun
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| |
Collapse
|