1
|
Wu X, Li X, Wang L, Bi X, Zhong W, Yue J, Chin YE. Lysine Deacetylation Is a Key Function of the Lysyl Oxidase Family of Proteins in Cancer. Cancer Res 2024; 84:652-658. [PMID: 38194336 DOI: 10.1158/0008-5472.can-23-2625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/05/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024]
Abstract
Mammalian members of the lysyl oxidase (LOX) family of proteins carry a copper-dependent monoamine oxidase domain exclusively within the C-terminal region, which catalyzes ε-amine oxidation of lysine residues of various proteins. However, recent studies have demonstrated that in LOX-like (LOXL) 2-4 the C-terminal canonical catalytic domain and N-terminal scavenger receptor cysteine-rich (SRCR) repeats domain exhibit lysine deacetylation and deacetylimination catalytic activities. Moreover, the N-terminal SRCR repeats domain is more catalytically active than the C-terminal oxidase domain. Thus, LOX is the third family of lysine deacetylases in addition to histone deacetylase and sirtuin families. In this review, we discuss how the LOX family targets different cellular proteins for deacetylation and deacetylimination to control the development and metastasis of cancer.
Collapse
Affiliation(s)
- Xingxing Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Xue Li
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Peninsular Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China
| | - Luwei Wang
- Peninsular Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China
| | - Xianxia Bi
- Peninsular Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China
| | - Weihong Zhong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Jicheng Yue
- Peninsular Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China
| | - Y Eugene Chin
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Peninsular Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
2
|
Peymanfar Y, Mahjour F, Shrestha N, de la Cueva A, Chen Y, Huang S, Kirsch KH, Han X, Trackman PC. The Lysyl Oxidase G473A Polymorphism Exacerbates Oral Cancer Development in Humans and Mice. Int J Mol Sci 2023; 24:9407. [PMID: 37298359 PMCID: PMC10254048 DOI: 10.3390/ijms24119407] [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: 04/21/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Oral cancer is primarily squamous-cell carcinoma with a 5-year survival rate of approximately 50%. Lysyl oxidase (LOX) participates in collagen and elastin maturation. The propeptide of LOX is released as an 18 kDa protein (LOX-PP) in the extracellular environment by procollagen C-proteinases and has tumor-inhibitory properties. A polymorphism in the propeptide region of LOX (rs1800449, G473A) results in a single amino acid substitution of Gln for Arg. Here we investigated the frequency of rs1800449 in OSCC employing TCGA database resources and determined the kinetics and severity of precancerous oral lesion development in wildtype and corresponding knockin mice after exposure to 4-nitroquinoline oxide (4 NQO) in drinking water. Data show that the OSCC is more common in humans carrying the variant compared to the wildtype. Knockin mice are more susceptible to lesion development. The immunohistochemistry of LOX in mouse tissues and in vitro studies point to a negative feedback pathway of wildtype LOX-PP on LOX expression that is deficient in knockin mice. Data further demonstrate modulations of T cell phenotype in knockin mice toward a more tumor-permissive condition. Data provide initial evidence for rs1800449 as an oral cancer susceptibility biomarker and point to opportunities to better understand the functional mechanism of LOX-PP cancer inhibitory activity.
Collapse
Affiliation(s)
- Yaser Peymanfar
- The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA; (Y.P.); (Y.C.)
| | - Faranak Mahjour
- Department of Translational Dental Medicine, Henry M. Goldman School of Dental Medicine, Boston University, 700 Albany Street, Boston, MA 02118, USA; (F.M.); (N.S.)
| | - Neha Shrestha
- Department of Translational Dental Medicine, Henry M. Goldman School of Dental Medicine, Boston University, 700 Albany Street, Boston, MA 02118, USA; (F.M.); (N.S.)
| | - Ana de la Cueva
- Department of Biochemistry, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; (A.d.l.C.); (K.H.K.)
| | - Ying Chen
- The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA; (Y.P.); (Y.C.)
| | - Shengyuan Huang
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.H.); (X.H.)
| | - Kathrin H. Kirsch
- Department of Biochemistry, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; (A.d.l.C.); (K.H.K.)
| | - Xiaozhe Han
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.H.); (X.H.)
| | - Philip C. Trackman
- The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA; (Y.P.); (Y.C.)
- Department of Translational Dental Medicine, Henry M. Goldman School of Dental Medicine, Boston University, 700 Albany Street, Boston, MA 02118, USA; (F.M.); (N.S.)
| |
Collapse
|
3
|
Kim YS, Lee HJ, Handoko GA, Kim J, Kim SB, Won M, Park JH, Ahn J. Production of a 135-residue long N-truncated human keratinocyte growth factor 1 in Escherichia coli. Microb Cell Fact 2023; 22:98. [PMID: 37170276 PMCID: PMC10173505 DOI: 10.1186/s12934-023-02097-z] [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: 04/27/2022] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Palifermin (trade name Kepivance®) is an amino-terminally truncated recombinant human keratinocyte growth factor 1 (KGF-1) with 140 residues that has been produced using Escherichia coli to prevent and treat oral mucositis following radiation or chemotherapy. In this study, an amino-terminally shortened KGF-1 variant with 135 residues was produced and purified in E. coli, and its cell proliferation activity was evaluated. RESULTS We expressed soluble KGF-1 fused to thioredoxin (TRX) in the cytoplasmic fraction of E. coli to improve its production yield. However, three N-truncated forms (KGF-1 with 140, 138, and 135 residues) were observed after the removal of the TRX protein from the fusion form by cleavage of the human enterokinase light chain C112S (hEKL C112S). The shortest KGF-1 variant, with 135 residues, was expressed by fusion with TRX via the hEKL cleavage site in E. coli and purified at high purity (> 99%). Circular dichroism spectroscopy shows that purified KGF-1135 had a structure similar to that of the KGF-1140 as a random coiled form, and MCF-7 cell proliferation assays demonstrate its biological activity. CONCLUSIONS We identified variations in N-terminus-truncated KGF-1 and selected the most stable form. Furthermore, by a simple two-step purification, highly purified KGF-1135 was obtained that showed biological activity. These results demonstrate that KGF-1135 may be considered an alternative protein to KGF-1.
Collapse
Affiliation(s)
- Young Su Kim
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
| | - Hye-Jeong Lee
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
| | - Gabriella Aphrodita Handoko
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Jaehui Kim
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Seong-Bo Kim
- Bio-Living Engineering Major, Global Leaders College, Yonsei University, 50 Yonsei-ro, Shinchon-dong, Seodaemun-gu, Seoul, 03722, Korea
| | - Minho Won
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea.
| | - Jung-Ho Park
- Bio-Evaluation Center, KRIBB, Cheongju, 20736, Republic of Korea.
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| | - Jungoh Ahn
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea.
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
4
|
Trackman PC, Peymanfar Y, Roy S. Functions and Mechanisms of Pro-Lysyl Oxidase Processing in Cancers and Eye Pathologies with a Focus on Diabetic Retinopathy. Int J Mol Sci 2022; 23:5088. [PMID: 35563478 PMCID: PMC9105217 DOI: 10.3390/ijms23095088] [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: 03/30/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Lysyl oxidases are multifunctional proteins derived from five lysyl oxidase paralogues (LOX) and lysyl oxidase-like 1 through lysyl oxidase-like 4 (LOXL1-LOXL4). All participate in the biosynthesis of and maturation of connective tissues by catalyzing the oxidative deamination of lysine residues in collagens and elastin, which ultimately results in the development of cross-links required to function. In addition, the five LOX genes have been linked to fibrosis and cancer when overexpressed, while tumor suppression by the propeptide derived from pro-LOX has been documented. Similarly, in diabetic retinopathy, LOX overexpression, activity, and elevated LOX propeptide have been documented. The proteolytic processing of pro-forms of the respective proteins is beginning to draw attention as the resultant peptides appear to exhibit their own biological activities. In this review we focus on the LOX paralogue, and what is known regarding its extracellular biosynthetic processing and the still incomplete knowledge regarding the activities and mechanisms of the released lysyl oxidase propeptide (LOX-PP). In addition, a summary of the roles of both LOX and LOX-PP in diabetic retinopathy, and brief mentions of the roles for LOX and closely related LOXL1 in glaucoma, and keratoconus, respectively, are included.
Collapse
Affiliation(s)
- Philip C. Trackman
- The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA;
- Department of Translational Dental Medicine, Boston University Henry M Goldman School of Dental Medicine, 700 Albany Street, Boston, MA 02118, USA
| | - Yaser Peymanfar
- The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA;
| | - Sayon Roy
- Department of Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
5
|
Liu S, Medina-Perez P, Ha-Thi MC, Wieland A, Stecklum M, Hoffmann J, Tchernitsa O, Sers C, Schäfer R. Rapid testing of candidate oncogenes and tumour suppressor genes in signal transduction and neoplastic transformation. Adv Biol Regul 2021; 83:100841. [PMID: 34866037 DOI: 10.1016/j.jbior.2021.100841] [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: 11/05/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/18/2022]
Abstract
The COSMIC database (version 94) lists 576 genes in the Cancer Gene Census which have a defined function as drivers of malignancy (oncogenes) or as tumour suppressors (Tier 1). In addition, there are 147 genes with similar functions, but which are less well characterised (Tier 2). Furthermore, next-generation sequencing projects in the context of precision oncology activities are constantly discovering new ones. Since cancer genes differ from their wild-type precursors in numerous molecular and biochemical properties and exert significant differential effects on downstream processes, simple assays that can uncover oncogenic or anti-oncogenic functionality are desirable and may precede more sophisticated analyses. We describe simple functional assays for PTPN11 (protein-tyrosine phosphatase, non-receptor-type 11)/SHP2 mutants, which are typically found in RASopathies and exhibit potential oncogenic activity. We have also designed a functional test for lysyl oxidase (LOX), a prototypical class II tumour suppressor gene whose loss of function may contribute to neoplastic transformation by RAS oncogenes. Moreover, we applied this test to analyse three co-regulated, RAS-responsive genes for transformation-suppressive activity. The integration of these tests into systems biology studies will contribute to a better understanding of cellular networks in cancer.
Collapse
Affiliation(s)
- Sha Liu
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Paula Medina-Perez
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Minh-Cam Ha-Thi
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Anja Wieland
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Maria Stecklum
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, D-13125, Berlin-Buch, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, D-13125, Berlin-Buch, Germany
| | - Oleg Tchernitsa
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Christine Sers
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany
| | - Reinhold Schäfer
- Laboratory of Molecular Tumour Pathology and Cancer Systems Biology, Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany; Charité Comprehensive Cancer Center Berlin, Germany.
| |
Collapse
|
6
|
Targeting Lysyl Oxidase Family Meditated Matrix Cross-Linking as an Anti-Stromal Therapy in Solid Tumours. Cancers (Basel) 2021; 13:cancers13030491. [PMID: 33513979 PMCID: PMC7865543 DOI: 10.3390/cancers13030491] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary To improve efficacy of solid cancer treatment, efforts have shifted towards targeting both the cancer cells and the surrounding tumour tissue they grow in. The lysyl oxidase (LOX) family of enzymes underpin the fibrotic remodeling of the tumour microenvironment to promote both cancer growth, spread throughout the body and modulate response to therapies. This review examines how the lysyl oxidase family is involved in tumour development, how they can be targeted, and their potential as diagnostic and prognostic biomarkers in solid tumours. Abstract The lysyl oxidase (LOX) family of enzymes are a major driver in the biogenesis of desmoplastic matrix at the primary tumour and secondary metastatic sites. With the increasing interest in and development of anti-stromal therapies aimed at improving clinical outcomes of cancer patients, the Lox family has emerged as a potentially powerful clinical target. This review examines how lysyl oxidase family dysregulation in solid cancers contributes to disease progression and poor patient outcomes, as well as an evaluation of the preclinical landscape of LOX family targeting therapeutics. We also discuss the suitability of the LOX family as a diagnostic and/or prognostic marker in solid tumours.
Collapse
|
7
|
Yang N, Cao DF, Yin XX, Zhou HH, Mao XY. Lysyl oxidases: Emerging biomarkers and therapeutic targets for various diseases. Biomed Pharmacother 2020; 131:110791. [PMID: 33152948 DOI: 10.1016/j.biopha.2020.110791] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic targeting of extracellular proteins has attracted huge attention in treating human diseases. The lysyl oxidases (LOXs) are a family of secreted copper-dependent enzymes which initiate the covalent crosslinking of collagen and elastin fibers in the extracellular microenvironment, thereby facilitating extracellular matrix (ECM) remodeling and ECM homeostasis. Apart from ECM-dependent roles, LOXs are also involved in other biological processes such as epithelial-to-mesenchymal transition (EMT) and transcriptional regulation, especially following hypoxic stress. Dysregulation of LOXs is found to underlie the onset and progression of multiple pathologies, such as carcinogenesis and cancer metastasis, fibrotic diseases, neurodegeneration and cardiovascular diseases. In this review, we make a comprehensive summarization of clinical and experimental evidences that support roles of for LOXs in disease pathology and points out LOXs as promising therapeutic targets for improving prognosis. Additionally, we also propose that LOXs reshape cell-ECM interaction or cell-cell interaction due to ECM-dependent and ECM-independent roles for LOXs. Therapeutic intervention of LOXs may have advantages in the maintenance of communication between ECM and cell or intercellular signaling, finally recovering organ function.
Collapse
Affiliation(s)
- Nan Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Dan-Feng Cao
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan, 410013, PR China
| | - Xi-Xi Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China.
| |
Collapse
|
8
|
Kim D, Lee D, Trackman PC, Roy S. Effects of High Glucose-Induced Lysyl Oxidase Propeptide on Retinal Endothelial Cell Survival: Implications for Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1945-1952. [PMID: 31537300 PMCID: PMC6880772 DOI: 10.1016/j.ajpath.2019.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 01/19/2023]
Abstract
Diabetic retinopathy (DR) is characterized by apoptotic cell loss in the retinal vasculature. Lysyl oxidase propeptide (LOX-PP), released during LOX processing, has been implicated in promoting apoptosis in various diseased tissues. However, its role in the development and progression of DR is unknown. We investigated whether high glucose (HG) or diabetes alters LOX-PP expression and thereby influences AKT pathway and affects retinal endothelial cell survival. Rat retinal endothelial cells were grown in normal medium, normal medium and exposed to recombinant LOX-PP (rLOX-PP) or HG medium and examined for LOX-PP expression, AKT and caspase-3 activation. Similarly, rats intravitreally injected with rLOX-PP were examined for changes in retinal LOX-PP levels, AKT phosphorylation, and the number of acellular capillaries and pericyte loss compared with those of control diabetic and nondiabetic rats. Results indicate that HG up-regulates LOX-PP expression and reduces AKT activation. In addition, cells exposed to rLOX-PP alone exhibited increased apoptosis concomitant with decreased AKT phosphorylation. In retinas of diabetic rats, increased LOX-PP level, decreased AKT phosphorylation, and increased number of acellular capillaries and pericyte loss compared with those of nondiabetic rats were observed. Of interest, similar changes were noted in the retinas of rats injected with rLOX-PP. Findings from this study suggest that hyperglycemia-induced LOX-PP overexpression may contribute to retinal vascular cell loss associated with DR.
Collapse
Affiliation(s)
- Dongjoon Kim
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Dayeun Lee
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Philip C Trackman
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts
| | - Sayon Roy
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
9
|
Kielosto M, Eriksson J, Nummela P, Yin M, Hölttä E. Divergent roles of lysyl oxidase family members in ornithine decarboxylase- and RAS-transformed mouse fibroblasts and human melanoma cells. Oncotarget 2018; 9:37733-37752. [PMID: 30701028 PMCID: PMC6340875 DOI: 10.18632/oncotarget.26508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that proto-oncoprotein c-Jun is activated in ornithine decarboxylase (ODC)- and RAS-transformed mouse fibroblasts, and that the transformed morphology of these cells can be reversed by expressing the transactivation domain deletion mutant of c-Jun (TAM67). Here, we found that lysyl oxidase (Lox), encoding an extracellular matrix-modifying enzyme, is downregulated in a c-Jun-dependent manner in ODC-transformed fibroblasts (Odc cells). In addition to Lox, the Lox family members Lox-like 1 and 3 (Loxl1 and Loxl3) were found to be downregulated in Odc as well as in RAS-transformed fibroblasts (E4), whereas Lox-like 4 (Loxl4) was upregulated in Odc and downregulated in E4 cells compared to normal N1 fibroblasts. Tetracycline-regulatable LOX re-expression in Odc cells led to inhibition of cell growth and invasion in three-dimensional Matrigel in an activity-independent manner. On the contrary, LOX and especially LOXL2, LOXL3, and LOXL4 were found to be upregulated in several human melanoma cell lines, and LOX inhibitor B-aminopropionitrile inhibited the invasive growth of these cells particularly when co-cultured with fibroblasts in Matrigel. Knocking down the expression of LOX and especially LOXL2 in melanoma cells almost completely abrogated the invasive growth capability. Further, LOXL2 was significantly upregulated in clinical human primary melanomas compared to benign nevi, and high expression of LOXL2 in primary melanomas was associated with formation of metastases and shorter survival of patients. Thus, our studies reveal that inactive pro-LOX (together with Lox propeptide) functions as a tumor suppressor in ODC- and RAS-transformed murine fibroblasts by inhibiting cell growth and invasion, and active LOX and LOXL2 as tumor promoters in human melanoma cells by promoting their invasive growth.
Collapse
Affiliation(s)
- Mari Kielosto
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Johanna Eriksson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Pirjo Nummela
- Department of Pathology, University of Helsinki, Helsinki, Finland.,Current address: University of Helsinki, Genome-Scale Biology Research Program, Helsinki, Finland
| | - Miao Yin
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Erkki Hölttä
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Saxena D, Mahjour F, Findlay A, Mously E, Kantarci A, Trackman P. Multiple Functions of Lysyl Oxidase Like-2 in Oral Fibroproliferative Processes. J Dent Res 2018; 97:1277-1284. [PMID: 29787337 PMCID: PMC6151912 DOI: 10.1177/0022034518775971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gingival overgrowth is a side effect of certain medications, including calcium channel blockers, cyclosporin A, and phenytoin. Phenytoin-induced gingival overgrowth is fibrotic. Lysyl oxidases are extracellular enzymes that are required for biosynthetic cross-linking of collagens, and members of this enzyme family are upregulated in fibrosis. Previous studies in humans and in a mouse model of phenytoin-induced gingival overgrowth have shown that LOXL2 is elevated in the epithelium and connective tissue in gingival overgrowth tissues and not in normal tissues. Here, using a novel LOXL2 isoform-selective inhibitor and knockdown studies in loss- and gain-of-function studies, we investigated roles for LOXL2 in promoting cultures of human gingival fibroblasts to proliferate and to accumulate collagen. Data indicate that LOXL2 stimulates gingival fibroblast proliferation, likely by a platelet-derived growth factor B receptor-mediated mechanism. Moreover, collagen accumulation was stimulated by LOXL2 enzyme and inhibited by LOXL2 inhibitor or gene knockdown. These studies suggest that LOXL2 could serve as a potential therapeutic target to address oral fibrotic conditions.
Collapse
Affiliation(s)
- D. Saxena
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - F. Mahjour
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | | | - E.A. Mously
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
- College of Dentistry, Taibah University,
Medina, Saudi Arabia
| | | | - P.C. Trackman
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
11
|
Nareshkumar RN, Sulochana KN, Coral K. Inhibition of angiogenesis in endothelial cells by Human Lysyl oxidase propeptide. Sci Rep 2018; 8:10426. [PMID: 29993014 PMCID: PMC6041307 DOI: 10.1038/s41598-018-28745-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/05/2018] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis is a critical process involved in normal physiology. Pathological angiogenesis is observed in vascular diseases and neoplasia. The propeptide domain of LOX (LOX-PP) has been shown to inhibit tumorigenesis in various cancers. In this study, we explored the role of both overexpressed and recombinant LOX-PP in naïve human umbilical vein endothelial cell with the addition of vascular endothelial growth factor (VEGF). Primarily, we observed a significant reduction in the angiogenesis signaling pathways upon LOX-PP overexpression by proteomic analysis. Further functional analysis showed that the VEGF induced cell proliferation, migration, adhesion and tube formation was inhibited by LOX-PP. Moreover, LOX-PP arrested cells at S-phase, reduced F-actin levels and decreased phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase (ERK). The anti-angiogenic effect of LOX-PP was further confirmed by the reduction in the vascular network formation in chick chorioallantoic membrane (CAM). These results indicate that inhibition of angiogenesis events is not only achieved by overexpressing LOX-PP but also by addition of rLOX-PP. Taken together our findings discovered the anti-angiogenic role of LOX-PP in endothelial cells which suggests that harnessing this potential can be a promising strategy to inhibit angiogenesis.
Collapse
Affiliation(s)
- Ragavachetty Nagaraj Nareshkumar
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, 41, College road, Chennai, India.,School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | | | - Karunakaran Coral
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, 41, College road, Chennai, India.
| |
Collapse
|
12
|
de la Cueva A, Emmerling M, Lim SL, Yang S, Trackman PC, Sonenshein GE, Kirsch KH. A polymorphism in the lysyl oxidase propeptide domain accelerates carcinogen-induced cancer. Carcinogenesis 2018; 39:921-930. [PMID: 29579155 PMCID: PMC6692853 DOI: 10.1093/carcin/bgy045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/17/2018] [Accepted: 03/20/2018] [Indexed: 01/19/2023] Open
Abstract
The propeptide (LOX-PP) domain of the lysyl oxidase proenzyme was shown to inhibit the transformed phenotype of breast, lung and pancreatic cells in culture and the formation of Her2/neu-driven breast cancer in a xenograft model. A single nucleotide polymorphism (SNP, rs1800449) positioned in a highly conserved region of LOX-PP results in an Arg158Gln substitution (humans). This arginine (Arg)→glutamine (Gln) substitution profoundly impaired the ability of LOX-PP to inhibit the invasive phenotype and xenograft tumor formation. To study the effect of the SNP in vivo, here we established a knock in (KI) mouse line (LOX-PPGln mice) expressing an Arg152Gln substitution corresponding to the human Arg158Gln polymorphism. Breast cancer was induced in wild-type (WT) and LOX-PPGln female mice beginning at 6 weeks of age by treatment with 7,12-dimethylbenz(a)anthracene (DMBA) in combination with progesterone. Time course analysis of tumor development demonstrated earlier tumor onset and shorter overall survival in LOX-PPGln versus WT mice. To further compare the tumor burden in WT and LOX-PPGln mice, inguinal mammary glands from both groups of mice were examined for microscopic lesion formation. LOX-PPGln glands contained more lesions (9.6 versus 6.9 lesions/#4 bilateral). In addition, more DMBA-treated LOX-PPGln mice had increased leukocyte infiltrations in their livers and were moribund compared with DMBA-treated WT mice. Thus, these data indicate that the Arg→Gln substitution in LOX-PP could be an important marker associated with a more aggressive cancer phenotype and that this KI model is ideal for further mechanistic studies regarding the tumor suppressor function of LOX-PP.
Collapse
Affiliation(s)
- Ana de la Cueva
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Michael Emmerling
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Sarah L Lim
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Shi Yang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Philip C Trackman
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Gail E Sonenshein
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Kathrin H Kirsch
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Li T, Wu C, Gao L, Qin F, Wei Q, Yuan J. Lysyl oxidase family members in urological tumorigenesis and fibrosis. Oncotarget 2018; 9:20156-20164. [PMID: 29732010 PMCID: PMC5929453 DOI: 10.18632/oncotarget.24948] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/11/2018] [Indexed: 02/05/2023] Open
Abstract
Lysyl oxidase (LOX) is an extracellular copper-dependent monoamine oxidase that catalyzes crosslinking of soluble collagen and elastin into insoluble, mature fibers. Lysyl oxidase-like proteins (LOXL), LOX isozymes with partial structural homology, exhibit similar catalytic activities. This review summarizes recent findings describing the roles of LOX family members in urological cancers and fibrosis. LOX/LOXL play key roles in extracellular matrix stability and integrity, which is essential for normal female pelvic floor function. LOX/LOXL inhibition may reverse kidney fibrosis and ischemic priapism. LOX and LOXL2 reportedly promote kidney carcinoma tumorigenesis, while LOX, LOXL1 and LOXL4 suppress bladder cancer growth. Multiple studies agree that the LOX propeptide may suppress tumor growth, but the role of LOX in prostate cancer remains controversial. Further studies are needed to clarify the exact effects and mechanism of LOX/LOXL on urological malignancies.
Collapse
Affiliation(s)
- Tao Li
- The Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changjing Wu
- The Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liang Gao
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Qin
- The Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiuhong Yuan
- The Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Johnston KA, Lopez KM. Lysyl oxidase in cancer inhibition and metastasis. Cancer Lett 2018; 417:174-181. [DOI: 10.1016/j.canlet.2018.01.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/20/2017] [Accepted: 01/02/2018] [Indexed: 11/16/2022]
|
15
|
Trackman PC. Functional importance of lysyl oxidase family propeptide regions. J Cell Commun Signal 2017; 12:45-53. [PMID: 29086201 DOI: 10.1007/s12079-017-0424-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
The lysyl oxidase family of proteins is primarily known for its critical role in catalyzing extracellular oxidative deamination of hydroxylysine and lysine residues in collagens, and lysine residues in elastin required for connective tissue structure and function. Lysyl oxidases have additional important biological functions in health and disease. While the enzyme domains are highly conserved, the propeptide regions are less uniform, and have biological activity, some of which are independent of their respective enzymes. This review summarizes what has been published regarding the functions of the propeptide regions of this family of proteins in the context of extracellular matrix biosynthesis, fibrosis and cancer biology. Although much has been learned, there is a need for greater attention to structure/function relationships and mechanisms to more fully understand these multifunctional proteins.
Collapse
Affiliation(s)
- Philip C Trackman
- Henry M. Goldman School of Dental Medicine, Department of Molecular and Cell Biology, Boston University, 700 Albany Street, W-201, Boston, MA, 02118, USA.
| |
Collapse
|
16
|
Alsaqer SF, Tashkandi MM, Kartha VK, Yang YT, Alkheriji Y, Salama A, Varelas X, Kukuruzinska M, Monti S, Bais MV. Inhibition of LSD1 epigenetically attenuates oral cancer growth and metastasis. Oncotarget 2017; 8:73372-73386. [PMID: 29088714 PMCID: PMC5650269 DOI: 10.18632/oncotarget.19637] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 07/14/2017] [Indexed: 01/26/2023] Open
Abstract
Lysine-specific demethylase 1 (LSD1) is a nuclear histone demethylase and a member of the amine oxidase (AO) family. LSD1 is a flavin-containing AO that specifically catalyzes the demethylation of mono- and di-methylated histone H3 lysine 4 through an FAD-dependent oxidative reaction. LSD1 is inappropriately upregulated in lung, liver, brain and esophageal cancers, where it promotes cancer initiation, progression, and metastasis. However, unlike other lysine-specific demethylases, the role and specific targets of LSD1 in oral squamous cell carcinoma (OSCC) pathogenesis remain unknown. We show that LSD1 protein expression was increased in malignant OSCC tissues in a clinical tissue microarray, and its expression correlated with progressive tumor stages. In an orthotopic oral cancer mouse model, LSD1 overexpression in aggressive HSC-3 cells promoted metastasis whereas knockdown of LSD1 inhibited tumor spread, suggesting that LSD1 is a key regulator of OSCC metastasis. Pharmacological inhibition of LSD1 using a specific small molecule inhibitor, GSK-LSD1, down-regulated EGF signaling pathway. Further, GSK-LSD1 attenuates CTGF/CCN2, MMP13, LOXL4 and vimentin expression but increased E-cadherin expression in pre-existing, patient-derived tonsillar OSCC xenografts. Similarly, GSK-LSD1 inhibited proliferation and CTGF expression in mesenchymal cells, including myoepithelial cells and osteosarcoma cells. In addition, gene set enrichment analysis revealed that GSK-LSD1 increased p53 expression and apoptosis while inhibiting c-myc, β-catenin and YAP-induced oncogenic transcriptional networks. These data reveal that aberrant LSD1 activation regulates key OSCC microenvironment and EMT promoting factors, including CTGF, LOXL4 and MMP13.
Collapse
Affiliation(s)
- Saqer F Alsaqer
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Mustafa M Tashkandi
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Vinay K Kartha
- Bioinformatics Program, Boston University, Boston, MA, USA.,Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Ya-Ting Yang
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Yazeed Alkheriji
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Andrew Salama
- Department of Oral and Maxillofacial Surgery, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Maria Kukuruzinska
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - Stefano Monti
- Bioinformatics Program, Boston University, Boston, MA, USA.,Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Manish V Bais
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
17
|
Sánchez-Morgan N, Kirsch KH, Trackman PC, Sonenshein GE. UXT Is a LOX-PP Interacting Protein That Modulates Estrogen Receptor Alpha Activity in Breast Cancer Cells. J Cell Biochem 2017; 118:2347-2356. [PMID: 28106301 DOI: 10.1002/jcb.25893] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023]
Abstract
The lysyl oxidase proenzyme propeptide region (LOX-PP) is a tumor suppressor protein whose mechanism of action is not completely understood. Here, the Ubiquitously expressed Transcript (UXT) was identified in a yeast two-hybrid assay with LOX-PP as bait and confirmed as a novel LOX-PP associating protein. UXT, a prefoldin-like protein, is ubiquitous in human and mouse. Since UXT modulates androgen receptor transcriptional activity in prostate cancer, we studied its role in breast cancer. Breast tumors and derived cell lines overexpressed UXT. UXT was able to associate with the estrogen receptor alpha (ER) and decrease its transcriptional activity and target gene expression. Conversely, UXT knockdown increased ER element-dependent transcriptional activity. Ectopic LOX-PP relocalized UXT to the cytoplasm and decreased its stability. UXT ubiquitination and depletion in the presence of LOX-PP was rescued by a proteasomal inhibitor. In summary, proteasome-mediated turnover of UXT upon interaction with LOX-PP releases repression of ER transcriptional activity. J. Cell. Biochem. 118: 2347-2356, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nuria Sánchez-Morgan
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Kathrin H Kirsch
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Philip C Trackman
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts
| | - Gail E Sonenshein
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
18
|
Major Action of Endogenous Lysyl Oxidase in Clear Cell Renal Cell Carcinoma Progression and Collagen Stiffness Revealed by Primary Cell Cultures. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2473-85. [PMID: 27449199 DOI: 10.1016/j.ajpath.2016.05.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 04/27/2016] [Accepted: 05/23/2016] [Indexed: 11/20/2022]
Abstract
Human clear cell renal cell carcinoma (ccRCC) is therapy resistant; therefore, it is worthwhile studying in depth the molecular aspects of its progression. In ccRCC the biallelic inactivation of the VHL gene leads to stabilization of hypoxia-inducible factors (HIFs). Among the targets of HIF-1α transcriptional activity is the LOX gene, which codes for the inactive proenzyme (Pro-Lox) from which, after extracellular secretion and proteolysis, derives the active enzyme (Lox) and the propeptide (Lox-PP). By increasing stiffness of extracellular matrix by collagen crosslinking, Lox promotes tumor progression and metastasis. Lox and Lox-PP can reenter the cells where Lox promotes cell proliferation and invasion, whereas Lox-PP acts as tumor suppressor because of its Ras recision and apoptotic activity. Few data are available concerning LOX in ccRCC. Using an in vitro model of ccRCC primary cell cultures, we performed, for the first time in ccRCC, a detailed study of endogenous LOX and also investigated their transcriptomic profile. We found that endogenous LOX is overexpressed in ccRCC, is involved in a positive-regulative loop with HIF-1α, and has a major action on ccRCC progression through cellular adhesion, migration, and collagen matrix stiffness increment; however, the oncosuppressive action of Lox-PP was not found to prevail. These findings may suggest translational approaches for new therapeutic strategies in ccRCC.
Collapse
|
19
|
Wang G, Shen Y, Cheng G, Bo H, Lin J, Zheng M, Li J, Zhao Y, Li W. Lysyl Oxidase Gene G473A Polymorphism and Cigarette Smoking in Association with a High Risk of Lung and Colorectal Cancers in a North Chinese Population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E635. [PMID: 27367711 PMCID: PMC4962176 DOI: 10.3390/ijerph13070635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/19/2022]
Abstract
The relationship among the lysyl oxidase (LOX) G473A single nucleotide polymorphism (SNP), cigarette smoking and lung, colorectal, colon and rectum cancer susceptibility was studied in 200 cases of lung cancer, 335 cases of colorectal cancer including 130 cases of colon cancer and 205 cases of rectum cancer, and 335 healthy people in Tangshan, China. Peripheral blood DNA samples were collected, DNA sequencing and polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) performed, followed by multivariate logistic regression analysis. In comparison to LOX473GG genotype carriers, individuals with LOX473AA exhibited a higher susceptibility to lung, colon-rectum, colon, and rectum cancers with OR values amounting to 3.84-, 2.74-, 2.75-, and 2.74-fold of the control, respectively. In the LOX 473AA-positive population, females were more susceptible than males to carcinogenesis with OR values (female vs. male): 5.25 vs. 3.23, 2.29 vs. 1.51, 2.27 vs. 1.45, and 2.25 vs. 1.53, respectively, for lung, colon-rectum combined, colon, and rectum cancers. LOX G473A polymorphism apparently elevated human sensitivity to cigarette smoking carcinogens for eliciting cancers in the lung and colon only. Thus, LOX G473A polymorphism positively correlates with carcinogenesis and it may be used as an ideal intrinsic biomarker for prediction or diagnosis of carcinogenesis in humans.
Collapse
Affiliation(s)
- Guoli Wang
- The Collage of Public Health, North China University of Science and Technology, Tangshan 063000, China.
| | - Yanqing Shen
- The Collage of Public Health, North China University of Science and Technology, Tangshan 063000, China.
| | - Guang Cheng
- The Clinic Medical College, North China University of Science and Technology, Tangshan 063000, China.
| | - Haimei Bo
- The Clinic Medical College, North China University of Science and Technology, Tangshan 063000, China.
| | - Jia Lin
- The College of Life Science, North China University of Science and Technology, Tangshan 063000, China.
| | - Maogen Zheng
- The Clinic Medical College, North China University of Science and Technology, Tangshan 063000, China.
| | - Jianmin Li
- The Clinic Medical College, North China University of Science and Technology, Tangshan 063000, China.
| | - Yinzhi Zhao
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Wande Li
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
20
|
Trackman PC. Lysyl Oxidase Isoforms and Potential Therapeutic Opportunities for Fibrosis and Cancer. Expert Opin Ther Targets 2016; 20:935-45. [PMID: 26848785 DOI: 10.1517/14728222.2016.1151003] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The lysyl oxidase family of enzymes is classically known as being required for connective tissue maturation by oxidizing lysine residues in elastin and lysine and hydroxylysine residues in collagen precursors. The resulting aldehydes then participate in cross-link formation, which is required for normal connective tissue integrity. These enzymes have biological functions that extend beyond this fundamental biosynthetic role, with contributions to angiogenesis, cell proliferation, and cell differentiation. Dysregulation of lysyl oxidases occurs in multiple pathologies including fibrosis, primary and metastatic cancers, and complications of diabetes in a variety of tissues. AREAS COVERED This review summarizes the major findings of novel roles for lysyl oxidases in pathologies, and highlights some of the potential therapeutic approaches that are in development and which stem from these new findings. EXPERT OPINION Fundamental questions remain regarding the mechanisms of novel biological functions of this family of proteins, and regarding functions that are independent of their catalytic enzyme activity. However, progress is underway in the development of isoform-specific pharmacologic inhibitors, potential therapeutic antibodies and gaining an increased understanding of both tumor suppressor and metastasis promotion activities. Ultimately, this is likely to lead to novel therapeutic agents.
Collapse
Affiliation(s)
- Philip C Trackman
- a Department of Molecular and Cell Biology , Boston University, Henry M. Goldman School of Dental Medicine , Boston , MA , USA
| |
Collapse
|
21
|
Alsulaiman M, Bais MV, Trackman PC. Lysyl oxidase propeptide stimulates osteoblast and osteoclast differentiation and enhances PC3 and DU145 prostate cancer cell effects on bone in vivo. J Cell Commun Signal 2015; 10:17-31. [PMID: 26627907 DOI: 10.1007/s12079-015-0311-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/18/2015] [Indexed: 12/11/2022] Open
Abstract
Lysyl oxidase pro-enzyme is secreted by tumor cells and normal cells as a 50 kDa pro-enzyme into the extracellular environment where it is cleaved into the ~30 kDa mature enzyme (LOX) and 18 kDa pro-peptide (LOX-PP). Extracellular LOX enzyme activity is required for normal collagen and elastin extracellular cross-linking and maturation of the extracellular matrix. Extracellular LOX-PP acts as a tumor suppressor and can re-enter cells from the extracellular environment to induce its effects. The underlying hypothesis is that LOX-PP has the potential to promote bone cell differentiation, while inhibiting cancer cell effects in bone. Here we investigate the effect of LOX-PP on bone marrow cell proliferation and differentiation towards osteoblasts or osteoclasts, and LOX-PP modulation of prostate cancer cell conditioned media-induced alterations of proliferation and differentiation of bone marrow cells in vitro. Effects of overexpression of rLOX-PP in DU145 and PC3 prostate cancer cell lines on bone structure in vivo after intramedullary injections were determined. Data show that prostate cancer cell conditioned media inhibited osteoblast differentiation in bone marrow-derived cells, which was reversed by rLOX-PP treatment. Prostate cancer conditioned media stimulated osteoclast differentiation which was further enhanced by rLOX-PP treatment. rLOX-PP stimulated osteoclast differentiation by inhibiting OPG expression, up-regulating CCN2 expression, and increasing osteoclast fusion. In vivo studies indicate that rLOX-PP expression by PC3 cells implanted into the tibia of mice further enhanced PC3 cell ability to resorb bone, while rLOX-PP expression in DU145 cells resulted in non-significant increases in net bone formation. rLOX-PP enhances both osteoclast and osteoblast differentiation. rLOX-PP may serve to enhance coupling interactions between osteoclasts and osteoblasts helping to maintain a normal bone turnover in health, while contributing to bone abnormalities in disease.
Collapse
Affiliation(s)
- Mona Alsulaiman
- Henry M. Goldman School of Dental Medicine, Department of Molecular and Cell Biology, Boston University, 700 Albany Street, W-201, Boston, MA, 02118, USA
| | - Manish V Bais
- Henry M. Goldman School of Dental Medicine, Department of Molecular and Cell Biology, Boston University, 700 Albany Street, W-201, Boston, MA, 02118, USA
| | - Philip C Trackman
- Henry M. Goldman School of Dental Medicine, Department of Molecular and Cell Biology, Boston University, 700 Albany Street, W-201, Boston, MA, 02118, USA.
| |
Collapse
|
22
|
Cidre-Aranaz F, Alonso J. EWS/FLI1 Target Genes and Therapeutic Opportunities in Ewing Sarcoma. Front Oncol 2015; 5:162. [PMID: 26258070 PMCID: PMC4507460 DOI: 10.3389/fonc.2015.00162] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/06/2015] [Indexed: 12/31/2022] Open
Abstract
Ewing sarcoma is an aggressive bone malignancy that affect children and young adults. Ewing sarcoma is the second most common primary bone malignancy in pediatric patients. Although significant progress has been made in the treatment of Ewing sarcoma since it was first described in the 1920s, in the last decade survival rates have remained unacceptably invariable, thus pointing to the need for new approaches centered in the molecular basis of the disease. Ewing sarcoma driving mutation, EWS–FLI1, which results from a chromosomal translocation, encodes an aberrant transcription factor. Since its first characterization in 1990s, many molecular targets have been described to be regulated by this chimeric transcription factor. Their contribution to orchestrate Ewing sarcoma phenotype has been reported over the last decades. In this work, we will focus on the description of a selection of EWS/FLI1 targets, their functional role, and their potential clinical relevance. We will also discuss their role in other types of cancer as well as the need for further studies to be performed in order to achieve a broader understanding of their particular contribution to Ewing sarcoma development.
Collapse
Affiliation(s)
- Florencia Cidre-Aranaz
- Unidad de Tumores Sólidos Infantiles, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III , Madrid , Spain
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III , Madrid , Spain
| |
Collapse
|
23
|
Abstract
Metastasis is the main reason for cancer-associated deaths and therapies are desperately needed to target the progression of cancer. Lysyl oxidase (LOX) plays a pivotal role in cancer progression, including metastasis, and is therefore is an attractive therapeutic target. In this review we will breakdown the process of cancer progression and the various roles that LOX plays has in the advancement of cancer. We will highlight why LOX is an exciting therapeutic target for the future.
Collapse
Affiliation(s)
- Lara Perryman
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, Copenhagen 2200, Denmark
| | | |
Collapse
|
24
|
Bais MV, Ozdener GB, Sonenshein GE, Trackman PC. Effects of tumor-suppressor lysyl oxidase propeptide on prostate cancer xenograft growth and its direct interactions with DNA repair pathways. Oncogene 2015; 34:1928-37. [PMID: 24882580 PMCID: PMC4254378 DOI: 10.1038/onc.2014.147] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 04/01/2014] [Accepted: 04/18/2014] [Indexed: 01/27/2023]
Abstract
Lysyl oxidase (LOX) is a multifunctional protein required for normal collagen and elastin biosynthesis and maturation. In addition, LOX has complex roles in cancer in which the lysyl oxidase propeptide (LOX-PP) domain of secreted pro-LOX has tumor-suppressor activity, while the active enzyme promotes metastasis. In prostate cancer cell lines, recombinant LOX-PP (rLOX-PP) inhibits the growth of PC3 cells in vitro by mechanisms that were not characterized, while in DU145 cells rLOX-PP targeted fibroblast growth factor signaling. Because rLOX-PP can enhance effects of a genotoxic chemotherapeutic on breast cancer cell apoptosis, we reasoned that rLOX-PP could target DNA repair pathways typically elevated in cancer. Here we demonstrate for the first time that rLOX-PP inhibits prostate xenograft growth in vivo and that activating phosphorylations of the key DNA repair molecules ataxia-telangiectasia mutated (ATM) and checkpoint kinase 2 (CHK2) are inhibited by rLOX-PP expression in vivo. In addition, in vitro studies showed that rLOX-PP inhibits radiation-induced activating phosphorylations of ATM and CHK2 and that exogenously added rLOX-PP protein can localize to the nucleus in both DU145 and PC3 cells. rLOX-PP pull-down studies resulted in detection of a protein complex with the nuclear DNA repair regulator MRE11 in both cell lines, and rLOX-PP localized to radiation-induced nuclear DNA repair foci. Finally, rLOX-PP was shown to sensitize both DU145 and PC3 cells to radiation-induced cell death determined in colony-formation assays. These data provide evidence that rLOX-PP has a nuclear mechanism of action in which it directly interacts with DNA repair proteins to sensitize prostate cancer cells to the effects of ionizing radiation.
Collapse
Affiliation(s)
- Manish V. Bais
- Boston University Henry M. Goldman School of Dental Medicine, Department of Periodontology and Oral Biology, Boston, MA 02118
| | - Gokhan Baris Ozdener
- Boston University Henry M. Goldman School of Dental Medicine, Department of Periodontology and Oral Biology, Boston, MA 02118
| | - Gail E. Sonenshein
- Tufts University School of Medicine, Department of Biochemistry, Boston, MA 02111
| | - Philip C. Trackman
- Boston University Henry M. Goldman School of Dental Medicine, Department of Periodontology and Oral Biology, Boston, MA 02118
| |
Collapse
|
25
|
Association of the G473A polymorphism and expression of lysyl oxidase with breast cancer risk and survival in European women: a hospital-based case-control study. PLoS One 2014; 9:e105579. [PMID: 25141126 PMCID: PMC4139364 DOI: 10.1371/journal.pone.0105579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/25/2014] [Indexed: 11/19/2022] Open
Abstract
Background Lysyl oxidase (LOX) is an extracellular enzyme essential for the covalent crosslinking of extracellular matrix proteins and may also have additional functions. LOX expression can be both up- and downregulated in cancer and is associated both with tumour suppression and metastasis progression. The G473A polymorphism (rs1800449) results in the Arg158Gln amino acid substitution in the LOX propeptide, compromises its tumour suppressive activity, and was associated with an increased breast cancer risk in a Chinese Han population. In the first hospital-based case-control study in European women, we aimed at investigating the association of LOX expression and the G473A polymorphism with breast cancer risk and survival in unselected and estrogen receptor (ER) negative patients. Methodology/Principal Findings The G473A polymorphism was genotyped in 386 breast cancer patients and 243 female controls. Moreover, LOX mRNA expression was quantified in the tumors of 105 patients by qRT-PCR. We found that the minor A-allele of this polymorphism is associated with a later age at breast cancer onset, a trend towards a decreased disease-free and metastasis-free survival, but not with an increased breast cancer risk. LOX mRNA expression was significantly elevated in tumours of patients older than 55 years, postmenopausal patients, estrogen receptor positive tumours, and p53 negative tumours, but was unaffected by G473A genotype in tumours and breast cancer cell lines. High LOX expression was associated with a poor disease-free and metastasis-free survival in ER negative but not ER positive patients. LOX expression was an independent prognostic parameter in multivariate analysis, whereas G473A genotype was not. A small, distinct subgroup of the ER negative patients was identified which exhibited a considerably elevated LOX expression and a very poor disease-free (p = 0.001) and metastasis-free survival (p = 0.0003). Conclusions/Significance This newly identified ER negative/LOX high subgroup may be a suitable collective for future individualized breast cancer diagnosis and therapy.
Collapse
|
26
|
Zheng Y, Wang X, Wang H, Yan W, Zhang Q, Chang X. Expression of the lysyl oxidase propeptide in hepatocellular carcinoma and its clinical relevance. Oncol Rep 2014; 31:1669-76. [PMID: 24573150 DOI: 10.3892/or.2014.3044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 01/20/2014] [Indexed: 11/05/2022] Open
Abstract
Lysyl oxidase is an important extracellular matrix remodeling enzyme and plays critical roles in tumor progression and development. Its tumor-suppressor activity has been shown to depend on the propeptide region. Previous studies have reported that the expression levels of lysyl oxidase propeptide (LOX-PP) are associated with cancer of the breast, pancreas, lung, prostate and gastrointestinal system. However, to date, the exact effects and molecular mechanisms of LOX-PP in hepatocellular carcinoma progression are still unclear. The present study aimed to investigate the expression and clinical significance of LOX-PP in human hepatocellular carcinoma. First, 42 cases of hepatocellular carcinoma and corresponding adjacent non-cancerous tissues (ANCTs) were collected, and the expression of LOX-PP in these samples was assessed by immunohistochemistry (IHC). The clinicopathological characteristics of all patients were recorded. Next, in in vitro studies, recombinant adenovirus LOX (ad-LOX-PP) was used to infect hepatocellular carcinoma cell lines to determine the function of LOX-PP. To determine whether ad-LOX-PP affects hepatocellular carcinoma cell survival, cell viability was examined by CCK-8 assay, and cell cycle progression was assessed by flow cytometry. We also investigated the effects of LOX-PP on the expression of cell cycle regulators (cyclin D1 and cyclin E) by western blot analysis. The migration and invasion capacities of hepatocellular carcinoma cells were observed by wound-healing and tranwell invasion assays. To further investigate how LOX-PP affects migration levels of matrix metallopeptidase (MMP)-2 and MMP-9 were assessed by western blot analysis. Additionally, markers of the PI3K and MAPK signaling pathway were detected to further confirm the mechanisms of LOX-PP. As a result, reduced expression of LOX-PP was found in hepatocellular carcinoma tissues, when compared with that in the ANCTs (15 vs. 83%, P<0.01), and its expression was associated with tumor stage and distant metastasis (each P<0.05). Proliferation in hepatocellular carcinoma cells was significantly decreased in the ad-LOX-PP group as indicated by CCK-8 assay. LOX-PP significantly reduced the expression of Ki-67, while prominent increases in the rate of apoptosis and cell cycle arrest were observed. Similarly, cell migration was significantly inhibited in the ad-LOX-PP group as evidenced by transwell invasion and wound-healing assays. The expression levels of MMP-2 and MMP-9 were attenuated in the ad-LOX-PP group, suggesting that LOX-PP inhibits hepatocellular carcinoma cell migration via down-regulation of MMPs expression. When LOX-PP expression was potentiated by an adenovirus containing LOX-PP, the expression of p-ERK was significantly downregulated, indicating that LOX-PP inhibits hepatocellular carcinoma cell proliferation and induces its apoptosis probably through downregulation of the MAPK/ERK pathway.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Anesthesia, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xuemei Wang
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - Haidong Wang
- Department of Obstetrics and Gynecology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Wei Yan
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Quan Zhang
- Department of Oncology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xin Chang
- Department of Imaging Medicine Center, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
27
|
Finney J, Moon HJ, Ronnebaum T, Lantz M, Mure M. Human copper-dependent amine oxidases. Arch Biochem Biophys 2014; 546:19-32. [PMID: 24407025 DOI: 10.1016/j.abb.2013.12.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 12/11/2022]
Abstract
Copper amine oxidases (CAOs) are a class of enzymes that contain Cu(2+) and a tyrosine-derived quinone cofactor, catalyze the conversion of a primary amine functional group to an aldehyde, and generate hydrogen peroxide and ammonia as byproducts. These enzymes can be classified into two non-homologous families: 2,4,5-trihydroxyphenylalanine quinone (TPQ)-dependent CAOs and the lysine tyrosylquinone (LTQ)-dependent lysyl oxidase (LOX) family of proteins. In this review, we will focus on recent developments in the field of research concerning human CAOs and the LOX family of proteins. The aberrant expression of these enzymes is linked to inflammation, fibrosis, tumor metastasis/invasion and other diseases. Consequently, there is a critical need to understand the functions of these proteins at the molecular level, so that strategies targeting these enzymes can be developed to combat human diseases.
Collapse
Affiliation(s)
- Joel Finney
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Hee-Jung Moon
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Trey Ronnebaum
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Mason Lantz
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Minae Mure
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
28
|
Abstract
Colorectal cancer is the third most prevalent form of cancer worldwide and fourth-leading cause of cancer-related mortality, leading to ~600,000 deaths annually, predominantly affecting the developed world. Lysyl oxidase is a secreted, extracellular matrix-modifying enzyme previously suggested to act as a tumor suppressor in colorectal cancer. However, emerging evidence has rapidly implicated lysyl oxidase in promoting metastasis of solid tumors and in particular colorectal cancer at multiple stages, affecting tumor cell proliferation, invasion, and angiogenesis. This emerging research has stimulated significant interest in lysyl oxidase as a strong candidate for developing and deploying inhibitors as functional efficacious cancer therapeutics. In this review, we discuss the rapidly expanding body of knowledge concerning lysyl oxidase in solid tumor progression, highlighting recent advancements in the field of colorectal cancer.
Collapse
Affiliation(s)
- Thomas R Cox
- Biotech Research and Innovation Centre (BRIC Univ. of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
29
|
Sato S, Zhao Y, Imai M, Simister PC, Feller SM, Trackman PC, Kirsch KH, Sonenshein GE. Inhibition of CIN85-mediated invasion by a novel SH3 domain binding motif in the lysyl oxidase propeptide. PLoS One 2013; 8:e77288. [PMID: 24167568 PMCID: PMC3805583 DOI: 10.1371/journal.pone.0077288] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/30/2013] [Indexed: 12/24/2022] Open
Abstract
The lysyl oxidase gene inhibits Ras signaling in transformed fibroblasts and breast cancer cells. Its activity was mapped to the 162 amino acid propeptide domain (LOX-PP) of the lysyl oxidase precursor protein. LOX-PP inhibited the Her-2/Ras signaling axis in breast cancer cells, and reduced the Her-2-driven breast tumor burden in a xenograft model. Since its mechanism of action is largely unknown, co-affinity-purification/mass spectrometry was performed and the “Cbl-interacting protein of 85-kDa” (CIN85) identified as an associating protein. CIN85 is an SH3-containing adapter protein that is overexpressed in invasive breast cancers. The CIN85 SH3 domains interact with c-Cbl, an E3 ubiquitin ligase, via an unconventional PxxxPR ligand sequence, with the highest affinity displayed by the SH3-B domain. Interaction with CIN85 recruits c-Cbl to the AMAP1 complex where its ubiquitination activity is necessary for cancer cells to develop an invasive phenotype and to degrade the matrix. Direct interaction of LOX-PP with CIN85 was confirmed using co-immunoprecipitation analysis of lysates from breast cancer cells and of purified expressed proteins. CIN85 interaction with c-Cbl was reduced by LOX-PP. Domain specific CIN85 regions and deletion mutants of LOX-PP were prepared and used to map the sites of interaction to the SH3-B domain of CIN85 and to an epitope encompassing amino acids 111 to 116 of LOX-PP. Specific LOX-PP point mutant proteins P111A and R116A failed to interact with CIN85 or to compete for CIN85 binding with c-Cbl. Structural modeling identified a new atypical PxpxxRh SH3-binding motif in this region of LOX-PP. The LOX-PP interaction with CIN85 was shown to reduce the invasive phenotype of breast cancer cells, including their ability to degrade the surrounding extracellular matrix and for Matrigel outgrowth. Thus, LOX-PP interacts with CIN85 via a novel SH3-binding motif and this association reduces CIN85-promoted invasion by breast cancer cells.
Collapse
Affiliation(s)
- Seiichi Sato
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Yingshe Zhao
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Misa Imai
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Philip C. Simister
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Stephan M. Feller
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Section Tumor Biology, Institute of Molecular Medicine, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Philip C. Trackman
- Division of Oral Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Kathrin H. Kirsch
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gail E. Sonenshein
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
30
|
XU XIN, WANG BIN, XU YANLING. Expression of lysyl oxidase in human osteosarcoma and its clinical significance: A tumor suppressive role of LOX in human osteosarcoma cells. Int J Oncol 2013; 43:1578-86. [DOI: 10.3892/ijo.2013.2067] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/18/2013] [Indexed: 11/06/2022] Open
|
31
|
Tadmor T, Bejar J, Attias D, Mischenko E, Sabo E, Neufeld G, Vadasz Z. The expression of lysyl-oxidase gene family members in myeloproliferative neoplasms. Am J Hematol 2013; 88:355-8. [PMID: 23494965 DOI: 10.1002/ajh.23409] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 01/28/2013] [Indexed: 12/16/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are malignant disorders originating from clonal expansion of a single neoplastic stem cell and characteristically show an increase in bone marrow reticulin fibers. Lysyl oxidases (LOXs) are copper-dependent amine oxidases that play a critical role in the biogenesis of connective tissue by crosslinking extracellular matrix proteins, collagen and elastin. Expression of LOX gene family members is increased in disorders associated with increased fibrosis. To evaluate involvement of LOX gene family in various MPNs. In-situ hybridization was used to detect Lysyl-Oxidase family members in bone marrow biopsies from patients with different MPNs. We compared normal bone marrows and those from patients with polycythemia vera, essential thrombocythemia, chronic myeloid leukemia, and primary myelofibrosis (PMF). Serum levels of lysyl-oxidase from patients with PMF and healthy controls were also examined. LOX gene family was not detected in normal bone marrows. All members of the LOX gene family were over expressed in PMF. In other MPNs a differential pattern of expression was observed. Differences in gene expression were statistically significant (P < 0.010). The medianserum LOX levels in normal controls was 28.4 ± 2.5 ng\ml and 44.6 ± 9.44 ng\ml in PMF (P = 0.02). The varying pattern of expression of LOX genes may reflect differences in the pathophysiology of bone marrow fibrosis in these MPNs. These observations could be used as the basis for future targeted therapy directed against bone marrow fibrosis.
Collapse
Affiliation(s)
| | - J. Bejar
- Department of Pathology; Bnai-Zion Medical Center; Haifa
| | - D. Attias
- The Hematology Unit; Bnai-Zion Medical Center; Haifa
| | - E. Mischenko
- The Hematology Unit; Bnai-Zion Medical Center; Haifa
| | | | - G. Neufeld
- The Bruce Rappaport Faculty of Medicine; Cancer and Vascular Biology Research Center; Rappaport Family Institute for Research in the Medical Sciences; Technion; Israel Institute of Haifa; Israel
| | | |
Collapse
|
32
|
Eliades A, Papadantonakis N, Matsuura S, Mi R, Bais MV, Trackman P, Ravid K. Megakaryocyte polyploidy is inhibited by lysyl oxidase propeptide. Cell Cycle 2013; 12:1242-50. [PMID: 23518500 DOI: 10.4161/cc.24312] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Megakaryocytes (MKs), the platelet precursors, undergo an endomitotic cell cycle that leads to polyploidy. Lysyl oxidase propeptide (LOX-PP) is generated from lysyl oxidase (LOX) pro-enzyme after proteolytical cleavage. We recently reported that LOX, a known matrix cross-linking enzyme, contributes to MK lineage expansion. In addition, LOX expression levels are ploidy-dependent, with polyploidy MKs having minimal levels. This led us to test the effects of LOX-PP on the number and ploidy of primary MKs. LOX-PP significantly decreases mouse bone marrow MK ploidy coupled with a reduction in MK size. MK number is unchanged upon LOX-PP treatment. Analysis of LOX-PP- or vehicle-treated MKs by western blotting revealed a reduction in ERK1/2 phosphorylation and in the levels of its downstream targets, cyclin D3 and cyclin E, which are known to play a central role in MK endomitosis. Pull-down assays and immunochemistry staining indicated that LOX-PP interacts with α-tubulin and the mictotubules, which can contribute to decreased MK ploidy. Thus, our findings defined a role for LOX-PP in reducing MK ploidy. This suggests that high-level expression of LOX in aberrantly proliferating MKs could play a part in inhibiting their polyploidization via LOX-PP.
Collapse
Affiliation(s)
- Alexia Eliades
- Department of Biochemistry, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Li L, Yang J, Wang WW, Yao YC, Fang SH, Dai ZY, Hong HH, Yang X, Shuai XT, Gao GQ. Pigment epithelium-derived factor gene loaded in cRGD–PEG–PEI suppresses colorectal cancer growth by targeting endothelial cells. Int J Pharm 2012; 438:1-10. [DOI: 10.1016/j.ijpharm.2012.08.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/30/2012] [Accepted: 08/21/2012] [Indexed: 01/19/2023]
|
34
|
Cano A, Santamaría PG, Moreno-Bueno G. LOXL2 in epithelial cell plasticity and tumor progression. Future Oncol 2012; 8:1095-108. [PMID: 23030485 DOI: 10.2217/fon.12.105] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Several members of the lysyl oxidase family have recently emerged as important regulators of tumor progression. Among them, LOXL2 has been shown to be involved in tumor progression and metastasis of several tumor types, including breast carcinomas. Secreted LOXL2 participates in the remodeling of the extracellular matrix of the tumor microenvironment, in a similar fashion to prototypical lysyl oxidase. In addition, new intracellular functions of LOXL2 have been described, such as its involvement in the regulation of the epithelial-to-mesenchymal transition, epithelial cell polarity and differentiation mediated by transcriptional repression mechanisms. Importantly, intracellular (perinuclear) expression of LOXL2 is associated with poor prognosis and distant metastasis of specific tumor types, such as larynx squamous cell carcinoma and basal breast carcinomas. These recent findings open new avenues for the therapeutic utility of LOXL2.
Collapse
Affiliation(s)
- Amparo Cano
- Departamento de Bioquímica, UAM, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, IdiPAZ, Arzobispo Morcillo, 2, 28029 Madrid, Spain.
| | | | | |
Collapse
|
35
|
Abstract
The therapeutic targeting of extracellular proteins is becoming hugely attractive in light of evidence implicating the tumour microenvironment as pivotal in all aspects of tumour initiation and progression. Members of the lysyl oxidase (LOX) family of proteins are secreted by tumours and are the subject of much effort to understand their roles in cancer. In this Review we discuss the roles of members of this family in the remodelling of the tumour microenvironment and their paradoxical roles in tumorigenesis and metastasis. We also discuss how targeting this family of proteins might lead to a new avenue of cancer therapeutics.
Collapse
Affiliation(s)
- Holly E Barker
- Hypoxia & Metastasis Team, The Institute of Cancer Research, London SW3 6JB, UK
| | | | | |
Collapse
|