1
|
Cetin M, Saatci O, Rezaeian AH, Rao CN, Beneker C, Sreenivas K, Taylor H, Pederson B, Chatzistamou I, Buckley B, Lessner S, Angel P, McInnes C, Sahin O. A highly potent bi-thiazole inhibitor of LOX rewires collagen architecture and enhances chemoresponse in triple-negative breast cancer. Cell Chem Biol 2024; 31:1926-1941.e11. [PMID: 39043186 PMCID: PMC11585458 DOI: 10.1016/j.chembiol.2024.06.012] [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: 09/14/2023] [Revised: 04/12/2024] [Accepted: 06/22/2024] [Indexed: 07/25/2024]
Abstract
Lysyl oxidase (LOX) is upregulated in highly stiff aggressive tumors, correlating with metastasis, resistance, and worse survival; however, there are currently no potent, safe, and orally bioavailable small molecule LOX inhibitors to treat these aggressive desmoplastic solid tumors in clinics. Here we discovered bi-thiazole derivatives as potent LOX inhibitors by robust screening of drug-like molecules combined with cell/recombinant protein-based assays. Structure-activity relationship analysis identified a potent lead compound (LXG6403) with ∼3.5-fold specificity for LOX compared to LOXL2 while not inhibiting LOXL1 with a competitive, time- and concentration-dependent irreversible mode of inhibition. LXG6403 shows favorable pharmacokinetic properties, globally changes ECM/collagen architecture, and reduces tumor stiffness. This leads to better drug penetration, inhibits FAK signaling, and induces ROS/DNA damage, G1 arrest, and apoptosis in chemoresistant triple-negative breast cancer (TNBC) cell lines, PDX organoids, and in vivo. Overall, our potent and tolerable bi-thiazole LOX inhibitor enhances chemoresponse in TNBC, the deadliest breast cancer subtype.
Collapse
Affiliation(s)
- Metin Cetin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Abdol-Hossein Rezaeian
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chintada Nageswara Rao
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chad Beneker
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kukkamudi Sreenivas
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Harrison Taylor
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Breanna Pederson
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology, University of South Carolina, Columbia, SC 29208, USA
| | - Brian Buckley
- Small Molecule Screening Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Susan Lessner
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Campbell McInnes
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
2
|
Steppan J, Wang H, Nandakumar K, Gadkari M, Poe A, Pak L, Brady T, Berkowitz DE, Shimoda LA, Santhanam L. LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L423-L438. [PMID: 39010824 PMCID: PMC11482525 DOI: 10.1152/ajplung.00327.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance have emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen cross-linking enzyme lysyl oxidase like 2 (LOXL2) in this study. Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from the pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe pulmonary hypertension (PH). Similarly, LOXL2 protein and mRNA levels were increased in the pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from the rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in vivo with PAT-1251. Importantly, PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Hypoxia-induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH and pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 could be a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH.NEW & NOTEWORTHY Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function, and PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.
Collapse
Affiliation(s)
- Jochen Steppan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Huilei Wang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Kavitha Nandakumar
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Mahin Gadkari
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore Maryland, United States
| | - Alan Poe
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lydia Pak
- Department of Molecular and Cellular Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Travis Brady
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Dan E Berkowitz
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore Maryland, United States
| |
Collapse
|
3
|
Jheng JR, Bai Y, Noda K, Huot JR, Cook T, Fisher A, Chen YY, Goncharov DA, Goncharova EA, Simon MA, Zhang Y, Forman DE, Rojas M, Machado RF, Auwerx J, Gladwin MT, Lai YC. Skeletal Muscle SIRT3 Deficiency Contributes to Pulmonary Vascular Remodeling in Pulmonary Hypertension Due to Heart Failure With Preserved Ejection Fraction. Circulation 2024; 150:867-883. [PMID: 38804138 PMCID: PMC11384544 DOI: 10.1161/circulationaha.124.068624] [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: 01/04/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a major complication linked to adverse outcomes in heart failure with preserved ejection fraction (HFpEF), yet no specific therapies exist for PH associated with HFpEF (PH-HFpEF). We have recently reported on the role of skeletal muscle SIRT3 (sirtuin-3) in modulation of PH-HFpEF, suggesting a novel endocrine signaling pathway for skeletal muscle modulation of pulmonary vascular remodeling. METHODS Using skeletal muscle-specific Sirt3 knockout mice (Sirt3skm-/-) and mass spectrometry-based comparative secretome analysis, we attempted to define the processes by which skeletal muscle SIRT3 defects affect pulmonary vascular health in PH-HFpEF. RESULTS Sirt3skm-/- mice exhibited reduced pulmonary vascular density accompanied by pulmonary vascular proliferative remodeling and elevated pulmonary pressures. Comparative analysis of secretome by mass spectrometry revealed elevated secretion levels of LOXL2 (lysyl oxidase homolog 2) in SIRT3-deficient skeletal muscle cells. Elevated circulation and protein expression levels of LOXL2 were also observed in plasma and skeletal muscle of Sirt3skm-/- mice, a rat model of PH-HFpEF, and humans with PH-HFpEF. In addition, expression levels of CNPY2 (canopy fibroblast growth factor signaling regulator 2), a known proliferative and angiogenic factor, were increased in pulmonary artery endothelial cells and pulmonary artery smooth muscle cells of Sirt3skm-/- mice and animal models of PH-HFpEF. CNPY2 levels were also higher in pulmonary artery smooth muscle cells of subjects with obesity compared with nonobese subjects. Moreover, treatment with recombinant LOXL2 protein promoted pulmonary artery endothelial cell migration/proliferation and pulmonary artery smooth muscle cell proliferation through regulation of CNPY2-p53 signaling. Last, skeletal muscle-specific Loxl2 deletion decreased pulmonary artery endothelial cell and pulmonary artery smooth muscle cell expression of CNPY2 and improved pulmonary pressures in mice with high-fat diet-induced PH-HFpEF. CONCLUSIONS This study demonstrates a systemic pathogenic impact of skeletal muscle SIRT3 deficiency in remote pulmonary vascular remodeling and PH-HFpEF. This study suggests a new endocrine signaling axis that links skeletal muscle health and SIRT3 deficiency to remote CNPY2 regulation in the pulmonary vasculature through myokine LOXL2. Our data also identify skeletal muscle SIRT3, myokine LOXL2, and CNPY2 as potential targets for the treatment of PH-HFpEF.
Collapse
MESH Headings
- Animals
- Sirtuin 3/metabolism
- Sirtuin 3/deficiency
- Sirtuin 3/genetics
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/etiology
- Vascular Remodeling
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Mice, Knockout
- Mice
- Humans
- Stroke Volume
- Male
- Rats
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Disease Models, Animal
- Female
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Yang Bai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang (Y.B.)
| | - Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, PA (K.N.)
| | - Joshua R Huot
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Todd Cook
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Amanda Fisher
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Yi-Yun Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan (Y.-Y.C.)
| | - Dmitry A Goncharov
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis (D.A.G., E.A.G.)
| | - Elena A Goncharova
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis (D.A.G., E.A.G.)
| | - Marc A Simon
- Division of Cardiology, University of California, San Francisco (M.A.S.)
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine (Y.Z.), University of Pittsburgh, PA
| | - Daniel E Forman
- Department of Medicine, Divisions of Geriatrics and Cardiology (D.E.F.), University of Pittsburgh, PA
- Geriatric Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, PA (D.E.F.)
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus (M.R.)
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Switzerland (J.A.)
| | - Mark T Gladwin
- Department of Medicine, University of Maryland, Baltimore (M.T.G.)
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| |
Collapse
|
4
|
He A, He L, Chen T, Li X, Cao C. Biomechanical Properties and Cellular Responses in Pulmonary Fibrosis. Bioengineering (Basel) 2024; 11:747. [PMID: 39199705 PMCID: PMC11351367 DOI: 10.3390/bioengineering11080747] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Pulmonary fibrosis is a fatal lung disease affecting approximately 5 million people worldwide, with a 5-year survival rate of less than 50%. Currently, the only available treatments are palliative care and lung transplantation, as there is no curative drug for this condition. The disease involves the excessive synthesis of the extracellular matrix (ECM) due to alveolar epithelial cell damage, leading to scarring and stiffening of the lung tissue and ultimately causing respiratory failure. Although multiple factors contribute to the disease, the exact causes remain unclear. The mechanical properties of lung tissue, including elasticity, viscoelasticity, and surface tension, are not only affected by fibrosis but also contribute to its progression. This paper reviews the alteration in these mechanical properties as pulmonary fibrosis progresses and how cells in the lung, including alveolar epithelial cells, fibroblasts, and macrophages, respond to these changes, contributing to disease exacerbation. Furthermore, it highlights the importance of developing advanced in vitro models, based on hydrogels and 3D bioprinting, which can accurately replicate the mechanical and structural properties of fibrotic lungs and are conducive to studying the effects of mechanical stimuli on cellular responses. This review aims to summarize the current understanding of the interaction between the progression of pulmonary fibrosis and the alterations in mechanical properties, which could aid in the development of novel therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Andong He
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Lizhe He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | - Tianwei Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
| |
Collapse
|
5
|
Deshpande H. Levoleucovorin inhibits LOXL2 (lysyl oxidase like-2) to control breast cancer proliferation: a repurposing approach. J Biomol Struct Dyn 2024; 42:5104-5113. [PMID: 37340696 DOI: 10.1080/07391102.2023.2224894] [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: 03/22/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Lysyl oxidase like-2 (LOXL2) belongs to copper dependent amine oxidase from the lysyl oxidase family and is associated with breast cancer metastasis This study used multi-stage computational screening and in vitro validations to repurpose FDA approved drugs targeting LOXL2 to control breast cancer progression.Molecular modeling techniques and high-throughput virtual-screening technique was employed to screen FDA-approved drug library for its avid binding to LOXL2.hLOXL2, MDA-MB231 and MCF 7 cells were used for in vitro.Collectively, this repurposing study identified levoleucovorin to bind the active site of LOXL2 protein to inhibit its activity. Further validation of levoleucovorin against LOXL2 activity is warranted toward repurposing levoleucovorin as a therapeutic agent for treating breast cancer patients. validations.Computational modeling of LOXL2 identified putative druggable region at the active site of LOXL2 protein. High-throughput virtual screening predicted levoleucovorin as a best lead drug candidate to have a favorable binding affinity for LOXL2 at its active site. Molecular dynamic simulation predicts levoleucovorin to bind stably and avidly to LOXL2 with favorable interactions. In vitro validations show levoleucovorin significantly inhibited hLOXL2 with and IC50 value of 68.81 μM. Levoleucovorin controlled cell proliferations in MDM-MB 231 and MCF-7 cells with GI50 values of 55.91 μM and 79.20 μM respectively. Further, a dose dependent inhibition of cancer cell migration was noted along with apoptosis induction in these cells with levoleucovorin treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hemali Deshpande
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Asir, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Ye C, Jiang S, Zeng T, He S, Cao J, Xiao J. The role of LOXL2 in tumor progression, immune response and cellular senescence: a comprehensive analysis. Discov Oncol 2024; 15:245. [PMID: 38922489 PMCID: PMC11208360 DOI: 10.1007/s12672-024-01107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
LOXL2, an enzyme belonging to the LOX family, facilitates the cross-linking of extracellular matrix (ECM) elements. However, the roles of the LOXL2 gene in mechanisms of oncogenesis and tumor development have not been clearly defined. In this pan-cancer study, we examined the notable disparity in LOXL2 expression at the mRNA and protein levels among various cancer types and elucidated its interconnected roles in tumor progression, mutational profile, immune response, and cellular senescence. Apart from investigating the hyperexpression of LOXL2 being related to poorer prognosis in different types of tumors, this study also unveiled noteworthy connections between LOXL2 and genetic mutations, infiltration of tumor immune cells, and genes in immune checkpoint pathways. Further analysis revealed the participation of LOXL2 in multiple pathways related to cancer extracellular matrix remodeling and cellular senescence. Moreover, our investigation uncovered that the knockdown and inhibition of LOXL2 significantly attenuated the proliferation and migration of PC-9 and HCC-LM3 cells. The knock-down and inhibition of LOXL2 enhanced cellular senescence in lung and liver cancer cells, as confirmed by SA-β-Gal staining and quantitative RT-PCR analyses. This comprehensive analysis offers valuable insights on the functions of LOXL2 in different types of cancer and its role in regulating the senescence of cancer cells.
Collapse
Affiliation(s)
- Chen Ye
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Sihan Jiang
- Graduate School, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Tanlun Zeng
- Graduate School, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Shaohui He
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jinjin Cao
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China.
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| | - Jianru Xiao
- School of Health Science and Technology, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200093, China.
- Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
7
|
Jalil SMA, Henry JC, Cameron AJM. Targets in the Tumour Matrisome to Promote Cancer Therapy Response. Cancers (Basel) 2024; 16:1847. [PMID: 38791926 PMCID: PMC11119821 DOI: 10.3390/cancers16101847] [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: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is composed of complex fibrillar proteins, proteoglycans, and macromolecules, generated by stromal, immune, and cancer cells. The components and organisation of the matrix evolves as tumours progress to invasive disease and metastasis. In many solid tumours, dense fibrotic ECM has been hypothesised to impede therapy response by limiting drug and immune cell access. Interventions to target individual components of the ECM, collectively termed the matrisome, have, however, revealed complex tumour-suppressor, tumour-promoter, and immune-modulatory functions, which have complicated clinical translation. The degree to which distinct components of the matrisome can dictate tumour phenotypes and response to therapy is the subject of intense study. A primary aim is to identify therapeutic opportunities within the matrisome, which might support a better response to existing therapies. Many matrix signatures have been developed which can predict prognosis, immune cell content, and immunotherapy responses. In this review, we will examine key components of the matrisome which have been associated with advanced tumours and therapy resistance. We have primarily focussed here on targeting matrisome components, rather than specific cell types, although several examples are described where cells of origin can dramatically affect tumour roles for matrix components. As we unravel the complex biochemical, biophysical, and intracellular transduction mechanisms associated with the ECM, numerous therapeutic opportunities will be identified to modify tumour progression and therapy response.
Collapse
Affiliation(s)
| | | | - Angus J. M. Cameron
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; (S.M.A.J.); (J.C.H.)
| |
Collapse
|
8
|
Pal S, Openy J, Krzyzanowski A, Noisier A, ‘t Hart P. On-Resin Photochemical Decarboxylative Arylation of Peptides. Org Lett 2024; 26:2795-2799. [PMID: 37819674 PMCID: PMC11019635 DOI: 10.1021/acs.orglett.3c03070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Indexed: 10/13/2023]
Abstract
Here we describe the application of photochemical decarboxylative arylation as a late-stage functionalization reaction for peptides. The reaction uses redox-active esters of aspartic acid and glutamic acid on the solid phase to provide analogues of aromatic amino acids. By using aryl bromides as arylation reagents, a wide variety of amino acids can be accessed without having to synthesize them individually in solution. The reaction is compatible with proteinogenic amino acids and was used to perform a structure-activity relationship study of a PRMT5 binding peptide.
Collapse
Affiliation(s)
- Sunit Pal
- Chemical
Genomics Centre, Max Planck Institute of
Molecular Physiology, 44227 Dortmund, Germany
| | - Joseph Openy
- Chemical
Genomics Centre, Max Planck Institute of
Molecular Physiology, 44227 Dortmund, Germany
| | - Adrian Krzyzanowski
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, 44227 Dortmund, Germany
| | - Anaïs Noisier
- Medicinal
Chemistry, Research and Early Development Cardiovascular, Renal and
Metabolism BioPharmaceutical R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Peter ‘t Hart
- Chemical
Genomics Centre, Max Planck Institute of
Molecular Physiology, 44227 Dortmund, Germany
| |
Collapse
|
9
|
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
|
10
|
Steppan J, Wang H, Nandakumar K, Poe A, Pak L, Brady T, Gadkari M, Berkowitz DE, Shimoda LA, Santhanam L. LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563874. [PMID: 37961202 PMCID: PMC10634806 DOI: 10.1101/2023.10.24.563874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance has emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen crosslinking enzyme lysyl oxidase like 2 (LOXL2) in this study. Methods and Results Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe PH. Similarly, LOXL2 protein and mRNA levels were increased in pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in-vivo with PAT-1251. Importantly PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Conclusion Hypoxia induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH, as well as pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 is a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH. New & Noteworthy Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function and improves PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.
Collapse
|
11
|
Lu Y, Deng Y, Ko H, Peng H, Lee H, Kuo MY, Cheng S. Lysyl oxidase-like 2 promotes stemness and enhances antitumor effects of gefitinib in head and neck cancer via IFIT1 and IFIT3. Cancer Sci 2023; 114:3957-3971. [PMID: 37496288 PMCID: PMC10551584 DOI: 10.1111/cas.15912] [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: 11/11/2022] [Revised: 06/22/2023] [Accepted: 07/09/2023] [Indexed: 07/28/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) is a matrix-remodeling enzyme that has recently been identified as an important regulator of tumor progression and metastasis. This study discovered that LOXL2 expression in oral squamous cell carcinoma (OSCC) tissues was significantly associated with tumor clinical stage, lymph node metastasis and patients' overall survival time. LOXL2-overexpressing human buccal SCC TW2.6 (TW2.6/LOXL2) and hypopharyngeal SCC FaDu (FaDu/LOXL2) cells exhibited enhanced migration, invasion, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) phenotypes, independently of its enzymatic activity. Moreover, TW2.6/LOXL2 significantly increased tumor-initiating frequency in SCID mice. We further demonstrated that LOXL2 increased the levels of interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) and IFIT3 in TW2.6/LOXL2 and FaDu/LOXL2 cells. We also identified IFIT1 and IFIT3 as key downstream components of LOXL2 action in migration, invasion, EMT, and CSC phenotypes in TW2.6 and FaDu cells. Furthermore, a significant positive correlation between LOXL2 expression and IFIT1 and IFIT3 overexpression in human OSCC tissues was observed. In addition, TW2.6/LOXL2 and FaDu/LOXL2 cells were 3.3- to 3.6-fold more susceptible to the epidermal growth factor receptor (EGFR) inhibitor gefitinib than were their respective control cells. The antitumor effect of gefitinib on orthotopic TW2.6/LOXL2 xenograft tumor was fourfold higher than that on controls. Our results indicate that LOXL2 expression is a strong prognostic factor for OSCC and may be used as a marker to identify patients most likely to respond to EGFR-targeted therapy.
Collapse
Affiliation(s)
- Yi‐Jie Lu
- Graduate Institute of Oral Biology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Yi‐Ting Deng
- Department of DentistryNational Taiwan University Hospital Hsin‐Chu BranchHsin‐ChuTaiwan
| | - Hui‐Hsin Ko
- Department of DentistryNational Taiwan University Hospital Hsin‐Chu BranchHsin‐ChuTaiwan
| | - Hsin‐Hui Peng
- Department of DentistryNational Taiwan University Hospital Hsin‐Chu BranchHsin‐ChuTaiwan
| | - Hsiang‐Chieh Lee
- Graduate Institute of Photonics and OptoelectronicsNational Taiwan UniversityTaipeiTaiwan
| | - Mark Yen‐Ping Kuo
- Department of Dentistry, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of DentistryNational Taiwan University HospitalTaipeiTaiwan
| | - Shih‐Jung Cheng
- Graduate Institute of Oral Biology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of Dentistry, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of DentistryNational Taiwan University HospitalTaipeiTaiwan
| |
Collapse
|
12
|
Cano A, Eraso P, Mazón MJ, Portillo F. LOXL2 in Cancer: A Two-Decade Perspective. Int J Mol Sci 2023; 24:14405. [PMID: 37762708 PMCID: PMC10532419 DOI: 10.3390/ijms241814405] [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: 07/28/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Lysyl Oxidase Like 2 (LOXL2) belongs to the lysyl oxidase (LOX) family, which comprises five lysine tyrosylquinone (LTQ)-dependent copper amine oxidases in humans. In 2003, LOXL2 was first identified as a promoter of tumour progression and, over the course of two decades, numerous studies have firmly established its involvement in multiple cancers. Extensive research with large cohorts of human tumour samples has demonstrated that dysregulated LOXL2 expression is strongly associated with poor prognosis in patients. Moreover, investigations have revealed the association of LOXL2 with various targets affecting diverse aspects of tumour progression. Additionally, the discovery of a complex network of signalling factors acting at the transcriptional, post-transcriptional, and post-translational levels has provided insights into the mechanisms underlying the aberrant expression of LOXL2 in tumours. Furthermore, the development of genetically modified mouse models with silenced or overexpressed LOXL2 has enabled in-depth exploration of its in vivo role in various cancer models. Given the significant role of LOXL2 in numerous cancers, extensive efforts are underway to identify specific inhibitors that could potentially improve patient prognosis. In this review, we aim to provide a comprehensive overview of two decades of research on the role of LOXL2 in cancer.
Collapse
Affiliation(s)
- Amparo Cano
- Departamento de Bioquímica UAM, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.C.); (P.E.); (M.J.M.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz—IdiPAZ, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red, Área de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pilar Eraso
- Departamento de Bioquímica UAM, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.C.); (P.E.); (M.J.M.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz—IdiPAZ, 28029 Madrid, Spain
| | - María J. Mazón
- Departamento de Bioquímica UAM, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.C.); (P.E.); (M.J.M.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz—IdiPAZ, 28029 Madrid, Spain
| | - Francisco Portillo
- Departamento de Bioquímica UAM, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (A.C.); (P.E.); (M.J.M.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz—IdiPAZ, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red, Área de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
13
|
Löser R, Kuchar M, Wodtke R, Neuber C, Belter B, Kopka K, Santhanam L, Pietzsch J. Lysyl Oxidases as Targets for Cancer Therapy and Diagnostic Imaging. ChemMedChem 2023; 18:e202300331. [PMID: 37565736 DOI: 10.1002/cmdc.202300331] [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: 06/28/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/12/2023]
Abstract
The understanding of the contribution of the tumour microenvironment to cancer progression and metastasis, in particular the interplay between tumour cells, fibroblasts and the extracellular matrix has grown tremendously over the last years. Lysyl oxidases are increasingly recognised as key players in this context, in addition to their function as drivers of fibrotic diseases. These insights have considerably stimulated drug discovery efforts towards lysyl oxidases as targets over the last decade. This review article summarises the biochemical and structural properties of theses enzymes. Their involvement in tumour progression and metastasis is highlighted from a biochemical point of view, taking into consideration both the extracellular and intracellular action of lysyl oxidases. More recently reported inhibitor compounds are discussed with an emphasis on their discovery, structure-activity relationships and the results of their biological characterisation. Molecular probes developed for imaging of lysyl oxidase activity are reviewed from the perspective of their detection principles, performance and biomedical applications.
Collapse
Affiliation(s)
- Reik Löser
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Manuela Kuchar
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Christin Neuber
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Birgit Belter
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Lakshmi Santhanam
- Departments of Anesthesiology and Critical Care Medicine and Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| |
Collapse
|
14
|
Radić J, Kožik B, Nikolić I, Kolarov-Bjelobrk I, Vasiljević T, Vranjković B, Despotović S. Multiple Roles of LOXL2 in the Progression of Hepatocellular Carcinoma and Its Potential for Therapeutic Targeting. Int J Mol Sci 2023; 24:11745. [PMID: 37511503 PMCID: PMC10380739 DOI: 10.3390/ijms241411745] [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: 06/10/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
LOXL2, a copper-dependent amine oxidase, has emerged as a promising therapeutic target in hepatocellular carcinoma (HCC). Increased LOXL2 expression in HCC has been linked with an aggressive phenotype and represents a poor prognostic factor. Here, we focus on the mechanisms through which LOXL2 orchestrates multiple oncogenic functions in HCC development. We performed a review of the current knowledge on the roles LOXL2 performs in the modulation of the HCC tumor microenvironment, formation of premetastatic niches, and epithelial-mesenchymal transition. We also highlighted the complex interplay between LOXL2 and hypoxia, angiogenesis, and vasculogenic mimicry in HCC. At the end of the review, we summarize the current LOXL2 inhibitors and discuss their potential in HCC precision treatment.
Collapse
Affiliation(s)
- Jelena Radić
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Bojana Kožik
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11100 Belgrade, Serbia
| | - Ivan Nikolić
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Ivana Kolarov-Bjelobrk
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Tijana Vasiljević
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Pathology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Bojana Vranjković
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Sanja Despotović
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
15
|
Caligiuri A, Parola M, Marra F, Cannito S, Gentilini A. Cholangiocarcinoma tumor microenvironment highlighting fibrosis and matrix components. HEPATOMA RESEARCH 2023. [DOI: 10.20517/2394-5079.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Cholangiocarcinoma (CCA) is an extremely aggressive malignancy characterized by a very limited prognosis and scarce treatment options. The majority of patients are diagnosed at an advanced stage and do not qualify for potentially curative surgical treatments, making CCA an increasingly prevalent global challenge. CCA is characterized by a highly reactive desmoplastic stroma, with complex mechanisms underlying the mutual interactions between tumor cells and stromal compartment. This review focuses on the recent studies examining CCA’s biological features, with particular reference to the tumor reactive stroma (TRS) and its role in CCA progression, including matrix remodeling, angiogenesis and lymphangiogenesis, metastasis, and immune evasion. After giving a panoramic view of the relationship between the tumoral and stromal compartment (cancer-associated fibroblast, CAFs and tumor-associated macrophages, TAMs), this review also discusses the current therapeutic approaches to counteract CAFs and TAMs effects on CCA progression.
Collapse
|
16
|
Cheng F, Yang F, Wang Y, Zhou J, Qian H, Yan Y. Mesenchymal stem cell-derived exosomal miR-27b-3p alleviates liver fibrosis via downregulating YAP/LOXL2 pathway. J Nanobiotechnology 2023; 21:195. [PMID: 37328872 DOI: 10.1186/s12951-023-01942-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/29/2023] [Indexed: 06/18/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) is an extracellular copper-dependent enzyme that plays a central role in fibrosis by catalyzing the crosslinking and deposition of collagen. Therapeutic LOXL2 inhibition has been shown to suppress liver fibrosis progression and promote its reversal. This study investigates the efficacy and underlying mechanisms of human umbilical cord-derived exosomes (MSC-ex) in LOXL2 inhibition of liver fibrosis. MSC-ex, nonselective LOX inhibitor β-aminopropionitrile (BAPN), or PBS were administered into carbon tetrachloride (CCl4)-induced fibrotic livers. Serum LOXL2 and collagen crosslinking were assessed histologically and biochemically. MSC-ex's mechanisms on LOXL2 regulation were investigated in human hepatic stellate cell line LX-2. We found that systemic administration of MSC-ex significantly reduced LOXL2 expression and collagen crosslinking, delaying the progression of CCl4-induced liver fibrosis. Mechanically, RNA-sequencing and fluorescence in situ hybridization (FISH) indicated that miR-27b-3p was enriched in MSC-ex and exosomal miR-27b-3p repressed Yes-associated protein (YAP) expression by targeting its 3' untranslated region in LX-2. LOXL2 was identified as a novel downstream target gene of YAP, and YAP bound to the LOXL2 promoter to positively regulate transcription. Additionally, the miR-27b-3p inhibitor abrogated the anti-LOXL2 abilities of MSC-ex and diminished the antifibrotic efficacy. miR-27b-3p overexpression promoted MSC-ex mediated YAP/LOXL2 inhibition. Thus, MSC-ex may suppress LOXL2 expression through exosomal miR-27b-3p mediated YAP down-regulation. The findings here may improve our understanding of MSC-ex in liver fibrosis alleviation and provide new opportunities for clinical treatment.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, 213017, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, 212001, China
| | - Fuji Yang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, 213017, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, 212001, China
| | - Yanjin Wang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, 213017, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, 212001, China
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, 212001, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, 213017, China.
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China.
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, China.
| |
Collapse
|
17
|
Vizier R, Garnier AR, Dias A, Moreau M, Claron M, Collin B, Denat F, Bellaye PS, Goncalves V. SPECT Imaging of Lysyl Oxidase-like 2 in a Model of Idiopathic Pulmonary Fibrosis. Mol Pharm 2023. [PMID: 37307296 DOI: 10.1021/acs.molpharmaceut.3c00232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Noninvasive imaging of idiopathic pulmonary fibrosis (IPF) remains a challenge. The aim of this study was to develop an antibody-based radiotracer targeting Lysyl Oxidase-like 2 (LOXL2), an enzyme involved in the fibrogenesis process, for SPECT/CT imaging of pulmonary fibrosis. The bifunctional chelator DOTAGA-PEG4-NH2 was chemoenzymatically conjugated to the murine antibody AB0023 using microbial transglutaminase, resulting in a degree of labeling (number of chelators per antibody) of 2.3. Biolayer interferometry confirmed that the binding affinity of DOTAGA-AB0023 to LOXL2 was preserved with a dissociation constant of 2.45 ± 0.04 nM. DOTAGA-AB0023 was then labeled with 111In and in vivo experiments were carried out in a mice model of progressive pulmonary fibrosis induced by intratracheal administration of bleomycin. [111In]In-DOTAGA-AB0023 was injected in three groups of mice (control, fibrotic, and treated with nintedanib). SPECT/CT images were recorded over 4 days p.i. and an ex vivo biodistribution study was performed by gamma counting. A significant accumulation of the tracer in the lungs of the fibrotic mice was observed at D18 post-bleomycin. Interestingly, the tracer uptake was found selectively upregulated in fibrotic lesions observed on CT scans. Images of mice that received the antifibrotic drug nintedanib from D8 up to D18 showed a decrease in [111In]In-DOTAGA-AB0023 lung uptake associated with a decrease in pulmonary fibrosis measured by CT scan. In conclusion, we report the first radioimmunotracer targeting the protein LOXL2 for nuclear imaging of IPF. The tracer showed promising results in a preclinical model of bleomycin-induced pulmonary fibrosis, with high lung uptake in fibrotic areas, and accounted for the antifibrotic activity of nintedanib.
Collapse
Affiliation(s)
- Romane Vizier
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
| | - Anaïs-Rachel Garnier
- Centre Georges François Leclerc, Service de Médecine Nucléaire, Plateforme d'Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon Cedex, France
| | - Alexandre Dias
- Centre Georges François Leclerc, Service de Médecine Nucléaire, Plateforme d'Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon Cedex, France
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
| | - Michael Claron
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
| | - Bertrand Collin
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
- Centre Georges François Leclerc, Service de Médecine Nucléaire, Plateforme d'Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon Cedex, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
| | - Pierre-Simon Bellaye
- Centre Georges François Leclerc, Service de Médecine Nucléaire, Plateforme d'Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon Cedex, France
| | - Victor Goncalves
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, 9 Avenue Alain Savary, 21078 Dijon Cedex, France
| |
Collapse
|
18
|
Meier AA, Kuczera K, Mure M. A 3D-Predicted Structure of the Amine Oxidase Domain of Lysyl Oxidase-Like 2. Int J Mol Sci 2022; 23:13385. [PMID: 36362176 PMCID: PMC9659206 DOI: 10.3390/ijms232113385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 07/30/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) has been recognized as an attractive drug target for anti-fibrotic and anti-tumor therapies. However, the structure-based drug design of LOXL2 has been very challenging due to the lack of structural information of the catalytically-competent LOXL2. In this study; we generated a 3D-predicted structure of the C-terminal amine oxidase domain of LOXL2 containing the lysine tyrosylquinone (LTQ) cofactor from the 2.4Å crystal structure of the Zn2+-bound precursor (lacking LTQ; PDB:5ZE3); this was achieved by molecular modeling and molecular dynamics simulation based on our solution studies of a mature LOXL2 that is inhibited by 2-hydrazinopyridine. The overall structures of the 3D-modeled mature LOXL2 and the Zn2+-bound precursor are very similar (RMSD = 1.070Å), and disulfide bonds are conserved. The major difference of the mature and the precursor LOXL2 is the secondary structure of the pentapeptide (His652-Lys653-Ala654-Ser655-Phe656) containing Lys653 (the precursor residue of the LTQ cofactor). We anticipate that this peptide is flexible in solution to accommodate the conformation that enables the LTQ cofactor formation as opposed to the β-sheet observed in 5ZE3. We discuss the active site environment surrounding LTQ and Cu2+ of the 3D-predicted structure.
Collapse
Affiliation(s)
- Alex A. Meier
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Krzysztof Kuczera
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Minae Mure
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
19
|
Gong L, Zhang Y, Yang Y, Yan Q, Ren J, Luo J, Tiu YC, Fang X, Liu B, Lam RHW, Lam K, Lee AW, Guan X. Inhibition of lysyl oxidase-like 2 overcomes adhesion-dependent drug resistance in the collagen-enriched liver cancer microenvironment. Hepatol Commun 2022; 6:3194-3211. [PMID: 35894804 PMCID: PMC9592791 DOI: 10.1002/hep4.1966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/13/2022] [Accepted: 02/27/2022] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) is considered to be one of the vital mediators of tumor progression. Extracellular matrix (ECM), infiltrating immune cells, and stromal cells collectively constitute the complex ecosystem with varied biochemical and biophysical properties. The development of liver cancer is strongly tied with fibrosis and cirrhosis that alters the microenvironmental landscape, especially ECM composition. Enhanced deposition and cross-linking of type I collagen are frequently detected in patients with liver cancer and have been shown to facilitate tumor growth and metastasis by epithelial-to-mesenchymal transition. However, information on the effect of collagen enrichment on drug resistance is lacking. Thus, the present study has comprehensively illustrated phenotypical and mechanistic changes in an in vitro mimicry of collagen-enriched TME and revealed that collagen enrichment could induce 5-fluorouracil (5FU) and sorafenib resistance in liver cancer cells through hypoxia-induced up-regulation of lysyl oxidase-like 2 (LOXL2). LOXL2, an enzyme that facilitates collagen cross-linking, enhances cell adhesion-mediated drug resistance by activating the integrin alpha 5 (ITGA5)/focal adhesion kinase (FAK)/phosphoinositide 3-kinase (PI3K)/rho-associated kinase 1 (ROCK1) signaling axis. Conclusion: We demonstrated that inhibition of LOXL2 in a collagen-enriched microenvironment synergistically promotes the efficacy of sorafenib and 5FU through deterioration of focal adhesion signaling. These findings have clinical implications for developing LOXL2-targeted strategies in patients with chemoresistant liver cancer and especially for those patients with advanced fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Lanqi Gong
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongChina
| | - Yu Zhang
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongChina
- Department of Pediatric OncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in Southern ChinaSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yuma Yang
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
| | - Qian Yan
- Department of Colorectal SurgeryGuangdong Institute Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Jifeng Ren
- Department of Biomedical EngineeringCity University of Hong KongHong KongChina
- School of Biomedical EngineeringCapital Medical UniversityBeijingChina
| | - Jie Luo
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
| | - Yuen Chak Tiu
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
| | - Xiaona Fang
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongChina
| | - Beilei Liu
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongChina
| | - Raymond Hiu Wai Lam
- Department of Biomedical EngineeringCity University of Hong KongHong KongChina
| | - Ka‐On Lam
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
| | - Anne Wing‐Mui Lee
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhouChina
| | - Xin‐Yuan Guan
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- Department of Clinical OncologyLi Ka Shing Faculty of MedicineHong KongChina
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongChina
- State Key Laboratory of Oncology in Southern ChinaSun Yat‐sen University Cancer CenterGuangzhouChina
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhouChina
| |
Collapse
|
20
|
Liburkin-Dan T, Nir-Zvi I, Razon H, Kessler O, Neufeld G. Knock-Out of the Five Lysyl-Oxidase Family Genes Enables Identification of Lysyl-Oxidase Pro-Enzyme Regulated Genes. Int J Mol Sci 2022; 23:ijms231911322. [PMID: 36232621 PMCID: PMC9570307 DOI: 10.3390/ijms231911322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 12/02/2022] Open
Abstract
The five lysyl-oxidase genes share similar enzymatic activities and contribute to tumor progression. We have knocked out the five lysyl-oxidase genes in MDA-MB-231 breast cancer cells using CRISPR/Cas9 in order to identify genes that are regulated by LOX but not by other lysyl-oxidases and in order to study such genes in more mechanistic detail in the future. Re-expression of the full-length cDNA encoding LOX identified four genes whose expression was downregulated in the knock-out cells and rescued following LOX re-expression but not re-expression of other lysyl-oxidases. These were the AGR2, STOX2, DNAJB11 and DNAJC3 genes. AGR2 and STOX2 were previously identified as promoters of tumor progression. In addition, we identified several genes that were not downregulated in the knock-out cells but were strongly upregulated following LOX or LOXL3 re-expression. Some of these, such as the DERL3 gene, also promote tumor progression. There was very little proteolytic processing of the re-expressed LOX pro-enzyme in the MDA-MB-231 cells, while in the HEK293 cells, the LOX pro-enzyme was efficiently cleaved. We introduced point mutations into the known BMP-1 and ADAMTS2/14 cleavage sites of LOX. The BMP-1 mutant was secreted but not cleaved, while the LOX double mutant dmutLOX was not cleaved or secreted. However, even in the presence of the irreversible LOX inhibitor β-aminoproprionitrile (BAPN), these point-mutated LOX variants induced the expression of these genes, suggesting that the LOX pro-enzyme has hitherto unrecognized biological functions.
Collapse
|
21
|
Meier AA, Go EP, Moon HJ, Desaire H, Mure M. Mass Spectrometry-Based Disulfide Mapping of Lysyl Oxidase-like 2. Int J Mol Sci 2022; 23:5879. [PMID: 35682561 PMCID: PMC9180022 DOI: 10.3390/ijms23115879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 01/09/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) catalyzes the oxidative deamination of peptidyl lysines and hydroxylysines to promote extracellular matrix remodeling. Aberrant activity of LOXL2 has been associated with organ fibrosis and tumor metastasis. The lysine tyrosylquinone (LTQ) cofactor is derived from Lys653 and Tyr689 in the amine oxidase domain via post-translational modification. Based on the similarity in hydrodynamic radius and radius of gyration, we recently proposed that the overall structures of the mature LOXL2 (containing LTQ) and the precursor LOXL2 (no LTQ) are very similar. In this study, we conducted a mass spectrometry-based disulfide mapping analysis of recombinant LOXL2 in three forms: a full-length LOXL2 (fl-LOXL2) containing a nearly stoichiometric amount of LTQ, Δ1-2SRCR-LOXL2 (SRCR1 and SRCR2 are truncated) in the precursor form, and Δ1-3SRCR-LOXL2 (SRCR1, SRCR2, SRCR3 are truncated) in a mixture of the precursor and the mature forms. We detected a set of five disulfide bonds that is conserved in both the precursor and the mature recombinant LOXL2s. In addition, we detected a set of four alternative disulfide bonds in low abundance that is not associated with the mature LOXL2. These results suggest that the major set of five disulfide bonds is retained post-LTQ formation.
Collapse
Affiliation(s)
| | | | | | - Heather Desaire
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (E.P.G.); (H.-J.M.)
| | - Minae Mure
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (E.P.G.); (H.-J.M.)
| |
Collapse
|
22
|
Peng T, Lin S, Meng Y, Gao P, Wu P, Zhi W, Ding W, Cao C, Wu P. LOXL2 small molecule inhibitor restrains malignant transformation of cervical cancer cells by repressing LOXL2-induced epithelial-mesenchymal transition (EMT). Cell Cycle 2022; 21:1827-1841. [PMID: 35509127 PMCID: PMC9359382 DOI: 10.1080/15384101.2022.2073047] [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] [Indexed: 12/24/2022] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) is a member of the lysine oxidase (LOX) family. Although its overexpression is known to play pivotal roles in carcinogenesis, its involvement in cervical cancer remains undefined. Here, we comprehensively explored the expression level and functional mechanism of LOXL2 in cervical cancer using bioinformatics and experimental methods. Bioinformatics analysis revealed that LOXL2 was significantly upregulated in cervical cancer compared to normal tissues. Enrichment analysis showed that most positively or negatively correlated genes of LOXL2 were correlated with extracellular matrix (ECM) formation and epithelial-mesenchymal transition (EMT). Further experiments confirmed that overexpression of LOXL2 greatly enhanced the malignant transformation abilities (e.g., proliferation, invasion, and migration) of cervical cancer cells via mediation of EMT. Furthermore, the small molecule inhibitor of LOXL2 ((2-Chloropyridin-4-yl) methanamine hydrochloride) significantly decreased the invasive ability of cervical cancer by reversing the process of LOXL2-induced EMT. In summary, LOXL2 may be a promising diagnostic and therapeutic biomarker for cervical cancer, and its small molecule inhibitor may be an effective anti-tumor drug.
Collapse
Affiliation(s)
- Ting Peng
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,These authors contributed equally to this work
| | - Shitong Lin
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,These authors contributed equally to this work
| | - Yifan Meng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Peipei Gao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Wu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhua Zhi
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wencheng Ding
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Canhui Cao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, P.R. China
| | - Peng Wu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
23
|
Meier AA, Moon HJ, Toth R, Folta-Stogniew E, Kuczera K, Middaugh CR, Mure M. Oligomeric States and Hydrodynamic Properties of Lysyl Oxidase-Like 2. Biomolecules 2021; 11:biom11121846. [PMID: 34944490 PMCID: PMC8699698 DOI: 10.3390/biom11121846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) has emerged as a promising therapeutic target against metastatic/invasive tumors and organ and tissue fibrosis. LOXL2 catalyzes the oxidative deamination of lysine and hydroxylysine residues in extracellular matrix (ECM) proteins to promote crosslinking of these proteins, and thereby plays a major role in ECM remodeling. LOXL2 secretes as 100-kDa full-length protein (fl-LOXL2) and then undergoes proteolytic cleavage of the first two scavenger receptor cysteine-rich (SRCR) domains to yield 60-kDa protein (Δ1-2SRCR-LOXL2). This processing does not affect the amine oxidase activity of LOXL2 in vitro. However, the physiological importance of this cleavage still remains elusive. In this study, we focused on characterization of biophysical properties of fl- and Δ1-2SRCR-LOXL2s (e.g., oligomeric states, molecular weights, and hydrodynamic radii in solution) to gain insight into the structural role of the first two SRCR domains. Our study reveals that fl-LOXL2 exists predominantly as monomer but also dimer to the lesser extent when its concentration is <~1 mM. The hydrodynamic radius (Rh) determined by multi-angle light scattering coupled with size exclusion chromatography (SEC-MALS) indicates that fl-LOXL2 is a moderately asymmetric protein. In contrast, Δ1-2SRCR-LOXL2 exists solely as monomer and its Rh is in good agreement with the predicted value. The Rh values calculated from a 3D modeled structure of fl-LOXL2 and the crystal structure of the precursor Δ1-2SRCR-LOXL2 are within a reasonable margin of error of the values determined by SEC-MALS for fl- and Δ1-2SRCR-LOXL2s in mature forms in this study. Based on superimposition of the 3D model and the crystal structure of Δ1-2SRCR-LOXL2 (PDB:5ZE3), we propose a configuration of fl-LOXL2 that explains the difference observed in Rh between fl- and Δ1-2SRCR-LOXL2s in solution.
Collapse
Affiliation(s)
- Alex A. Meier
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (H.-J.M.); (K.K.)
| | - Hee-Jung Moon
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (H.-J.M.); (K.K.)
| | - Ronald Toth
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA; (R.T.IV); (C.R.M.)
| | - Ewa Folta-Stogniew
- W.M. Keck Biotechnology Resource Laboratory, Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Krzysztof Kuczera
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (H.-J.M.); (K.K.)
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - C. Russell Middaugh
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA; (R.T.IV); (C.R.M.)
| | - Minae Mure
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA; (A.A.M.); (H.-J.M.); (K.K.)
- Correspondence:
| |
Collapse
|
24
|
The Hepatic Sinusoid in Chronic Liver Disease: The Optimal Milieu for Cancer. Cancers (Basel) 2021; 13:cancers13225719. [PMID: 34830874 PMCID: PMC8616349 DOI: 10.3390/cancers13225719] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary During the development of chronic liver disease, the hepatic sinusoid undergoes major changes that further compromise the hepatic function, inducing persistent inflammation and the formation of scar tissue, together with alterations in liver hemodynamics. This diseased background may induce the formation and development of hepatocellular carcinoma (HCC), which is the most common form of primary liver cancer and a major cause of mortality. In this review, we describe the ways in which the dysregulation of hepatic sinusoidal cells—including liver sinusoidal cells, Kupffer cells, and hepatic stellate cells—may have an important role in the development of HCC. Our review summarizes all of the known sinusoidal processes in both health and disease, and possible treatments focusing on the dysregulation of the sinusoid; finally, we discuss how some of these alterations occurring during chronic injury are shared with the pathology of HCC and may contribute to its development. Abstract The liver sinusoids are a unique type of microvascular beds. The specialized phenotype of sinusoidal cells is essential for their communication, and for the function of all hepatic cell types, including hepatocytes. Liver sinusoidal endothelial cells (LSECs) conform the inner layer of the sinusoids, which is permeable due to the fenestrae across the cytoplasm; hepatic stellate cells (HSCs) surround LSECs, regulate the vascular tone, and synthetize the extracellular matrix, and Kupffer cells (KCs) are the liver-resident macrophages. Upon injury, the harmonic equilibrium in sinusoidal communication is disrupted, leading to phenotypic alterations that may affect the function of the whole liver if the damage persists. Understanding how the specialized sinusoidal cells work in coordination with each other in healthy livers and chronic liver disease is of the utmost importance for the discovery of new therapeutic targets and the design of novel pharmacological strategies. In this manuscript, we summarize the current knowledge on the role of sinusoidal cells and their communication both in health and chronic liver diseases, and their potential pharmacologic modulation. Finally, we discuss how alterations occurring during chronic injury may contribute to the development of hepatocellular carcinoma, which is usually developed in the background of chronic liver disease.
Collapse
|
25
|
Lyakhovich MS, Murashkina AV, Panchenko SP, Averin AD, Abel AS, Maloshitskaya OA, Savelyev EN, Orlinson BS, Novakov IA, Beletskaya IP. Arylation of Adamantanamines: XI. Comparison of the Catalytic Efficiency of Palladium and Copper Complexes in Reactions of Adamantanamines with Fluorinated 2-Bromopyridines. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1070428021050031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Lin L, Zou H, Li W, Xu LY, Li EM, Dong G. Redox Potentials of Disulfide Bonds in LOXL2 Studied by Nonequilibrium Alchemical Simulation. Front Chem 2021; 9:797036. [PMID: 34970534 PMCID: PMC8713139 DOI: 10.3389/fchem.2021.797036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) is a metalloenzyme that catalyzes the oxidative deamination ε-amino group of lysine. It is found that LOXL2 is a promotor for the metastasis and invasion of cancer cells. Disulfide bonds are important components in LOXL2, and they play a stabilizing role for protein structure or a functional role for regulating protein bioactivity. The redox potential of disulfide bond is one important property to determine the functional role of disulfide bond. In this study, we have calculated the reduction potential of all the disulfide bonds in LOXL2 by non-equilibrium alchemical simulations. Our results show that seven of seventeen disulfide bonds have high redox potentials between -182 and -298 mV and could have a functional role, viz., Cys573-Cys625, Cys579-Cys695, Cys657-Cys673, and Cys663-Cys685 in the catalytic domain, Cys351-Cys414, Cys464-Cys530, and Cys477-Cys543 in the scavenger receptor cysteine-rich (SRCR) domains. The disulfide bond of Cys351-Cys414 is predicted to play an allosteric function role, which could affect the metastasis and invasion of cancer cells. Other functional bonds have a catalytic role related to enzyme activity. The rest of disulfide bonds are predicted to play a structural role. Our study provides an important insight for the classification of disulfide bonds in LOXL2 and can be utilized for the drug design that targets the cysteine residues in LOXL2.
Collapse
Affiliation(s)
- Lirui Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- Medical Informatics Research Center, Shantou University Medical College, Shantou, China
| | - Haiying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| | - Wenjin Li
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Li-Yan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
- Cancer Research Center, Shantou University Medical College, Shantou, China
- *Correspondence: Li-Yan Xu, ; En-Min Li, ; Geng Dong,
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
- *Correspondence: Li-Yan Xu, ; En-Min Li, ; Geng Dong,
| | - Geng Dong
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- Medical Informatics Research Center, Shantou University Medical College, Shantou, China
- *Correspondence: Li-Yan Xu, ; En-Min Li, ; Geng Dong,
| |
Collapse
|
27
|
Roles of Lysyl Oxidase Family Members in the Tumor Microenvironment and Progression of Liver Cancer. Int J Mol Sci 2020; 21:ijms21249751. [PMID: 33371259 PMCID: PMC7766343 DOI: 10.3390/ijms21249751] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
The lysyl oxidase (LOX) family members are secreted copper-dependent amine oxidases, comprised of five paralogues: LOX and LOX-like l-4 (LOXL1-4), which are characterized by catalytic activity contributing to the remodeling of the cross-linking of the structural extracellular matrix (ECM). ECM remodeling plays a key role in the angiogenesis surrounding tumors, whereby a corrupt tumor microenvironment (TME) takes shape. Primary liver cancer includes hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), ranked as the seventh most common cancer globally, with limited therapeutic options for advanced stages. In recent years, a growing body of evidence has revealed the key roles of LOX family members in the pathogenesis of liver cancer and the shaping of TME, indicating their notable potential as therapeutic targets. We herein review the clinical value and novel biological roles of LOX family members in tumor progression and the TME of liver cancers. In addition, we highlight recent insights into their mechanisms and their potential involvement in the development of target therapy for liver cancer.
Collapse
|
28
|
Wen B, Xu LY, Li EM. LOXL2 in cancer: regulation, downstream effectors and novel roles. Biochim Biophys Acta Rev Cancer 2020; 1874:188435. [PMID: 32976981 DOI: 10.1016/j.bbcan.2020.188435] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/19/2020] [Accepted: 09/19/2020] [Indexed: 02/05/2023]
Abstract
Lysyl oxidase-like 2 (LOXL2) is a copper and lysine tyrosyl-quinone (LTQ)-dependent amine oxidase belonging to the lysyl oxidase (LOX) family, the canonical function of which is to catalyze the crosslinking of elastin and collagen in the extracellular matrix (ECM). Many studies have revealed that the aberrant expression of LOXL2 in multiple cancers is associated with epithelial-mesenchymal transition (EMT), metastasis, poor prognosis, chemoradiotherapy resistance, and tumor progression. LOXL2 is regulated in many ways, such as transcriptional regulation, alternative splicing, microRNA regulation, posttranslational modification, and cleavage. Beyond affecting the extracellular environment, various intracellular roles, such as oxidation and deacetylation activities in the nucleus, have been reported for LOXL2. Additionally, LOXL2 contributes to tumor cell invasion by promoting cytoskeletal reorganization. Targeting LOXL2 has become a potential therapeutic strategy to combat many types of cancers. Here, we provide an overview of the regulation and downstream effectors of LOXL2 and discuss the intracellular role of LOXL2 in cancer.
Collapse
Affiliation(s)
- Bing Wen
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| |
Collapse
|
29
|
van Haaften WT, Blokzijl T, Hofker HS, Olinga P, Dijkstra G, Bank RA, Boersema M. Intestinal stenosis in Crohn's disease shows a generalized upregulation of genes involved in collagen metabolism and recognition that could serve as novel anti-fibrotic drug targets. Therap Adv Gastroenterol 2020; 13:1756284820952578. [PMID: 32922514 PMCID: PMC7457685 DOI: 10.1177/1756284820952578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/31/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND AND AIMS Crohn's disease (CD) can be complicated by intestinal fibrosis. Pharmacological therapies against intestinal fibrosis are not available. The aim of this study was to determine whether pathways involved in collagen metabolism are upregulated in intestinal fibrosis, and to discuss which drugs might be suitable to inhibit excessive extracellular matrix formation targeting these pathways. METHODS Human fibrotic and non-fibrotic terminal ileum was obtained from patients with CD undergoing ileocecal resection due to stenosis. Genes involved in collagen metabolism were analyzed using a microfluidic low-density TaqMan array. A literature search was performed to find potential anti-fibrotic drugs that target proteins/enzymes involved in collagen synthesis, its degradation and its recognition. RESULTS mRNA expression of collagen type I (COL1A1, 0.76 ± 0.28 versus 37.82 ± 49.85, p = 0.02) and III (COL3A1, 2.01 ± 2.61 versus 68.65 ± 84.07, p = 0.02) was increased in fibrotic CD compared with non-fibrotic CD. mRNA expression of proteins involved in both intra- and extracellular post-translational modification of collagens (prolyl- and lysyl hydroxylases, lysyl oxidases, chaperones), collagen-degrading enzymes (MMPs and cathepsin-K), and collagen receptors were upregulated in the fibrosis-affected part. A literature search on the upregulated genes revealed several potential anti-fibrotic drugs. CONCLUSION Expression of genes involved in collagen metabolism in intestinal fibrosis affected terminal ileum of patients with CD reveals a plethora of drug targets. Inhibition of post-translational modification and altering collagen metabolism might attenuate fibrosis formation in the intestine in CD. Which compound has the highest potential depends on a combination anti-fibrotic efficacy and safety, especially since some of the enzymes play key roles in the physiology of collagen.
Collapse
Affiliation(s)
- Wouter Tobias van Haaften
- Department of Gastroenterology and Hepatology,
University Medical Center Groningen, University of Groningen, Groningen, the
Netherlands
- Department of Pharmaceutical Technology and
Biopharmacy, University of Groningen, Groningen, the Netherlands
| | - Tjasso Blokzijl
- Department of Laboratory Medicine, University of
Groningen, University Medical Center Groningen, Groningen, The
Netherlands
| | - Hendrik Sijbrand Hofker
- Department of Surgery, University Medical Center
Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and
Biopharmacy, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713
AV, the Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology,
University Medical Center Groningen, University of Groningen, Groningen, the
Netherlands
| | - Ruud A. Bank
- Department of Pathology and Medical Biology,
University Medical Center Groningen, University of Groningen, Groningen, the
Netherlands
| | - Miriam Boersema
- Department of Pharmaceutical Technology and
Biopharmacy, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
30
|
Ye M, Song Y, Pan S, Chu M, Wang ZW, Zhu X. Evolving roles of lysyl oxidase family in tumorigenesis and cancer therapy. Pharmacol Ther 2020; 215:107633. [PMID: 32693113 DOI: 10.1016/j.pharmthera.2020.107633] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
The lysyl oxidase (LOX) family is comprised of LOX and four LOX-like proteins (LOXL1, LOXL2, LOXL3, and LOXL4), and mainly functions in the remodeling of extracellular matrix (ECM) and the cross-linking of collagen and elastic fibers. Recently, a growing body of research has demonstrated that LOX family is critically involved in the regulation of cancer cell proliferation, migration, invasion and metastasis. In this review, we discuss the roles of LOX family members in the development and progression of different types of human cancers. Furthermore, we also describe the potential inhibitors of LOX family proteins and highlight that LOX family might be an important therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shuya Pan
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Man Chu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China..
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
31
|
Using Topomer Comparative Molecular Field Analysis to Elucidate Activity Differences of Aminomethylenethiophene Derivatives as Lysyl Oxidase Inhibitors: Implications for Rational Design of Antimetastatic Agents for Cancer Therapy. J CHEM-NY 2020. [DOI: 10.1155/2020/2036585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Topomer comparative molecular field analysis (topomer CoMFA) is applied to the quantitative structure-activity relationship (QSAR) study of aminomethylenethiophene (AMT) derivatives as lysyl oxidase (LOX) inhibitors. A total of thirty-six AMT derivatives were selected to build the QSAR model. The established topomer CoMFA model has the non-cross-validated correlation coefficient (r2) of 0.912 and the leave-one-out correlation coefficient (q2) of 0.540, which is statistically significant. The theoretically predicted anti-LOX potency agrees well with the experimentally observed inhibitory activity, proving the reasonable predictive ability of the QSAR model. The effect of molecular field information on the LOX inhibition of substituted aminomethylenethiophene was discussed in detail. The structural modification of the aminomethylenethiophene scaffold was carried out, and novel AMT derivatives with theoretically decent LOX inhibition were proposed. The topomer CoMFA modeling could provide a quantitative perspective into the structure-activity relationship of AMT derivatives and potentially speed up the rational design of LOX inhibitors as antimetastatic agents for cancer therapy.
Collapse
|
32
|
Targeting lysyl oxidase (LOX) overcomes chemotherapy resistance in triple negative breast cancer. Nat Commun 2020; 11:2416. [PMID: 32415208 PMCID: PMC7229173 DOI: 10.1038/s41467-020-16199-4] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
Chemoresistance is a major obstacle in triple negative breast cancer (TNBC), the most aggressive breast cancer subtype. Here we identify hypoxia-induced ECM re-modeler, lysyl oxidase (LOX) as a key inducer of chemoresistance by developing chemoresistant TNBC tumors in vivo and characterizing their transcriptomes by RNA-sequencing. Inhibiting LOX reduces collagen cross-linking and fibronectin assembly, increases drug penetration, and downregulates ITGA5/FN1 expression, resulting in inhibition of FAK/Src signaling, induction of apoptosis and re-sensitization to chemotherapy. Similarly, inhibiting FAK/Src results in chemosensitization. These effects are observed in 3D-cultured cell lines, tumor organoids, chemoresistant xenografts, syngeneic tumors and PDX models. Re-expressing the hypoxia-repressed miR-142-3p, which targets HIF1A, LOX and ITGA5, causes further suppression of the HIF-1α/LOX/ITGA5/FN1 axis. Notably, higher LOX, ITGA5, or FN1, or lower miR-142-3p levels are associated with shorter survival in chemotherapy-treated TNBC patients. These results provide strong pre-clinical rationale for developing and testing LOX inhibitors to overcome chemoresistance in TNBC patients. The development of chemoresistance is a major hurdle in triple negative breast cancer (TNBC). Here, the authors show that lysyl oxidase (LOX) is overexpressed in chemoresistant TNBCs, and when inhibited reduces collagen cross-linking, fibronectin fibril assembly, and downstream integrin signalling, overcoming resistance.
Collapse
|
33
|
Tschumperlin DJ, Lagares D. Mechano-therapeutics: Targeting Mechanical Signaling in Fibrosis and Tumor Stroma. Pharmacol Ther 2020; 212:107575. [PMID: 32437826 DOI: 10.1016/j.pharmthera.2020.107575] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Pathological remodeling of the extracellular matrix (ECM) by activated myofibroblasts is a hallmark of fibrotic diseases and desmoplastic tumors. Activation of myofibroblasts occurs in response to fibrogenic tissue injury as well as in tumor-associated fibrotic reactions. The molecular determinants of myofibroblast activation in fibrosis and tumor stroma have traditionally been viewed to include biochemical agents, such as dysregulated growth factor and cytokine signaling, which profoundly alter the biology of fibroblasts, ultimately leading to overexuberant matrix deposition and fibrosis. More recently, compelling evidence has shown that altered mechanical properties of the ECM such as matrix stiffness are major drivers of tissue fibrogenesis by promoting mechano-activation of fibroblasts. In this Review, we discuss new insights into the role of the biophysical microenvironment in the amplified activation of fibrogenic myofibroblasts during the development and progression of fibrotic diseases and desmoplastic tumors. We also summarize novel therapeutic targets for anti-fibrotic therapy based on the mechanobiology of tissue fibrosis and tumor stroma, a class of drugs known as "mechano-therapeutics".
Collapse
Affiliation(s)
- Daniel J Tschumperlin
- Tissue Repair and Mechanobiology Laboratory, Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1(st) St SW, Rochester, MN 55905, USA.
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Brys R, Gibson K, Poljak T, Van Der Plas S, Amantini D. Discovery and development of ASK1 inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2020; 59:101-179. [PMID: 32362327 DOI: 10.1016/bs.pmch.2020.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant activation of mitogen-activated protein kinases (MAPKs) like c-Jun N-terminal kinase (JNK) and p38 is an event involved in the pathophysiology of numerous human diseases. The apoptosis signal-regulating kinase 1 (ASK1) is an upstream target that gets activated only under pathological conditions and as such is a promising target for therapeutic intervention. In the first part of this review the molecular mechanisms leading to ASK1 activation and regulation will be described as well as the evidences supporting a pathogenic role for ASK1 in human disease. In the second part, an update on drug discovery efforts towards the discovery and development of ASK1-targeting therapies will be provided.
Collapse
Affiliation(s)
| | - Karl Gibson
- Sandexis Medicinal Chemistry Ltd, Innovation House Discovery ParkSandwich, Kent, United Kingdom
| | | | | | | |
Collapse
|
35
|
Fan Z, Zheng W, Li H, Wu W, Liu X, Sun Z, Hu H, Du L, Jia Q, Liu Q. LOXL2 upregulates hypoxia‑inducible factor‑1α signaling through Snail‑FBP1 axis in hepatocellular carcinoma cells. Oncol Rep 2020; 43:1641-1649. [PMID: 32323822 PMCID: PMC7107812 DOI: 10.3892/or.2020.7541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Lysyl oxidase‑like 2 (LOXL2), a member of the lysyl oxidase gene family, is involved in the progression of hepatocellular carcinoma progression and metastasis. Increased expression of LOXL2 has been identified in several types of cancer, including hepatocellular carcinoma. Recently, LOXL2 has been reported to promote epithelial‑mesenchymal transition by reducing E‑cadherin expression via the upregulation of Snail expression. The present study provided evidence demonstrating that LOXL2 inhibited the expression of fructose‑1, 6‑biphosphatase (FBP1) and enhanced the glycolysis of Huh7 and Hep3B hepatocellular carcinoma cell lines in a Snail‑dependent manner. Overexpression of the point‑mutated form of LOXL2 [LOXL2(Y689F)], which lacks enzymatic activity, does not affect the expression of Snail1 or FBP1. Notably, targeting extracellular LOXL2 of Huh7 cells with a therapeutic antibody was unable to abolish its regulation on the expression of Snail and FBP1. Knockdown of LOXL2 also interrupted the angiogenesis of Huh7 and Hep3B cells, and this effect could be rescued by the overexpression of Snail. Furthermore, upregulation of hypoxia‑inducible factor 1α (HIF‑1α) and vascular endothelial growth factor (VEGF) expression was observed in Huh7 and Hep3B cells expressing wild‑type LOXL2. Notably, the selective LOXL2 inhibitor LOXL2‑IN‑1 could upregulate the expression of FBP1 and inhibit the expression of Snail, HIF‑1α and VEGF in HCC cells, but not in FBP1‑knockdown cells. The results of the present study indicated that the intracellular activity of LOXL2 upregulated HIF‑1α/VEGF signaling pathways via the Snail‑FBP1 axis, and this phenomenon could be inhibited by LOXL2 inhibition. Collectively, these findings further support that LOXL2 exhibits an important role in the progression of hepatocellular carcinoma and implicates LOXL2 as a potential therapeutic agent for the treatment of this disease.
Collapse
Affiliation(s)
- Zhiyong Fan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Zheng
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Hui Li
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Wujun Wu
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiaogang Liu
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Zhongjie Sun
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Haitian Hu
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Lixue Du
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Qingan Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
36
|
Cu(I)- and Pd(0)-Catalyzed Arylation of Oxadiamines with Fluorinated Halogenobenzenes: Comparison of Efficiency. Molecules 2020; 25:molecules25051084. [PMID: 32121113 PMCID: PMC7179129 DOI: 10.3390/molecules25051084] [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: 02/04/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/25/2022] Open
Abstract
The comparison of the possibilities of Pd- and Cu-catalyzed amination reactions using fluorine-containing aryl bromides and iodides with oxadiamines to produce their N,N′-diaryl derivatives was carried out. The dependence of the reactivity of the aryl halides on the nature of the substituents and halogen atoms as well as on the structure of oxadiamines was investigated. It was found that the copper-catalyzed reactions were somewhat comparable with the palladium-mediated processes in the majority of cases, especially in the reactions with para-fluorine- and para-(trifluoromethyl)-substituted aryl halides, although the necessity to use aryl iodides in the Cu(I)-catalyzed amination was obvious. Pd catalysis was found inevitable for the successful amination of more sterically hindered ortho-(trifluoromethyl)aryl bromides.
Collapse
|
37
|
Chopra V, Sangarappillai RM, Romero‐Canelón I, Jones AM. Lysyl Oxidase Like‐2 (LOXL2): An Emerging Oncology Target. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Vriddhi Chopra
- School of PharmacyUniversity of Birmingham Birmingham B15 2TT UK
| | | | | | - Alan M. Jones
- School of PharmacyUniversity of Birmingham Birmingham B15 2TT UK
| |
Collapse
|
38
|
Vania V, Wang L, Tjakra M, Zhang T, Qiu J, Tan Y, Wang G. The interplay of signaling pathway in endothelial cells-matrix stiffness dependency with targeted-therapeutic drugs. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165645. [PMID: 31866415 DOI: 10.1016/j.bbadis.2019.165645] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/17/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVDs) have been one of the major causes of human deaths in the world. The study of CVDs has focused on cell chemotaxis for decades. With the advances in mechanobiology, accumulating evidence has demonstrated the influence of mechanical stimuli on arterial pathophysiology and endothelial dysfunction that is a hallmark of atherosclerosis development. An increasing number of drugs have been exploited to decrease the stiffness of vascular tissue for CVDs therapy. However, the underlying mechanisms have yet to be explored. This review aims to summarize how matrix stiffness mediates atherogenesis through various important signaling pathways in endothelial cells and cellular mechanophenotype, including RhoA/Rho-associated protein kinase (ROCK), mitogen-activated protein kinase (MAPK), and Hippo pathways. We also highlight the roles of putative mechanosensitive non-coding RNAs in matrix stiffness-mediated atherogenesis. Finally, we describe the usage of tunable hydrogel and its future strategy to improve our knowledge underlying matrix stiffness-mediated CVDs mechanism.
Collapse
Affiliation(s)
- Vicki Vania
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lu Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Marco Tjakra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tao Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
39
|
Targeting the lysyl oxidases in tumour desmoplasia. Biochem Soc Trans 2019; 47:1661-1678. [DOI: 10.1042/bst20190098] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 02/08/2023]
Abstract
The extracellular matrix (ECM) is a fundamental component of tissue microenvironments and its dysregulation has been implicated in a number of diseases, in particular cancer. Tumour desmoplasia (fibrosis) accompanies the progression of many solid cancers, and is also often induced as a result of many frontline chemotherapies. This has recently led to an increased interest in targeting the underlying processes. The major structural components of the ECM contributing to desmoplasia are the fibrillar collagens, whose key assembly mechanism is the enzymatic stabilisation of procollagen monomers by the lysyl oxidases. The lysyl oxidase family of copper-dependent amine oxidase enzymes are required for covalent cross-linking of collagen (as well as elastin) molecules into the mature ECM. This key step in the assembly of collagens is of particular interest in the cancer field since it is essential to the tumour desmoplastic response. LOX family members are dysregulated in many cancers and consequently the development of small molecule inhibitors targeting their enzymatic activity has been initiated by many groups. Development of specific small molecule inhibitors however has been hindered by the lack of crystal structures of the active sites, and therefore alternate indirect approaches to target LOX have also been explored. In this review, we introduce the importance of, and assembly steps of the ECM in the tumour desmoplastic response focussing on the role of the lysyl oxidases. We also discuss recent progress in targeting this family of enzymes as a potential therapeutic approach.
Collapse
|
40
|
Levada K, Omelyanchik A, Rodionova V, Weiskirchen R, Bartneck M. Magnetic-Assisted Treatment of Liver Fibrosis. Cells 2019; 8:E1279. [PMID: 31635053 PMCID: PMC6830324 DOI: 10.3390/cells8101279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic liver injury can be induced by viruses, toxins, cellular activation, and metabolic dysregulation and can lead to liver fibrosis. Hepatic fibrosis still remains a major burden on the global health systems. Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are considered the main cause of liver fibrosis. Hepatic stellate cells are key targets in antifibrotic treatment, but selective engagement of these cells is an unresolved issue. Current strategies for antifibrotic drugs, which are at the critical stage 3 clinical trials, target metabolic regulation, immune cell activation, and cell death. Here, we report on the critical factors for liver fibrosis, and on prospective novel drugs, which might soon enter the market. Apart from the current clinical trials, novel perspectives for anti-fibrotic treatment may arise from magnetic particles and controlled magnetic forces in various different fields. Magnetic-assisted techniques can, for instance, enable cell engineering and cell therapy to fight cancer, might enable to control the shape or orientation of single cells or tissues mechanically. Furthermore, magnetic forces may improve localized drug delivery mediated by magnetism-induced conformational changes, and they may also enhance non-invasive imaging applications.
Collapse
Affiliation(s)
- Kateryna Levada
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Alexander Omelyanchik
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Valeria Rodionova
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
- National University of Science and Technology "MISiS", 119049 Moscow, Russia.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, D-52074 Aachen, Germany.
| | - Matthias Bartneck
- Department of Medicine III, Medical Faculty, RWTH Aachen, D-52074 Aachen, Germany.
| |
Collapse
|
41
|
Lysyl oxidases: from enzyme activity to extracellular matrix cross-links. Essays Biochem 2019; 63:349-364. [DOI: 10.1042/ebc20180050] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
Abstract
AbstractThe lysyl oxidase family comprises five members in mammals, lysyl oxidase (LOX) and four lysyl oxidase like proteins (LOXL1-4). They are copper amine oxidases with a highly conserved catalytic domain, a lysine tyrosylquinone cofactor, and a conserved copper-binding site. They catalyze the first step of the covalent cross-linking of the extracellular matrix (ECM) proteins collagens and elastin, which contribute to ECM stiffness and mechanical properties. The role of LOX and LOXL2 in fibrosis, tumorigenesis, and metastasis, including changes in their expression level and their regulation of cell signaling pathways, have been extensively reviewed, and both enzymes have been identified as therapeutic targets. We review here the molecular features and three-dimensional structure/models of LOX and LOXLs, their role in ECM cross-linking, and the regulation of their cross-linking activity by ECM proteins, proteoglycans, and by inhibitors. We also make an overview of the major ECM cross-links, because they are the ultimate molecular readouts of LOX/LOXL activity in tissues. The recent 3D model of LOX, which recapitulates its known structural and biochemical features, will be useful to decipher the molecular mechanisms of LOX interaction with its various substrates, and to design substrate-specific inhibitors, which are potential antifibrotic and antitumor drugs.
Collapse
|
42
|
Leung L, Niculescu-Duvaz D, Smithen D, Lopes F, Callens C, McLeary R, Saturno G, Davies L, Aljarah M, Brown M, Johnson L, Zambon A, Chambers T, Ménard D, Bayliss N, Knight R, Fish L, Lawrence R, Challinor M, Tang H, Marais R, Springer C. Anti-metastatic Inhibitors of Lysyl Oxidase (LOX): Design and Structure-Activity Relationships. J Med Chem 2019; 62:5863-5884. [PMID: 31070916 PMCID: PMC6937593 DOI: 10.1021/acs.jmedchem.9b00335] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Indexed: 12/11/2022]
Abstract
Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that cross-links collagens and elastin in the extracellular matrix and is a critical mediator of tumor growth and metastatic spread. LOX is a target for cancer therapy, and thus the search for therapeutic agents against LOX has been widely sought. We report herein the medicinal chemistry discovery of a series of LOX inhibitors bearing an aminomethylenethiophene (AMT) scaffold. High-throughput screening provided the initial hits. Structure-activity relationship (SAR) studies led to the discovery of AMT inhibitors with sub-micromolar half-maximal inhibitory concentrations (IC50) in a LOX enzyme activity assay. Further SAR optimization yielded the orally bioavailable LOX inhibitor CCT365623 with good anti-LOX potency, selectivity, pharmacokinetic properties, as well as anti-metastatic efficacy.
Collapse
Affiliation(s)
- Leo Leung
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Dan Niculescu-Duvaz
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Deborah Smithen
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Filipa Lopes
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Cedric Callens
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Robert McLeary
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Grazia Saturno
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Lawrence Davies
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Mohammed Aljarah
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Michael Brown
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Louise Johnson
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Alfonso Zambon
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Tim Chambers
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Delphine Ménard
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Natasha Bayliss
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Ruth Knight
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Laura Fish
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| | - Rae Lawrence
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Mairi Challinor
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - HaoRan Tang
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Richard Marais
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Caroline Springer
- Drug
Discovery Unit, Cancer Research UK Manchester Institute and Molecular Oncology
Team, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield SK10 4TG, United Kingdom
- Cancer
Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, United Kingdom
| |
Collapse
|
43
|
Amendola PG, Reuten R, Erler JT. Interplay Between LOX Enzymes and Integrins in the Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11050729. [PMID: 31130685 PMCID: PMC6562985 DOI: 10.3390/cancers11050729] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/24/2022] Open
Abstract
Members of the lysyl oxidase (LOX) family are secreted copper-dependent amine oxidases that catalyze the covalent crosslinking of collagens and elastin in the extracellular matrix (ECM), an essential process for the structural integrity of all tissues. LOX enzymes can also remodel the tumor microenvironment and have been implicated in all stages of tumor initiation and progression of many cancer types. Changes in the ECM can influence several cancer cell phenotypes. Integrin adhesion complexes (IACs) physically connect cells with their microenvironment. This review article summarizes the main findings on the role of LOX proteins in modulating the tumor microenvironment, with a particular focus on how ECM changes are integrated by IACs to modulate cells behavior. Finally, we discuss how the development of selective LOX inhibitors may lead to novel and effective therapies in cancer treatment.
Collapse
Affiliation(s)
- Pier Giorgio Amendola
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Raphael Reuten
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Janine Terra Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
44
|
Kii I. Periostin Functions as a Scaffold for Assembly of Extracellular Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:23-32. [DOI: 10.1007/978-981-13-6657-4_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Hajdú I, Kardos J, Major B, Fabó G, Lőrincz Z, Cseh S, Dormán G. Inhibition of the LOX enzyme family members with old and new ligands. Selectivity analysis revisited. Bioorg Med Chem Lett 2018; 28:3113-3118. [PMID: 30098867 DOI: 10.1016/j.bmcl.2018.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 01/08/2023]
Abstract
Lysyl oxidase (LOX) enzymes as potential drug targets maintain constant attention in the therapy of fibrosis, cancer and metastasis. In order to measure the inhibitory activity of small molecules on the LOX enzyme family members a fluorometric activity screening method was developed. During assay validation, previously reported non-selective small inhibitor molecules (BAPN, MCP-1, thiram, disulfiram) were investigated on all of the major LOX enzymes. We confirmed that MCP-1, thiram, disulfiram are in fact pan-inhibitors, while BAPN inhibits only LOX-like enzymes (preferably LOX-like-protein-2, LOXL2) in contrast to the previous reports. We measured the LOX inhibitory profile of a small targeted library generated by 2D ligand-based chemoinformatics methods. Ten hits (10.4% hit rate) were identified, and the compounds showed distinct activity profiles. Potential inhibitors were also identified for LOX-like-protein-3 (LOXL3) and LOX-like-protein-4 (LOXL4), that are considered as emerging drug targets in the therapy of melanoma and gastric cancer.
Collapse
Affiliation(s)
- István Hajdú
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary; Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - József Kardos
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest, Hungary
| | - Balázs Major
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gabriella Fabó
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - Zsolt Lőrincz
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - Sándor Cseh
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - György Dormán
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary.
| |
Collapse
|
46
|
Juillerat-Jeanneret L, Aubert JD, Mikulic J, Golshayan D. Fibrogenic Disorders in Human Diseases: From Inflammation to Organ Dysfunction. J Med Chem 2018; 61:9811-9840. [DOI: 10.1021/acs.jmedchem.8b00294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - John-David Aubert
- Pneumology Division and Transplantation Center, Centre Hospitalier Universitaire Vaudois (CHUV), CH1011 Lausanne, Switzerland
| | - Josip Mikulic
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
47
|
Cosgrove D, Dufek B, Meehan DT, Delimont D, Hartnett M, Samuelson G, Gratton MA, Phillips G, MacKenna DA, Bain G. Lysyl oxidase like-2 contributes to renal fibrosis in Col4α3/Alport mice. Kidney Int 2018; 94:303-314. [PMID: 29759420 DOI: 10.1016/j.kint.2018.02.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/23/2018] [Accepted: 02/08/2018] [Indexed: 12/28/2022]
Abstract
Lysyl oxidase like-2 (LOXL2) is an amine oxidase with both intracellular and extracellular functions. Extracellularly, LOXL2 promotes collagen and elastin crosslinking, whereas intracellularly, LOXL2 has been reported to modify histone H3, stabilize SNAIL, and reduce cell polarity. Although LOXL2 promotes liver and lung fibrosis, little is known regarding its role in renal fibrosis. Here we determine whether LOXL2 influences kidney disease in COL4A3 (-/-) Alport mice. These mice were treated with a small molecule inhibitor selective for LOXL2 or with vehicle and assessed for glomerular sclerosis and fibrosis, albuminuria, blood urea nitrogen, lifespan, pro-fibrotic gene expression and ultrastructure of the glomerular basement membrane. Laminin α2 deposition in the glomerular basement membrane and mesangial filopodial invasion of the glomerular capillaries were also assessed. LOXL2 inhibition significantly reduced interstitial fibrosis and mRNA expression of MMP-2, MMP-9, TGF-β1, and TNF-α. LOXL2 inhibitor treatment also reduced glomerulosclerosis, expression of MMP-10, MMP-12, and MCP-1 mRNA in glomeruli, and decreased albuminuria and blood urea nitrogen. Mesangial filopodial invasion of the capillary tufts was blunted, as was laminin α2 deposition in the glomerular basement membrane, and glomerular basement membrane ultrastructure was normalized. There was no effect on lifespan. Thus, LOXL2 plays an important role in promoting both glomerular and interstitial pathogenesis associated with Alport syndrome in mice. Other etiologies of chronic kidney disease are implicated with our observations.
Collapse
Affiliation(s)
| | - Brianna Dufek
- Boys Town National Research Hospital, Omaha, Nebraska, USA
| | | | - Duane Delimont
- Boys Town National Research Hospital, Omaha, Nebraska, USA
| | | | - Gina Samuelson
- Boys Town National Research Hospital, Omaha, Nebraska, USA
| | | | | | | | | |
Collapse
|
48
|
Grosche J, Meißner J, Eble JA. More than a syllable in fib-ROS-is: The role of ROS on the fibrotic extracellular matrix and on cellular contacts. Mol Aspects Med 2018; 63:30-46. [PMID: 29596842 DOI: 10.1016/j.mam.2018.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023]
Abstract
Fibrosis is characterized by excess deposition of extracellular matrix (ECM). However, the ECM changes during fibrosis not only quantitatively but also qualitatively. Thus, the composition is altered as the expression of various ECM proteins changes. Moreover, also posttranslational modifications, secretion, deposition and crosslinkage as well as the proteolytic degradation of ECM components run differently during fibrosis. As several of these processes involve redox reactions and some of them are even redox-regulated, reactive oxygen species (ROS) influence fibrotic diseases. Redox regulation of the ECM has not been studied intensively, although evidences exist that the alteration of the ECM, including the redox-relevant processes of its formation and degradation, may be of key importance not only as a cause but also as a consequence of fibrotic diseases. Myofibroblasts, which have differentiated from fibroblasts during fibrosis, produce most of the ECM components and in return obtain important environmental cues of the ECM, including their redox-dependent fibrotic alterations. Thus, myofibroblast differentiation and fibrotic changes of the ECM are interdependent processes and linked with each other via cell-matrix contacts, which are mediated by integrins and other cell adhesion molecules. These cell-matrix contacts are also regulated by redox processes and by ROS. However, most of the redox-catalyzing enzymes are localized within cells. Little is known about redox-regulating enzymes, especially the ones that control the formation and cleavage of redox-sensitive disulfide bridges within the extracellular space. They are also important players in the redox-regulative crosstalk between ECM and cells during fibrosis.
Collapse
Affiliation(s)
- Julius Grosche
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Juliane Meißner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany.
| |
Collapse
|
49
|
Afratis NA, Klepfish M, Karamanos NK, Sagi I. The apparent competitive action of ECM proteases and cross-linking enzymes during fibrosis: Applications to drug discovery. Adv Drug Deliv Rev 2018; 129:4-15. [PMID: 29627371 DOI: 10.1016/j.addr.2018.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/11/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Progressive loss of organ function in most organs is associated with fibrosis, a tissue state associated with abnormal matrix buildup. If highly progressive, the fibrotic process eventually leads to organ failure and death. Fibrosis is a basic connective tissue lesion defined by the increase in the amount of fibrillar extracellular matrix (ECM) components in a tissue or organ. In addition, intrinsic changes in important structural cells can induce the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. ECM enzymes belonging to the family of matrix metalloproteinases (MMPs) and lysyl oxidases (LOXs) play a crucial role in ECM remodeling and regeneration. MMPs have a catalytic role in degradation of ECM, whereas LOX/LOXLs mediate ECM, especially collagen, cross-linking and stiffening. Importantly, enzymes from both families are elevated during the fibrotic response to tissue injury and its resolution. Yet, the apparent molecular competition or antagonistic activities of these enzyme families during the various stages of fibrosis is often overlooked. In this review, we discuss the diverse roles of MMPs and LOX/LOXL2 in chronic organ fibrosis. Finally, we review contemporary therapeutic strategies for fibrosis treatment, based on neutralization of MMP and LOX activity, as well as the development of novel drug delivery approaches.
Collapse
Affiliation(s)
- Nikolaos A Afratis
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mordehay Klepfish
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26500, Greece
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
50
|
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: 8.0] [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]
|