101
|
Withana NP, Garland M, Verdoes M, Ofori LO, Segal E, Bogyo M. Labeling of active proteases in fresh-frozen tissues by topical application of quenched activity-based probes. Nat Protoc 2015; 11:184-91. [PMID: 26716706 DOI: 10.1038/nprot.2016.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Active enzymes, such as proteases, often serve as valuable biomarkers for various disease pathologies. Therefore, methods to detect specific enzyme activities in biological samples can provide information to guide disease detection and diagnosis and to increase our understanding of the biological roles of specific enzyme targets. In this protocol, we outline methods for the topical application of fluorescently quenched activity-based probes (qABPs) to fresh-frozen tissue samples. This technique enables rapid imaging of enzyme activity at cellular resolution, and it can be combined with antibody labeling for immunodiagnosis. In this method, fresh-frozen tissue sections are fixed, incubated with the probe and imaged using fluorescence microscopy. This provides an advance over classical immunohistochemistry (IHC) in that it is rapid (4-8 h) and inexpensive, and it provides information on enzyme activity. Furthermore, it can be used with any of the growing number of fluorescent ABPs to provide data for more effective disease monitoring and diagnosis.
Collapse
Affiliation(s)
- Nimali P Withana
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Megan Garland
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Martijn Verdoes
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Leslie O Ofori
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Ehud Segal
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Department of Chemical Systems and Biology, Stanford University School of Medicine, Stanford, California, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
102
|
Ruan H, Hao S, Young P, Zhang H. Targeting Cathepsin B for Cancer Therapies. HORIZONS IN CANCER RESEARCH 2015; 56:23-40. [PMID: 26623174 PMCID: PMC4662557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cathepsin B is a member of the papain family of cysteine proteases normally present in the lysosome, but it can translocate and function to degrade components of the extracellular matrix. It exhibits carboxyopeptidase, peptidyldipepidase, and endopeptidase activity. Aberrant overexpression of cathepsin B has been reported in invasive and metastatic cancers, including breast cancer, melanoma and colorectal cancer. It has been shown that oncogenic activation, such as the signaling of the ErbB pathways, can lead to cathepsin B overexpression. The degradation of the extracellular matrix is a key factor for cathepsin B to contribute to development and metastasis of tumors. An example of substrates for cathepsin B is E-cadherin, which is involved in adherens junctions, and the downregulation of E-cadherin in cancer is directly linked to invasion and metastasis. Recent studies also point to a role for cathepsin B in macrophages in the tumor microenvironment. The structure of cathepsin B is crystallographically solved, and several highly selective and potent inhibitors for cathepsin B have been developed. Yet it remains to be a challenge to demonstrate the clinical utility or benefit of any cathepsin B inhibitor. As cathepsin B is required for a cellular process called lysosomal membrane permeabilization (LMP), inhibition of cathepsin B would protect cancer cells from cell death induced by chemotherapeutic agents. It is expected that combining cathepsin B inhibitors with other approaches, such as nanoparticles, to direct the inhibition to the extracellular space may lead to better clinical approaches to treat cancers and metastasis.
Collapse
Affiliation(s)
| | | | | | - Hongtao Zhang
- Corresponding author: Hongtao Zhang, Ph.D., 252 John Morgan Building, 3620 Hamilton Walk, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, Phone: 215-573-9256, Fax: 215-898-2401,
| |
Collapse
|
103
|
Verbovšek U, Van Noorden CJ, Lah TT. Complexity of cancer protease biology: Cathepsin K expression and function in cancer progression. Semin Cancer Biol 2015; 35:71-84. [DOI: 10.1016/j.semcancer.2015.08.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 12/18/2022]
|
104
|
Abstract
Lysosomes are membrane-bound intracellular organelles that receive macromolecules delivered by endocytosis, phagocytosis, and autophagy for degradation and recycling. Over the last decade, advances in lysosome research have established a broad role for the lysosome in the pathophysiology of disease. In this review, we highlight the recent discoveries in lysosome biology, with an emphasis on their implications for cancer therapy. We focus on targeting the lysosome in cancer by exploring lysosomal biogenesis and its role in the crosstalk between apoptosis and autophagy. We also discuss how lysosomal inhibition could emerge as a new therapeutic strategy to overcome drug resistance in cancer.
Collapse
Affiliation(s)
- Shengfu Piao
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ravi K Amaravadi
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
105
|
Sun T, Jiang D, Zhang L, Su Q, Mao W, Jiang C. Expression profile of cathepsins indicates the potential of cathepsins B and D as prognostic factors in breast cancer patients. Oncol Lett 2015; 11:575-583. [PMID: 26870250 PMCID: PMC4727043 DOI: 10.3892/ol.2015.3960] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 06/16/2015] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is one of the most prevalent types of cancer in women and contributes to 32% of all female cancer cases. Cathepsins, a family of proteins, are known to have a critical role in human cancers. However, previous studies on the systematic analysis of the role of cathepsin family members in breast cancer are limited. The aim of the present study was to identify biological markers to predict prognosis and treatment response of breast cancer patients, as well as to elucidate novel therapeutic targets. The present study analyzed the expression of six members of cathepsin family, including cathepsins B, G, D, K, L and V in 188 breast cancer tissue specimens using immunohistochemistry. The data showed that all members of the tested cathepsin families featured cytoplasmic staining. Notably, expression of cathepsin L was associated with advanced tumor stages, while cathepsins B and K expression levels were associated with positive estrogen receptor expression; in addition, cathepsin K expression was also demonstrated to be associated with progesterone receptor expression. Cathepsins V and D expression levels were found to be associated with breast cancer metastasis, while the expression levels of cathepsins B and D were associated with poor disease-free survival in breast cancer patients. In addition, univariate analysis demonstrated that breast cancer metastasis to the bone and the expression of cathepsin B protein were associated with poor disease-free survival. In conclusion, the results of the present study indicated that the altered expression of cathepsins, in particular cathepsins B and D, contributed to the progression of breast cancer and poor disease-free survival in breast cancer patients.
Collapse
Affiliation(s)
- Tao Sun
- Department of Medical Oncology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110000, P.R. China
| | - Daqing Jiang
- Department of Breast Oncology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110000, P.R. China
| | - Liang Zhang
- Department of Medical Oncology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110000, P.R. China
| | - Qinglong Su
- Department of Medical Oncology, Central Hospital of Chaoyang, Chaoyang, Liaoning 100000, P.R. China
| | - Wanli Mao
- Department of Medical Oncology, Yongchuan People's Hospital, Yongchuan, Chongqing 404000, P.R. China
| | - Cui Jiang
- Department of Medical Oncology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
106
|
Weber E, Barbulescu E, Medek R, Reinheckel T, Sameni M, Anbalagan A, Moin K, Sloane BF. Cathepsin B-deficient mice as source of monoclonal anti-cathepsin B antibodies. Biol Chem 2015; 396:277-81. [PMID: 25205719 DOI: 10.1515/hsz-2014-0191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/16/2014] [Indexed: 01/18/2023]
Abstract
Cathepsin B has been demonstrated to be involved in several proteolytic processes that support tumor progression and metastasis and neurodegeneration. To further clarify its role, defined monoclonal antibodies are needed. As the primary structure of human cathepsin B is almost identical to that of the mouse, cathepsin B-deficient mice were used in a novel approach for generating such antibodies, providing the chance of an increased immune response to the antigen, human cathepsin B. Thirty clones were found to produce cathepsin B-specific antibodies. Seven of these antibodies were used to detect cathepsin B in MCF10-DCIS human breast cancer cells by immunocytochemistry and immunoblotting. Five different binding sites were identified by epitope mapping giving the opportunity to combine these antibodies in oligoclonal antibody mixtures for an improved detection of cathepsin B.
Collapse
|
107
|
Ramalho SD, Sharma R, White JK, Aggarwal N, Chalasani A, Sameni M, Moin K, Vieira PC, Turro C, Kodanko JJ, Sloane BF. Imaging Sites of Inhibition of Proteolysis in Pathomimetic Human Breast Cancer Cultures by Light-Activated Ruthenium Compound. PLoS One 2015; 10:e0142527. [PMID: 26562785 PMCID: PMC4643019 DOI: 10.1371/journal.pone.0142527] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/22/2015] [Indexed: 11/21/2022] Open
Abstract
The cysteine protease cathepsin B has been causally linked to progression and metastasis of breast cancers. We demonstrate inhibition by a dipeptidyl nitrile inhibitor (compound 1) of cathepsin B activity and also of pericellular degradation of dye-quenched collagen IV by living breast cancer cells. To image, localize and quantify collagen IV degradation in real-time we used 3D pathomimetic breast cancer models designed to mimic the in vivo microenvironment of breast cancers. We further report the synthesis and characterization of a caged version of compound 1, [Ru(bpy)2(1)2](BF4)2 (compound 2), which can be photoactivated with visible light. Upon light activation, compound 2, like compound 1, inhibited cathepsin B activity and pericellular collagen IV degradation by the 3D pathomimetic models of living breast cancer cells, without causing toxicity. We suggest that caged inhibitor 2 is a prototype for cathepsin B inhibitors that can control both the site and timing of inhibition in cancer.
Collapse
Affiliation(s)
- Suelem D. Ramalho
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Rajgopal Sharma
- Department of Chemistry, Wayne State University, Detroit, Michigan, United States of America
| | - Jessica K. White
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Neha Aggarwal
- Department of Physiology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Anita Chalasani
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Mansoureh Sameni
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Kamiar Moin
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Paulo C. Vieira
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Claudia Turro
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Jeremy J. Kodanko
- Department of Chemistry, Wayne State University, Detroit, Michigan, United States of America
- * E-mail: (BFS); (JJK)
| | - Bonnie F. Sloane
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- * E-mail: (BFS); (JJK)
| |
Collapse
|
108
|
Cysteine proteases as therapeutic targets: does selectivity matter? A systematic review of calpain and cathepsin inhibitors. Acta Pharm Sin B 2015; 5:506-19. [PMID: 26713267 PMCID: PMC4675809 DOI: 10.1016/j.apsb.2015.08.001] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/09/2015] [Accepted: 07/14/2015] [Indexed: 01/17/2023] Open
Abstract
Cysteine proteases continue to provide validated targets for treatment of human diseases. In neurodegenerative disorders, multiple cysteine proteases provide targets for enzyme inhibitors, notably caspases, calpains, and cathepsins. The reactive, active-site cysteine provides specificity for many inhibitor designs over other families of proteases, such as aspartate and serine; however, a) inhibitor strategies often use covalent enzyme modification, and b) obtaining selectivity within families of cysteine proteases and their isozymes is problematic. This review provides a general update on strategies for cysteine protease inhibitor design and a focus on cathepsin B and calpain 1 as drug targets for neurodegenerative disorders; the latter focus providing an interesting query for the contemporary assumptions that irreversible, covalent protein modification and low selectivity are anathema to therapeutic safety and efficacy.
Collapse
Key Words
- AD, Alzheimer׳s disease
- ALS, amyotrophic lateral sclerosis
- APP, amyloid precursor protein
- APP/PS1, Aβ overexpressing mice APP (K670N/M671L) and PS1 (M146L) mutants
- Ala, alanine
- Alzheimer׳s disease
- AppLon, London familial amyloid precursor protein mutation, APP (V717I)
- AppSwe, Swedish amyloid precursor protein mutation, APP (K670N/M671L)
- Arg, arginine
- Aβ, amyloid β
- Aβ1-42, amyloid β, 42 amino acid protein
- BACE-1, β-amyloid cleaving enzyme
- BBB, blood–brain barrier
- CANP, calcium-activated neutral protease
- CNS, central nervous system
- CREB, cyclic adenosine monophosphate response element binding protein
- CaMKII, Ca2+/calmodulin-dependent protein kinases II
- Calpain
- Cathepsin
- Cdk5/p35, activator of cyclin-dependent kinase 5
- Cysteine protease
- DTT, dithioerythritol
- EGFR, epidermal growth factor receptor
- ERK1/2, extracellular signal-regulated kinase 1/2
- Enzyme inhibitors
- GSH, glutathione
- Gln, glutamine
- Glu, glutamic acid
- Gly, glutamine
- Hsp70.1, heat shock protein 70.1
- Ile, isoleucine
- KO, knockout
- Leu, leucine
- Lys, lysine
- MAP-2, microtubule-associated protein 2
- MMP-9, matrix metalloproteinase 9
- Met, methionine
- NFT, neurofibrilliary tangles
- Neurodegeneration
- Nle, norleucine
- PD, Parkinson׳s disease
- PK, pharmacokinetic
- PKC, protein kinase C
- PTP1B, protein-tyrosine phosphatase 1B
- Phe, phenylalanine
- Pro, proline
- SP, senile plaques
- TBI, traumatic brain injury
- TNF, tumor necrosis factor
- Thr, threonine
- Tyr, tyrosine
- Val, valine
- WRX, Trp-Arg containing epoxysuccinate cysteine protease inhibitor
- WT, wildtype
- isoAsp, isoaspartate
- pGlu, pyroglutamate
- pyroGluAβ, pyroglutamate-amyloid β
Collapse
|
109
|
Ji K, Heyza J, Cavallo-Medved D, Sloane BF. Pathomimetic cancer avatars for live-cell imaging of protease activity. Biochimie 2015; 122:68-76. [PMID: 26375517 DOI: 10.1016/j.biochi.2015.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/10/2015] [Indexed: 12/12/2022]
Abstract
Proteases are essential for normal physiology as well as multiple diseases, e.g., playing a causative role in cancer progression, including in tumor angiogenesis, invasion, and metastasis. Identification of dynamic alterations in protease activity may allow us to detect early stage cancers and to assess the efficacy of anti-cancer therapies. Despite the clinical importance of proteases in cancer progression, their functional roles individually and within the context of complex protease networks have not yet been well defined. These gaps in our understanding might be addressed with: 1) accurate and sensitive tools and methods to directly identify changes in protease activities in live cells, and 2) pathomimetic avatars for cancer that recapitulate in vitro the tumor in the context of its cellular and non-cellular microenvironment. Such avatars should be designed to facilitate mechanistic studies that can be translated to animal models and ultimately the clinic. Here, we will describe basic principles and recent applications of live-cell imaging for identification of active proteases. The avatars optimized by our laboratory are three-dimensional (3D) human breast cancer models in a matrix of reconstituted basement membrane (rBM). They are designated mammary architecture and microenvironment engineering (MAME) models as they have been designed to mimic the structural and functional interactions among cell types in the normal and cancerous human breast. We have demonstrated the usefulness of these pathomimetic avatars for following dynamic and temporal changes in cell:cell interactions and quantifying changes in protease activity associated with these interactions in real-time (4D). We also briefly describe adaptation of the avatars to custom-designed and fabricated tissue architecture and microenvironment engineering (TAME) chambers that enhance our ability to analyze concomitant changes in the malignant phenotype and the associated tumor microenvironment.
Collapse
Affiliation(s)
- Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Joshua Heyza
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Dora Cavallo-Medved
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biological Sciences, University of Windsor, Windsor, Canada.
| | - Bonnie F Sloane
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biological Sciences, University of Windsor, Windsor, Canada.
| |
Collapse
|
110
|
Dykes SS, Friday E, Pruitt K, Cardelli JA. The histone deacetylase inhibitor cambinol prevents acidic pH e-induced anterograde lysosome trafficking independently of sirtuin activity. Biochem Biophys Rep 2015; 3:83-93. [PMID: 29124170 PMCID: PMC5668693 DOI: 10.1016/j.bbrep.2015.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/17/2015] [Accepted: 07/23/2015] [Indexed: 01/03/2023] Open
Abstract
Common features of the solid tumor microenvironment, such as acidic extracellular pH and growth factors, are known to induce the redistribution of lysosomes from a perinuclear region to a position near the plasma membrane. Lysosome/plasma membrane juxtaposition facilitates invasion by allowing for the release of lysosomal proteases, including cathepsin B, which contribute to matrix degradation. In this study we identified the sirtuin 1/sirtuin 2 (SIRT1/2) inhibitor cambinol acts as a drug that inhibits lysosome redistribution and tumor invasion. Treatment of cells with cambinol resulted in a juxtanuclear lysosome aggregation (JLA) similar to that seen upon treatment with the PPARγ agonist, troglitazone (Tro). Like Tro, cambinol required the activity of ERK1/2 in order to induce this lysosome clustering phenotype. However, cambinol did not require the activity of Rab7, suggesting that this drug causes JLA by a mechanism different from what is known for Tro. Additionally, cambinol-induced JLA was not a result of autophagy induction. Further investigation revealed that cambinol triggered JLA independently of its activity as a SIRT1/2 inhibitor, suggesting that this drug could have effects in addition to SIRT1/2 inhibition that could be developed into a novel anti-cancer therapy. Cambinol prevents acidic pHe-induced anterograde lysosome trafficking. Cambinol-mediated lysosome aggregation is not dependent on sirtuin activity. ERK1/2 activity is necessary for cambinol-driven juxtanuclear lysosome aggregation.
Collapse
Affiliation(s)
- Samantha S. Dykes
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Ellen Friday
- Department of Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 7113, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Kevin Pruitt
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - James A. Cardelli
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States
- Corresponding author at: Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, United States.
| |
Collapse
|
111
|
Pantano F, Iuliani M, Zoccoli A, Fioramonti M, De Lisi D, Fioroni I, Ribelli G, Santoni M, Vincenzi B, Tonini G, Santini D. Emerging drugs for the treatment of bone metastasis. Expert Opin Emerg Drugs 2015; 20:637-51. [PMID: 26113304 DOI: 10.1517/14728214.2015.1062876] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Bone metastases are virtually incurable resulting in significant disease morbidity, reduced quality of life and mortality. Bone provides a unique microenvironment whose local interactions with tumor cells offer novel targets for therapeutic interventions. Increased understanding of the pathogenesis of bone disease has led to the discovery and clinical utility of bone-targeted agents other than bisphosphonates and denosumab, currently, the standard of care in this setting. AREAS COVERED In this review, we present the recent advances in molecular targeted therapies focusing on therapies that inhibit bone resorption and/or stimulate bone formation and novel anti-tumoral agents that exerts significant effects on skeletal metastases, nowadays available in clinical practice or in phase of development. EXPERT OPINION New emergent bone target therapies radium-223, mTOR inhibitors, anti-androgens have demonstrated the ability to increase overall survival in bone metastatic patients, other compounds, such as ET-1 and SRC inhibitors, up to now failed to clearly confirm in clinical trials their promising preclinical data.
Collapse
Affiliation(s)
- Francesco Pantano
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Michele Iuliani
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Alice Zoccoli
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Marco Fioramonti
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Delia De Lisi
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Iacopo Fioroni
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Giulia Ribelli
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Matteo Santoni
- b 2 Università Politecnica delle Marche, AOU Ospedali Riuniti, Medical Oncology Department , Ancona, Italy
| | - Bruno Vincenzi
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Giuseppe Tonini
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Daniele Santini
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| |
Collapse
|
112
|
Löser R, Pietzsch J. Cysteine cathepsins: their role in tumor progression and recent trends in the development of imaging probes. Front Chem 2015; 3:37. [PMID: 26157794 PMCID: PMC4477214 DOI: 10.3389/fchem.2015.00037] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/29/2015] [Indexed: 12/16/2022] Open
Abstract
Papain-like cysteine proteases bear an enormous potential as drug discovery targets for both infectious and systemic human diseases. The considerable progress in this field over the last two decades has also raised interest in the visualization of these enzymes in their native context, especially with regard to tumor imaging. After a short introduction to structure and general functions of human cysteine cathepsins, we highlight their importance for drug discovery and development and provide a critical update on the current state of knowledge toward their involvement in tumor progression, with a special emphasis on their role in therapy response. In accordance with a radiopharmaceutical point of view, the main focus of this review article will be the discussion of recently developed fluorescence and radiotracer-based imaging agents together with related molecular probes.
Collapse
Affiliation(s)
- Reik Löser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Dresden, Germany ; Department of Chemistry and Food Chemistry, Technische Universität Dresden Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Dresden, Germany ; Department of Chemistry and Food Chemistry, Technische Universität Dresden Dresden, Germany
| |
Collapse
|
113
|
Wang X, Lao Y, Xu N, Xi Z, Wu M, Wang H, Li X, Tan H, Sun M, Xu H. Oblongifolin C inhibits metastasis by up-regulating keratin 18 and tubulins. Sci Rep 2015; 5:10293. [PMID: 25973684 PMCID: PMC4431421 DOI: 10.1038/srep10293] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 04/08/2015] [Indexed: 12/22/2022] Open
Abstract
Tumor metastasis is the main cause of cancer-related patient death. In this study, we performed a wound healing migration screen to search for a metastatic inhibitor within our library of natural compounds. We found that oblongifolin C (OC), a natural compound extracted from Garcinia yunnanensis Hu, is an effective inhibitor of metastasis in human esophageal squamous carcinoma Eca109 cells. The transwell migration and matrigel invasion assay results also showed that OC inhibits the migration of Eca109 cells and HepG2 cells. OC can increase the expression of tubulin, indicating that OC inhibits metastasis via tubulin aggregation. In addition, the Western blotting, real-time PCR, and immunostaining results indicated that OC increases the expression of keratin18. Furthermore, the knockdown of keratin 18 by small interfering RNAs inhibited the expression of tubulin and increased the metastasis of cancer cells, suggesting that keratin 18 is the upstream signal of tubulin and plays a vital role in metastasis. A subsequent study in a tail vein injection metastasis model showed that OC can significantly inhibit pulmonary metastasis, as revealed by immunohistochemistry staining. Taken together, our results suggest that OC inhibits metastasis through the induction of the expression of keratin 18 and may be useful in cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, P.R. China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| | - Man Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| | - Hua Wang
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, P. R. China
| | - Xiyi Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
| | - Hongsheng Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| | - Menghong Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China
| |
Collapse
|
114
|
Zhou ZJ, Qiu R, Zhang J. Molecular characterization of the cathepsin B of turbot (Scophthalmus maximus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2015; 41:473-483. [PMID: 25326658 DOI: 10.1007/s10695-014-9998-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/08/2014] [Indexed: 06/04/2023]
Abstract
Cathepsin B is an enzymatic protein belonging to the peptidase C1 family. It is involved in diverse physiological and pathological functions that include immune response. In this study, we identified and characterized a cathepsin B homolog (SmCatB) from turbot (Scophthalmus maximus). SmCatB is composed of 330 amino acid residues and possesses typical domain architecture of cathepsin B, which contains a propeptide region and a cysteine protease domain, and the latter processes four conserved residues (Q101, C107, H277, and N297) in the active site. SmCatB shares 80.6-87.6% overall sequence identities with the cathepsin B of a number of teleost. SmCatB expression was detected in a wide range of tissues and upregulated by bacterial infection in a time-dependent manner. Recombinant SmCatB (rSmCatB-WT) purified from Escherichia coli exhibited apparent protease activity, which was optimal at 50 °C and pH 5.5. Compared to rSmCatB-WT, the mutant proteins rSmCatB-C107S, rSmCatB-H277A, and rSmCatB-N297A, which bear C107S, H277A, and N297A mutations, respectively, were significantly reduced in protease activity, with the highest reduction observed with rSmCatB-N297A. These results indicate that SmCatB is a bioactive protease that depends on the conserved structural features and that SmCatB is involved in pathogen-induced immune response.
Collapse
Affiliation(s)
- Ze-jun Zhou
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | | | | |
Collapse
|
115
|
Bian B, Mongrain S, Cagnol S, Langlois MJ, Boulanger J, Bernatchez G, Carrier JC, Boudreau F, Rivard N. Cathepsin B promotes colorectal tumorigenesis, cell invasion, and metastasis. Mol Carcinog 2015; 55:671-87. [PMID: 25808857 PMCID: PMC4832390 DOI: 10.1002/mc.22312] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 02/05/2015] [Accepted: 02/21/2015] [Indexed: 12/14/2022]
Abstract
Cathepsin B is a cysteine proteinase that primarily functions as an endopeptidase within endolysosomal compartments in normal cells. However, during tumoral expansion, the regulation of cathepsin B can be altered at multiple levels, thereby resulting in its overexpression and export outside of the cell. This may suggest a possible role of cathepsin B in alterations leading to cancer progression. The aim of this study was to determine the contribution of intracellular and extracellular cathepsin B in growth, tumorigenesis, and invasion of colorectal cancer (CRC) cells. Results show that mRNA and activated levels of cathepsin B were both increased in human adenomas and in CRCs of all stages. Treatment of CRC cells with the highly selective and non‐permeant cathepsin B inhibitor Ca074 revealed that extracellular cathepsin B actively contributed to the invasiveness of human CRC cells while not essential for their growth in soft agar. Cathepsin B silencing by RNAi in human CRC cells inhibited their growth in soft agar, as well as their invasion capacity, tumoral expansion, and metastatic spread in immunodeficient mice. Higher levels of the cell cycle inhibitor p27Kip1 were observed in cathepsin B‐deficient tumors as well as an increase in cyclin B1. Finally, cathepsin B colocalized with p27Kip1 within the lysosomes and efficiently degraded the inhibitor. In conclusion, the present data demonstrate that cathepsin B is a significant factor in colorectal tumor development, invasion, and metastatic spreading and may, therefore, represent a potential pharmacological target for colorectal tumor therapy. © 2015 The Authors. Molecular Carcinogenesis, published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Benjamin Bian
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sébastien Mongrain
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sébastien Cagnol
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marie-Josée Langlois
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jim Boulanger
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gérald Bernatchez
- Gastroenterology Service, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Julie C Carrier
- Gastroenterology Service, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Boudreau
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Nathalie Rivard
- Department of Anatomy and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
116
|
A novel cysteine cathepsin inhibitor yields macrophage cell death and mammary tumor regression. Oncogene 2015; 34:6066-78. [PMID: 25798843 DOI: 10.1038/onc.2015.51] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Although cysteine cathepsins have been identified as key regulators of cancer growth, their specific role in tumor development remains unclear. Recent studies have shown that high activity levels of tumor cathepsins are primarily a result of increased cathepsin activity in cancer-promoting tumor-associated macrophages (TAMs). To further investigate the role of cysteine cathepsin activity in normal and polarized macrophages, we established in vitro and in vivo models of macrophage differentiation and polarization and used a novel cysteine cathepsin inhibitor, GB111-NH2, to block the activity of cathepsins B, L and S. Here we show that in vitro, cysteine cathepsin inhibition yields both apoptosis and proliferation of macrophages, owing to increased oxidative stress. Proteomic analysis of cathepsin- inhibited macrophages demonstrates inhibition of autophagy, suggesting a likely cause of elevated reactive oxygen species (ROS) levels. In vivo models of mammary cancer further show that cathepsin inhibition yields TAM death owing to increased ROS levels. Strikingly, apoptosis in TAMs yields a seemingly cell non-autonomous death of neighboring cancer cells, and regression of the primary growth. These results show that cysteine cathepsin inhibitors can specifically trigger macrophage cell death and may function as an effective anticancer therapy in tumors with high levels of TAMs.
Collapse
|
117
|
Clézardin P. Physiopathologie des métastases osseuses. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2489-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
118
|
Ramalho SD, de Sousa LRF, Burger MCM, Lima MIS, da Silva MFDGF, Fernandes JB, Vieira PC. Evaluation of flavonols and derivatives as human cathepsin B inhibitor. Nat Prod Res 2015; 29:2212-4. [DOI: 10.1080/14786419.2014.1002404] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
119
|
Brennan-Fournet ME, Huerta M, Zhang Y, Malliaras G, Owens RM. Detection of fibronectin conformational changes in the extracellular matrix of live cells using plasmonic nanoplates. J Mater Chem B 2015; 3:9140-9147. [PMID: 32263128 DOI: 10.1039/c5tb02060c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoplates enable high sensitive spectral monitoring of fibronectin conformational transitions and fibril formation within the extracellular matrix of live cells.
Collapse
Affiliation(s)
| | - Miriam Huerta
- Ecole Nationale Supérieure des Mines
- CMP-EMSE
- Centre Microélectronique de Provence
- 13541 Gardanne
- France
| | - Yi Zhang
- Ecole Nationale Supérieure des Mines
- CMP-EMSE
- Centre Microélectronique de Provence
- 13541 Gardanne
- France
| | - George Malliaras
- Ecole Nationale Supérieure des Mines
- CMP-EMSE
- Centre Microélectronique de Provence
- 13541 Gardanne
- France
| | - Roisin M. Owens
- Ecole Nationale Supérieure des Mines
- CMP-EMSE
- Centre Microélectronique de Provence
- 13541 Gardanne
- France
| |
Collapse
|
120
|
Kędzior M, Seredyński R, Godzik U, Tomczyk D, Gutowicz J, Terlecka E, Całkosiński I, Terlecki G. Inhibition of cathepsin B activity by 2,3,7,8-tetrachlorodibenzo-p-dioxin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:733-737. [PMID: 25163566 DOI: 10.1007/s11356-014-3482-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/18/2014] [Indexed: 06/03/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is the most potent toxic isomer in the dioxin-like family. Due to its resistance to metabolic degradation, this ubiquitous environmental pollutant readily accumulates in multiple organs. Cathepsin B is a lysosomal cysteine protease playing an essential role in the intracellular protein turnover. Alterations in its expression, activity, and localization may facilitate the development of many pathologies, including cancer. TCDD, due to its extremely lipophilic nature, may diffuse through biological membranes and affect lysosomal enzymes, including cathepsins. Therefore, in this study we performed two enzymatic assays, spectrofluorimetry and gelatin zymography, in order to evaluate the effect of TCDD on purified bovine cathepsin B. We showed that the dioxin decreases the enzyme's activity in a dose-dependent manner. The reversibility of TCDD-induced inhibition of the protease was also examined, suggesting that TCDD does not bind covalently to the enzyme's active site, acting rather as a reversible inhibitor.
Collapse
Affiliation(s)
- Mateusz Kędzior
- Department of Physical Chemistry of Microorganisms, Institute of Genetics and Microbiology, University of Wrocław, Przybyszewskiego 63/77, 51-148, Wrocław, Poland
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Xu ZZ, Xiu P, Lv JW, Wang FH, Dong XF, Liu F, Li T, Li J. Integrin αvβ3 is required for cathepsin B-induced hepatocellular carcinoma progression. Mol Med Rep 2014; 11:3499-504. [PMID: 25572981 DOI: 10.3892/mmr.2014.3140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 08/14/2014] [Indexed: 11/05/2022] Open
Abstract
The cysteine protease cathepsin B (Cat B) is important in the progression of tumor cells, however, the function and molecular mechanisms of Cat B in hepatocellular carcinoma (HCC) remain to be elucidated. Our previous study demonstrated that integrin αvβ3 regulated the biological behavior of HCC. The present study demonstrated that Cat B was also important in cell proliferation and apoptosis in HCC. Notably, Cat B was observed to activate the phosphoinositide 3‑kinase (PI3K)/Akt signaling pathway to promote HCC proliferation. Furthermore, inhibition of integrin αvβ3 significantly prevented Cat B‑induced activation of PI3K/Akt and the progression of HCC. Thus, the results of the present study suggested the presence of a Cat B/integrin αvβ3/PI3K/Akt axis in the regulation of the progression of HCC.
Collapse
Affiliation(s)
- Zong-Zhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ju-Wei Lv
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fu-Hai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xiao-Feng Dong
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
122
|
Cotter K, Capecci J, Sennoune S, Huss M, Maier M, Martinez-Zaguilan R, Forgac M. Activity of plasma membrane V-ATPases is critical for the invasion of MDA-MB231 breast cancer cells. J Biol Chem 2014; 290:3680-92. [PMID: 25505184 DOI: 10.1074/jbc.m114.611210] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The vacuolar (H(+))-ATPases (V-ATPases) are a family of ATP-driven proton pumps that couple ATP hydrolysis with translocation of protons across membranes. Previous studies have implicated V-ATPases in cancer cell invasion. It has been proposed that V-ATPases participate in invasion by localizing to the plasma membrane and causing acidification of the extracellular space. To test this hypothesis, we utilized two separate approaches to specifically inhibit plasma membrane V-ATPases. First, we stably transfected highly invasive MDA-MB231 cells with a V5-tagged construct of the membrane-embedded c subunit of the V-ATPase, allowing for extracellular expression of the V5 epitope. We evaluated the effect of addition of a monoclonal antibody directed against the V5 epitope on both V-ATPase-mediated proton translocation across the plasma membrane and invasion using an in vitro Matrigel assay. The addition of anti-V5 antibody resulted in acidification of the cytosol and a decrease in V-ATPase-dependent proton flux across the plasma membrane in transfected but not control (untransfected) cells. These results demonstrate that the anti-V5 antibody inhibits activity of plasma membrane V-ATPases in transfected cells. Addition of the anti-V5 antibody also inhibited in vitro invasion of transfected (but not untransfected) cells. Second, we utilized a biotin-conjugated form of the specific V-ATPase inhibitor bafilomycin. When bound to streptavidin, this compound cannot cross the plasma membrane. Addition of this compound to MDA-MB231 cells also inhibited in vitro invasion. These studies suggest that plasma membrane V-ATPases play an important role in invasion of breast cancer cells.
Collapse
Affiliation(s)
- Kristina Cotter
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Joseph Capecci
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Souad Sennoune
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Markus Huss
- the Department of Biology/Chemistry, Division of Animal Physiology, University of Osnabrück, 49069 Osnabrück, Germany, and
| | - Martin Maier
- the Institute of Organic Chemistry, University of Tuebingen, 72076 Tuebingen, Germany
| | - Raul Martinez-Zaguilan
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Michael Forgac
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111,
| |
Collapse
|
123
|
Inactivation of tristetraprolin in chronic hypoxia provokes the expression of cathepsin B. Mol Cell Biol 2014; 35:619-30. [PMID: 25452305 DOI: 10.1128/mcb.01034-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Macrophages play important roles in many diseases and are frequently found in hypoxic areas. A chronic hypoxic microenvironment alters global cellular protein expression, but molecular details remain poorly understood. Although hypoxia-inducible factor (HIF) is an established transcription factor allowing adaption to acute hypoxia, responses to chronic hypoxia are more complex. Based on a two-dimensional differential gel electrophoresis (2D-DIGE) approach, we aimed to identify proteins that are exclusively expressed under chronic but not acute hypoxia (1% O2). One of the identified proteins was cathepsin B (CTSB), and a knockdown of either HIF-1α or -2α in primary human macrophages pointed to an HIF-2α dependency. Although chromatin immunoprecipitation (ChIP) experiments confirmed HIF-2 binding to a CTSB enhancer in acute hypoxia, an increase of CTSB mRNA was evident only under chronic hypoxia. Along those lines, CTSB mRNA stability increased at 48 h but not at 8 h of hypoxia. However, RNA stability at 8 h of hypoxia was enhanced by a knockdown of tristetraprolin (TTP). Inactivation of TTP under prolonged hypoxia was facilitated by c-Jun N-terminal kinase (JNK), and inhibition of this kinase lowered CTSB mRNA levels and stability. We postulate a TTP-dependent mechanism to explain delayed expression of CTSB under chronic hypoxia.
Collapse
|
124
|
Theocharis AD, Gialeli C, Bouris P, Giannopoulou E, Skandalis SS, Aletras AJ, Iozzo RV, Karamanos NK. Cell-matrix interactions: focus on proteoglycan-proteinase interplay and pharmacological targeting in cancer. FEBS J 2014; 281:5023-42. [PMID: 25333340 PMCID: PMC5036392 DOI: 10.1111/febs.12927] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/04/2014] [Accepted: 07/09/2014] [Indexed: 01/10/2023]
Abstract
Proteoglycans are major constituents of extracellular matrices, as well as cell surfaces and basement membranes. They play key roles in supporting the dynamic extracellular matrix by generating complex structural networks with other macromolecules and by regulating cellular phenotypes and signaling. It is becoming evident, however, that proteolytic enzymes are required partners for matrix remodeling and for modulating cell signaling via matrix constituents. Proteinases contribute to all stages of diseases, particularly cancer development and progression, and contextually participate in either the removal of damaged products or in the processing of matrix molecules and signaling receptors. The dynamic interplay between proteoglycans and proteolytic enzymes is a crucial biological step that contributes to the pathophysiology of cancer and inflammation. Moreover, proteoglycans are implicated in the expression and secretion of proteolytic enzymes and often modulate their activities. In this review, we describe the emerging biological roles of proteoglycans and proteinases, with a special emphasis on their complex interplay. We critically evaluate this important proteoglycan-proteinase interactome and discuss future challenges with respect to targeting this axis in the treatment of cancer.
Collapse
Affiliation(s)
- Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Chrisostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Panagiotis Bouris
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Efstathia Giannopoulou
- Clinical Oncology Laboratory, Division of Oncology, University Hospital of Patras, Patras Medical School, Patras 26110, Greece
| | - Spyros S. Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Alexios J. Aletras
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| |
Collapse
|
125
|
Schmitz J, Beckmann AM, Dudic A, Li T, Sellier R, Bartz U, Gütschow M. 3-Cyano-3-aza-β-amino Acid Derivatives as Inhibitors of Human Cysteine Cathepsins. ACS Med Chem Lett 2014; 5:1076-81. [PMID: 25313316 DOI: 10.1021/ml500238q] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/11/2014] [Indexed: 12/22/2022] Open
Abstract
Nitrile-type inhibitors are known to interact with cysteine proteases in a covalent-reversible manner. The chemotype of 3-cyano-3-aza-β-amino acid derivatives was designed in which the N-cyano group is centrally arranged in the molecule to allow for interactions with the nonprimed and primed binding regions of the target enzymes. These compounds were evaluated as inhibitors of the human cysteine cathepsins K, S, B, and L. They exhibited slow-binding behavior and were found to be exceptionally potent, in particular toward cathepsin K, with second-order rate constants up to 52 900 × 10(3) M(-1) s(-1).
Collapse
Affiliation(s)
- Janina Schmitz
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
- Department
of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, von-Liebig-Strasse 20, D-53359 Rheinbach, Germany
| | - Anna-Madeleine Beckmann
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Adela Dudic
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Tianwei Li
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Robert Sellier
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Ulrike Bartz
- Department
of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, von-Liebig-Strasse 20, D-53359 Rheinbach, Germany
| | - Michael Gütschow
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| |
Collapse
|
126
|
Shi QQ, Xiang JQ, Chen L, Zhan LL, Lv XP. uPA/PAI system, cathepsin B and hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2014; 22:3941-3946. [DOI: 10.11569/wcjd.v22.i26.3941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Urokinase-type plasminogen activator and plasminogen activator inhibitor (uPA/PAI) are a pair of proteolytic enzyme activator/activator inhibitor. Cathepsin B is a lysosomal cysteine protease. It has been proved that cathepsin B can activate uPA. uPA/PAI and cathepsin B are closely related to the invasion, migration and tumor angiogenesis of malignant neoplasms. The uPA/PAI system and cathepsin B play an important role in the occurrence and development of liver cancer.
Collapse
|
127
|
Sun S, Kennedy RT. Droplet electrospray ionization mass spectrometry for high throughput screening for enzyme inhibitors. Anal Chem 2014; 86:9309-14. [PMID: 25137241 PMCID: PMC4165461 DOI: 10.1021/ac502542z] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/19/2014] [Indexed: 12/20/2022]
Abstract
High throughput screening (HTS) is important for identifying molecules with desired properties. Mass spectrometry (MS) is potentially powerful for label-free HTS due to its high sensitivity, speed, and resolution. Segmented flow, where samples are manipulated as droplets separated by an immiscible fluid, is an intriguing format for high throughput MS because it can be used to reliably and precisely manipulate nanoliter volumes and can be directly coupled to electrospray ionization (ESI) MS for rapid analysis. In this study, we describe a "MS Plate Reader" that couples standard multiwell plate HTS workflow to droplet ESI-MS. The MS plate reader can reformat 3072 samples from eight 384-well plates into nanoliter droplets segmented by an immiscible oil at 4.5 samples/s and sequentially analyze them by MS at 2 samples/s. Using the system, a label-free screen for cathepsin B modulators against 1280 chemicals was completed in 45 min with a high Z-factor (>0.72) and no false positives (24 of 24 hits confirmed). The assay revealed 11 structures not previously linked to cathepsin inhibition. For even larger scale screening, reformatting and analysis could be conducted simultaneously, which would enable more than 145,000 samples to be analyzed in 1 day.
Collapse
Affiliation(s)
- Shuwen Sun
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
128
|
The current stage of cathepsin B inhibitors as potential anticancer agents. Future Med Chem 2014; 6:1355-71. [DOI: 10.4155/fmc.14.73] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cathepsin B is a lysosomal cysteine peptidase, with an important role in the development and progression of cancer. It is involved in the degradation of extracellular matrix proteins, a process promoting invasion and metastasis of tumor cells and tumor angiogenesis. Cathepsin B is unique among cathepsins in possessing both carboxypeptidase and endopeptidase activities. While the former is associated with its physiological role, the latter is involved in pathological degradation of the extracellular matrix. Its activities are regulated by different means, the most important being its endogenous inhibitors, the cystatins. In cancer this peptidase/inhibitor balance is altered, leading to harmful cathepsin B activity. The latter can be prevented by exogenous inhibitors. They differ in modes of inhibition, size, structure, binding affinity, selectivity, toxicity and bioavailability. In this article, we review the properties and function of endogenous and exogenous cathepsin B inhibitors and indicate their application as possible anticancer agents.
Collapse
|
129
|
Theoretical insight into the mechanism for the inhibition of the cysteine protease cathepsin B by 1,2,4-thiadiazole derivatives. J Mol Model 2014; 20:2254. [DOI: 10.1007/s00894-014-2254-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/21/2014] [Indexed: 10/25/2022]
|
130
|
Aggarwal N, Sloane BF. Cathepsin B: multiple roles in cancer. Proteomics Clin Appl 2014; 8:427-37. [PMID: 24677670 PMCID: PMC4205946 DOI: 10.1002/prca.201300105] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/05/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
Proteases, including intracellular proteases, play roles at many different stages of malignant progression. Our focus here is cathepsin B, a lysosomal cysteine cathepsin. High levels of cathepsin B are found in a wide variety of human cancers, levels that often induce secretion and association of cathepsin B with the tumor cell membrane. In experimental models, such as transgenic models of murine pancreatic and mammary carcinomas, causal roles for cathepsin B have been demonstrated in initiation, growth/tumor cell proliferation, angiogenesis, invasion, and metastasis. Tumor growth in transgenic models is promoted by cathepsin B in tumor-associated cells, for example, tumor-associated macrophages, as well as in tumor cells. In transgenic models, the absence of cathepsin B has been associated with enhanced apoptosis, yet cathepsin B also has been shown to contribute to apoptosis. Cathepsin B is part of a proteolytic pathway identified in xenograft models of human glioma; targeting only cathepsin B in these tumors is less effective than targeting cathepsin B in combination with other proteases or protease receptors. Understanding the mechanisms responsible for increased expression of cathepsin B in tumors and association of cathepsin B with tumor cell membranes is needed to determine whether targeting cathepsin B could be of therapeutic benefit.
Collapse
Affiliation(s)
- Neha Aggarwal
- Department of Physiology, Wayne State University School of Medicine, Detroit, Ml, USA
| | - Bonnie F. Sloane
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Ml, USA
| |
Collapse
|
131
|
Zhang ZY, Mai Y, Yang H, Dong PY, Zheng XL, Yang GS. CTSB promotes porcine preadipocytes differentiation by degrading fibronectin and attenuating the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2014; 395:53-64. [PMID: 24878992 DOI: 10.1007/s11010-014-2111-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/15/2014] [Indexed: 01/21/2023]
Abstract
The process of preadipocytes differentiation plays a vital role in adipose tissue expansion and many factors are involved in this event. Cathepsin B (CTSB), secreted from lysosome, has been reported in regulating a variety of physiological processes. In this study, we demonstrated CTSB promotes lipid accumulation and adipogenic genes expression in porcine primary preadipocytes by degrading fibronectin (Fn), a key component of extracellular matrix. Lithium chloride (LiCl) is an activator of Wnt/β-catenin signaling through stabilizing β-catenin. We found that CTSB can relieve the anti-adipogenic effects of LiCl, indicating that CTSB could impact Wnt/β-catenin signaling pathway. Interestingly, Fn is an important target gene of Wnt/β-catenin. So we considered that CTSB promote preadipocytes differentiation by suppressing these two pathways.
Collapse
Affiliation(s)
- Zhen-Yu Zhang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, 22 Xinong Road, Yangling, 712100, People's Republic of China
| | | | | | | | | | | |
Collapse
|
132
|
Abstract
The extracellular matrix (ECM) is composed of highly variable and dynamic components that regulate cell behavior. The protein composition and physical properties of the ECM govern cell fate through biochemical and biomechanical mechanisms. This requires a carefully orchestrated and thorough regulation considering that a disturbed ECM can have serious consequences and lead to pathological conditions like cancer. In breast cancer, many ECM proteins are significantly deregulated and specific matrix components promote tumor progression and metastatic spread. Intriguingly, several ECM proteins that are associated with breast cancer development, overlap substantially with a group of ECM proteins induced during the state of tissue remodeling such as mammary gland involution. Fibrillar collagens, fibronectin, hyaluronan and matricellular proteins are matrix components that are common to both involution and cancer. Moreover, some of these proteins have in recent years been identified as important constituents of metastatic niches in breast cancer. In addition, specific ECM molecules, their receptors or enzymatic modifiers are significantly involved in resistance to therapeutic intervention. Further analysis of these ECM proteins and the downstream ECM mediated signaling pathways may provide a range of possibilities to identify druggable targets against advanced breast cancer.
Collapse
Affiliation(s)
- Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; Divison of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
133
|
Molecular Cloning and Characterization of Cystatin, a Cysteine Protease Inhibitor, fromBufo melanostictus. Biosci Biotechnol Biochem 2014; 77:2077-81. [DOI: 10.1271/bbb.130424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
134
|
Li Y, Lu J, Peng Z, Tan G, Liu N, Huang D, Zhang Z, Duan C, Tang X, Tang F. N,N'-dinitrosopiperazine-mediated AGR2 is involved in metastasis of nasopharyngeal carcinoma. PLoS One 2014; 9:e92081. [PMID: 24717913 PMCID: PMC3981702 DOI: 10.1371/journal.pone.0092081] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/19/2014] [Indexed: 12/15/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has a high metastatic character in the clinic, but its mechanism is not clear. As a carcinogen with organ specificity for the nasopharyngeal epithelium, N,N'-Dinitrosopiperazine (DNP) is involved in NPC metastasis. Herein, our data revealed that anterior gradient 2 (AGR2) was overexpressed in human NPC tissues, particularly in cervical lymph node metastatic NPC (LMNPC). High AGR2 expression was associated with NPC metastasis. Importantly, DNP induced AGR2 expression, and increased cell motility and invasion in the NPC cell line 6-10B. However, DNP-mediated cell motility and invasion was dramatically decreased when transfected with siRNA-AGR2. Further, AGR2 directly regulated cathepsin (CTS) B and D by binding them in vitro. These results indicate that DNP induces AGR2 expression, regulates CTSB and CTSD, increases cell motility and invasion, and promotes NPC tumor metastasis. Therefore, DNP-mediated AGR2 expression may be an important factor in prolific NPC metastasis.
Collapse
Affiliation(s)
- Yuejin Li
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Jinping Lu
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Zhengke Peng
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Gongjun Tan
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Na Liu
- Medical Research Center and Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Damao Huang
- Medical Research Center and Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhenlin Zhang
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Chaojun Duan
- Medical Research Center and Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiaowei Tang
- Metallurgical Science and Engineering, Central South University, Changsha, People’s Republic of China
| | - Faqing Tang
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People’s Hospital, Zhuhai, Guangdong, People’s Republic of China
- * E-mail:
| |
Collapse
|
135
|
Kim CJ, Lee DI, Kim C, Lee K, Lee CH, Ahn IS. Gold Nanoparticles-Based Colorimetric Assay for Cathepsin B Activity and the Efficiency of Its Inhibitors. Anal Chem 2014; 86:3825-33. [DOI: 10.1021/ac4039064] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Chan-Jin Kim
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Dong-Ik Lee
- Dityrosine Innovation Chemical (D. I. Chemical), Seoul 120-749, South Korea
| | - Cheonghee Kim
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Kangtaek Lee
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Chang-Ha Lee
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Ik-Sung Ahn
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| |
Collapse
|
136
|
Yang M, Liu J, Shao J, Qin Y, Ji Q, Zhang X, Du J. Cathepsin S-mediated autophagic flux in tumor-associated macrophages accelerate tumor development by promoting M2 polarization. Mol Cancer 2014; 13:43. [PMID: 24580730 PMCID: PMC4015740 DOI: 10.1186/1476-4598-13-43] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/19/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating leukocytes. TAMs are heterogeneous, with distinct phenotypes influenced by the microenvironment surrounding tumor tissues, but relatively little is known about the key molecular in these cells that contribute to malignant phenotypes. Autophagic activity is a critical factor in tumor development that contributes to enhancing cellular fitness and survival in the hostile tumor microenvironment. However, the molecular basis and relations between autophagy and TAMs polarization remain unclear. METHODS Cathepsin S (Cat S) expression was analyzed in human colon carcinoma and normal colon tissues. In vivo effects were evaluated using PancO2 subcutaneous tumor model and SL4 hepatic metastasis model. Immunofluorescence staining, flow cytometry and real-time PCR were done to examine TAMs polarization. Western blotting assay, transmission electron microscopy, mCherry-GFP-LC3 transfection and DQ-BSA degradation assays were carried out to determine its role in regulating autophagy. RESULTS In the present study, we showed that the enhanced expression of Cat S correlated with the severity of histologic grade as well as clinical stage, metastasis, and recurrence, which are known indicators of a relatively poor prognosis of human colon carcinoma. Cat S knockout led to decreased tumor growth and metastasis. Moreover, Cat S knockout inhibited M2 macrophage polarization during tumor development. We further demonstrated that Cat S was required for not only autophagic flux but also the fusion processes of autophagosomes and lysosomes in TAMs. Importantly, we found that Cat S contributed to tumor development by regulating the M2 phenotype of TAMs through the activation of autophagy. CONCLUSIONS These results indicated that Cat S-mediated autophagic flux is an important mechanism for inducing M2-type polarization of TAMs, which leads to tumor development. These data provide strong evidence for a tumor-promoting role of autophagy in TAMs and suggest Cat S could be a potential target for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
137
|
Lao Y, Wan G, Liu Z, Wang X, Ruan P, Xu W, Xu D, Xie W, Zhang Y, Xu H, Xu N. The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 2014; 10:736-49. [PMID: 24642486 PMCID: PMC5119057 DOI: 10.4161/auto.28034] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metabolic stress induces autophagy as an alternative source of energy and metabolites. Insufficient autophagy in nutrient-deprived cancer cells would be beneficial for cancer therapy. Here, we performed a functional screen in search of novel autophagy regulators from natural products. We showed that oblongifolin C (OC), a natural small molecule compound extracted from Garcinia yunnanensis Hu, is a potent autophagic flux inhibitor. Exposure to OC results in an increased number of autophagosomes and impaired degradation of SQSTM1/p62. Costaining of GFP-LC3B with LysoTracker Red or LAMP1 antibody demonstrates that autophagosome-lysosome fusion is blocked by OC treatment. Furthermore, OC inhibits lysosomal proteolytic activity by altering lysosomal acidification and downregulating the expression of lysosomal cathepsins. Importantly, OC can eliminate the tolerance of cancer cells to nutrient starvation. Starvation dramatically increases the susceptibility of cancer cells to OC-induced CASP3-dependent apoptosis in vitro. Subsequent studies in xenograft mouse model showed that OC has anticancer potency as revealed by increased staining of cleaved CASP3, LC3 puncta, and SQSTM1, as well as reduced expression of lysosomal cathepsins. Combined treatment with OC and caloric restriction potentiates anticancer efficacy of OC in vivo. Collectively, these data demonstrated that OC is a novel autophagic flux inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Yuanzhi Lao
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Gang Wan
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Zhenyan Liu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Xiaoyu Wang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Ping Ruan
- Department of Pathology; Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine; Nanning, China
| | - Wei Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Danqing Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China; Department of Discovery Technologies; Roche R&D Center (China) Ltd; Shanghai, China
| | - Weidong Xie
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Hongxi Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| |
Collapse
|
138
|
[Pathophysiology of bone metastases and new molecular targets involved in bone remodelling]. Bull Cancer 2014; 100:1083-91. [PMID: 24152978 DOI: 10.1684/bdc.2013.1836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bone metastases are common complications of cancers. These skeletal lesions are usually osteolytic (excess of bone destruction), osteosclerostic (excess of bone formation) or mixed. Metastatic cancer cells residing in the bone marrow alter the functions of bone-resorbing (osteoclasts) and bone-forming (osteoblasts) cells and hijack signals coming from the bone matrix. In this review, we first described cellular and molecular mechanisms that drive cancer cells to colonize the bone marrow. We next show how cancer cells alter bone remodelling to promote the formation of osteolytic or osteosclerotic lesions.
Collapse
|
139
|
Sarg MTM, El-Shaer SS. Efficient Utilization of 6-Aminouracil to Synthesize Fused and Related Heterocyclic Compounds and Their Evaluation as Prostate Cytotoxic Agents with Cathepsin B Inhibition. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/ojmc.2014.42003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
140
|
Ryu JH, Na JH, Ko HK, You DG, Park S, Jun E, Yeom HJ, Seo DH, Park JH, Jeong SY, Kim IS, Kim BS, Kwon IC, Choi K, Kim K. Non-invasive optical imaging of cathepsin B with activatable fluorogenic nanoprobes in various metastatic models. Biomaterials 2013; 35:2302-11. [PMID: 24360720 DOI: 10.1016/j.biomaterials.2013.11.080] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 11/26/2013] [Indexed: 02/07/2023]
Abstract
An increasing number of treatments of metastases rely on diagnostics and imaging these days. The facts that the activity of cathepsin B (CB) is markedly linked to the metastatic process and that CB is found highly expressed in the pericellular regions in this process make CB an attractive target for diagnosing metastases. We have developed a CB-sensitive nanoprobe (CB-CNP) consisting of self-quenched CB-sensitive fluorogenic peptide probes conjugated onto the surface of tumor-targeting glycol chitosan nanoparticles (CNPs). The freshly prepared CB-CNP formed a spherical nanoparticle structure (280 nm in diameter) and the fluorescence intensity of CB-CNP was strongly quenched in physiological condition. However, self-quenched CB-CNP boosted strong fluorescence signals in the presence of CB, not of cathepsin l or cathepsin d, due to the CB-specific cleavage of self-quenched peptide probes. Importantly, the intravenously injected CB-CNP demonstrated the potential to discriminate metastases in vivo in three metastatic mouse models, including 4T1-luc2 liver metastases, RFP-B16F10 lung metastases and HT1080 peritoneal metastases. Indeed, Western blot analysis confirmed that the CB expression of metastases had increased compared to normal organ in these metastatic mouse models. CB-CNPs may be useful for depicting metastases through non-invasive CB molecular imaging.
Collapse
Affiliation(s)
- Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, San 56-1, Sillim-dong, Gwanak-gu, Seoul 151-744, Republic of Korea
| | - Jin Hee Na
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Ho Kyung Ko
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Dong Gil You
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Departments of Polymer Science and Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Subin Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Departments of Polymer Science and Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Eunsung Jun
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Ho Jun Yeom
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Deok Ho Seo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Jae Hyung Park
- Departments of Polymer Science and Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Seo Young Jeong
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, San 56-1, Sillim-dong, Gwanak-gu, Seoul 151-744, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea; KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, Republic of Korea
| | - Kuiwon Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea.
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, Republic of Korea.
| |
Collapse
|
141
|
Kubisch R, Fröhlich T, Arnold GJ, Schreiner L, von Schwarzenberg K, Roidl A, Vollmar AM, Wagner E. V-ATPase inhibition by archazolid leads to lysosomal dysfunction resulting in impaired cathepsin B activation in vivo. Int J Cancer 2013; 134:2478-88. [PMID: 24166050 DOI: 10.1002/ijc.28562] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/02/2013] [Accepted: 10/08/2013] [Indexed: 01/08/2023]
Abstract
The myxobacterial agent archazolid inhibits the vacuolar proton pump V-ATPase. V-ATPases are ubiquitously expressed ATP-dependent proton pumps, which are known to regulate the pH in endomembrane systems and thus play a crucial role in endo- and exocytotic processes of the cell. As cancer cells depend on a highly active secretion of proteolytic proteins in order to invade tissue and form metastases, inhibition of V-ATPase is proposed to affect the secretion profile of cancer cells and thus potentially abrogate their metastatic properties. Archazolid is a novel V-ATPase inhibitor. Here, we show that the secretion pattern of archazolid treated cancer cells includes various prometastatic lysosomal proteins like cathepsin A, B, C, D and Z. In particular, archazolid induced the secretion of the proforms of cathepsin B and D. Archazolid treatment abrogates the cathepsin B maturation process leading to reduced intracellular mature cathepsin B protein abundance and finally decreased cathepsin B activity, by inhibiting mannose-6-phoshate receptor-dependent trafficking. Importantly, in vivo reduced cathepsin B protein as well as a decreased proteolytic cathepsin B activity was detected in tumor tissue of archazolid-treated mice. Our results show that inhibition of V-ATPase by archazolid reduces the activity of prometastatic proteases like cathepsin B in vitro and in vivo.
Collapse
Affiliation(s)
- Rebekka Kubisch
- Pharmaceutical Biotechnology Department of Pharmacy, Ludwig Maximilians University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Pantano F, Zoccoli A, Iuliani M, Fioramonti M, Lanzetta G, Tonini G, Santini D. Targeting Bone Metastases: New Drugs for New Targets. Clin Rev Bone Miner Metab 2013. [DOI: 10.1007/s12018-013-9150-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
143
|
Esposito M, Kang Y. Targeting tumor-stromal interactions in bone metastasis. Pharmacol Ther 2013; 141:222-33. [PMID: 24140083 DOI: 10.1016/j.pharmthera.2013.10.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 12/17/2022]
Abstract
Bone metastasis is a frequent occurrence in late stage solid tumors, including breast cancers, prostate or lung. However, the causes for this proclivity have only recently been elucidated. Significant progress has been made in the past decade toward understanding the molecular underpinnings of bone metastasis, and much of this research reveals a crucial role of the host stroma in each step of the metastatic cascade. Tumor-stromal interactions are crucial in engineering a pre-metastatic niche, accommodating metastatic seeding, and establishing the vicious cycle of bone metastasis. Current treatments in bone metastasis focus on latter steps of the metastatic cascade, with most treatments targeting the process of bone remodeling; however, emerging research identifies many other candidates as promising targets. Host stromal cells including platelets and endothelial cells are important in the early steps of metastatic homing, attachment and extravasation while a variety of immune cells, parenchymal cells and mesenchymal cells of the bone marrow are important in the establishment of overt, immune-suppressed metastatic lesions. Many participants during these steps have been identified and functionally validated. Significant contributors include integrins, (αvβ3, α2β1, α4β1), TGFβ family members, bone resident proteins (BSP, OPG, SPARC, OPN), RANKL, and PTHrP. In this review, we will discuss the contribution of host stromal cells to pre-metastatic niche conditioning, seeding, dormancy, bone-remodeling, immune regulation, and chemotherapeutic shielding in bone metastasis. Research exploring these interactions between bone metastases and stromal cells has yielded many therapeutic targets, and we will discuss both the current and future therapeutic avenues in treating bone metastasis.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States.
| |
Collapse
|
144
|
Conformational mobility of active and E-64-inhibited actinidin. Biochim Biophys Acta Gen Subj 2013; 1830:4790-9. [DOI: 10.1016/j.bbagen.2013.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 06/12/2013] [Accepted: 06/13/2013] [Indexed: 11/22/2022]
|
145
|
Cell type-dependent pathogenic functions of overexpressed human cathepsin B in murine breast cancer progression. Oncogene 2013; 33:4474-84. [PMID: 24077280 DOI: 10.1038/onc.2013.395] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 07/03/2013] [Accepted: 08/19/2013] [Indexed: 12/13/2022]
Abstract
The cysteine protease cathepsin B (CTSB) is frequently overexpressed in human breast cancer and correlated with a poor prognosis. Genetic deficiency or pharmacological inhibition of CTSB attenuates tumor growth, invasion and metastasis in mouse models of human cancers. CTSB is expressed in both cancer cells and cells of the tumor stroma, in particular in tumor-associated macrophages (TAM). In order to evaluate the impact of tumor- or stromal cell-derived CTSB on Polyoma Middle T (PyMT)-induced breast cancer progression, we used in vivo and in vitro approaches to induce human CTSB overexpression in PyMT cancer cells or stromal cells alone or in combination. Orthotopic transplantation experiments revealed that CTSB overexpression in cancer cells rather than in the stroma affects PyMT tumor progression. In 3D cultures, primary PyMT tumor cells showed higher extracellular matrix proteolysis and enhanced collective cell invasion when CTSB was overexpressed and proteolytically active. Coculture of PyMT cells with bone marrow-derived macrophages induced a TAM-like macrophage phenotype in vitro, and the presence of such M2-polarized macrophages in 3D cultures enhanced sprouting of tumor spheroids. We employed a doxycycline (DOX)-inducible CTSB expression system to selectively overexpress human CTSB either in cancer cells or in macrophages in 3D cocultures. Tumor spheroid invasiveness was only enhanced when CTSB was overexpressed in cancer cells, whereas CTSB expression in macrophages alone did not further promote invasiveness of tumor spheroids. We conclude that CTSB overexpression in the PyMT mouse model promotes tumor progression not by a stromal effect, but by a direct, cancer cell-inherent mode of action: CTSB overexpression renders the PyMT cancers more invasive by increasing proteolytic extracellular matrix protein degradation fostering collective cell invasion into adjacent tissue.
Collapse
|
146
|
Song SJ, Poliseno L, Song MS, Ala U, Webster K, Ng C, Beringer G, Brikbak NJ, Yuan X, Cantley LC, Richardson AL, Pandolfi PP. MicroRNA-antagonism regulates breast cancer stemness and metastasis via TET-family-dependent chromatin remodeling. Cell 2013; 154:311-324. [PMID: 23830207 DOI: 10.1016/j.cell.2013.06.026] [Citation(s) in RCA: 368] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 12/21/2012] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
Tumor cells metastasize to distant organs through genetic and epigenetic alterations, including changes in microRNA (miR) expression. Here we find miR-22 triggers epithelial-mesenchymal transition (EMT), enhances invasiveness and promotes metastasis in mouse xenografts. In a conditional mammary gland-specific transgenic (TG) mouse model, we show that miR-22 enhances mammary gland side-branching, expands the stem cell compartment, and promotes tumor development. Critically, miR-22 promotes aggressive metastatic disease in MMTV-miR-22 TG mice, as well as compound MMTV-neu or -PyVT-miR-22 TG mice. We demonstrate that miR-22 exerts its metastatic potential by silencing antimetastatic miR-200 through direct targeting of the TET (Ten eleven translocation) family of methylcytosine dioxygenases, thereby inhibiting demethylation of the mir-200 promoter. Finally, we show that miR-22 overexpression correlates with poor clinical outcomes and silencing of the TET-miR-200 axis in patients. Taken together, our findings implicate miR-22 as a crucial epigenetic modifier and promoter of EMT and breast cancer stemness toward metastasis.
Collapse
Affiliation(s)
- Su Jung Song
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Laura Poliseno
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Min Sup Song
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Ugo Ala
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Kaitlyn Webster
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Christopher Ng
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Gary Beringer
- Department of Systems Biology, Department of Medicine, Division of Signal Transduction, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Nicolai J Brikbak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Xin Yuan
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Lewis C Cantley
- Department of Systems Biology, Department of Medicine, Division of Signal Transduction, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Andrea L Richardson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Pier Paolo Pandolfi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
147
|
Löser R, Bergmann R, Frizler M, Mosch B, Dombrowski L, Kuchar M, Steinbach J, Gütschow M, Pietzsch J. Synthesis and radiopharmacological characterisation of a fluorine-18-labelled azadipeptide nitrile as a potential PET tracer for in vivo imaging of cysteine cathepsins. ChemMedChem 2013; 8:1330-44. [PMID: 23785011 DOI: 10.1002/cmdc.201300135] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Indexed: 12/26/2022]
Abstract
A fluorinated cathepsin inhibitor based on the azadipeptide nitrile chemotype was prepared and selected for positron emission tomography (PET) tracer development owing to its high affinity for the oncologically relevant cathepsins L, S, K and B. Labelling with fluorine-18 was accomplished in an efficient and reliable two-step, one-pot radiosynthesis by using 2-[(18) F]fluoroethylnosylate as a prosthetic agent. The pharmacokinetic properties of the resulting radiotracer compound were studied in vitro, ex vivo and in vivo in normal rats by radiometabolite analysis and small-animal positron emission tomography. These investigations revealed rapid conjugate formation of the tracer with glutathione in the blood, which is associated with slow blood clearance. The potential of the developed (18) F-labelled probe to image tumour-associated cathepsin activity was investigated by dynamic small-animal PET imaging in nude mice bearing tumours derived from the human NCI-H292 lung carcinoma cell line. Computational analysis of the obtained image data indicated the time-dependent accumulation of the radiotracer in the tumours. The expression of the target enzymes in the tumours was confirmed by immunohistochemistry with specific antibodies. This indicates that azadipeptide nitriles have the potential to target thiol-dependent cathepsins in vivo despite their disadvantageous pharmacokinetics.
Collapse
Affiliation(s)
- Reik Löser
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Kallunki T, Olsen OD, Jäättelä M. Cancer-associated lysosomal changes: friends or foes? Oncogene 2013; 32:1995-2004. [PMID: 22777359 DOI: 10.1038/onc.2012.292] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 06/01/2012] [Indexed: 12/28/2022]
Abstract
Rapidly dividing and invasive cancer cells are strongly dependent on effective lysosomal function. Accordingly, transformation and cancer progression are characterized by dramatic changes in lysosomal volume, composition and cellular distribution. Depending on one's point of view, the cancer-associated changes in the lysosomal compartment can be regarded as friends or foes. Most of them are clearly transforming as they promote invasive growth, angiogenesis and drug resistance. The same changes can, however, strongly sensitize cells to lysosomal membrane permeabilization and thereby to lysosome-targeting anti-cancer drugs. In this review we compile our current knowledge on cancer-associated changes in lysosomal composition and discuss the consequences of these alterations to cancer progression and the possibilities they can bring to cancer therapy.
Collapse
Affiliation(s)
- T Kallunki
- Cell Death and Metabolism and Centre for Genotoxic Stress Research, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | | |
Collapse
|
149
|
4-tert-Octylphenol stimulates the expression of cathepsins in human breast cancer cells and xenografted breast tumors of a mouse model via an estrogen receptor-mediated signaling pathway. Toxicology 2013; 304:13-20. [DOI: 10.1016/j.tox.2012.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/06/2012] [Accepted: 10/09/2012] [Indexed: 01/13/2023]
|
150
|
Valkenburg KC, Steensma MR, Williams BO, Zhong Z. Skeletal metastasis: treatments, mouse models, and the Wnt signaling. CHINESE JOURNAL OF CANCER 2013; 32:380-96. [PMID: 23327798 PMCID: PMC3845601 DOI: 10.5732/cjc.012.10218] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Skeletal metastases result in significant morbidity and mortality. This is particularly true of cancers with a strong predilection for the bone, such as breast, prostate, and lung cancers. There is currently no reliable cure for skeletal metastasis, and palliative therapy options are limited. The Wnt signaling pathway has been found to play an integral role in the process of skeletal metastasis and may be an important clinical target. Several experimental models of skeletal metastasis have been used to find new biomarkers and test new treatments. In this review, we discuss pathologic process of bone metastasis, the roles of the Wnt signaling, and the available experimental models and treatments.
Collapse
Affiliation(s)
- Kenneth C Valkenburg
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | | | | |
Collapse
|