851
|
Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nat Commun 2015; 6:8752. [PMID: 26658462 PMCID: PMC4682176 DOI: 10.1038/ncomms9752] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/22/2015] [Indexed: 12/18/2022] Open
Abstract
Early cancers are avascular and hence, profoundly acidic. Pre-malignant cells must adapt to acidosis to thrive in this hostile microenvironment. Here, we investigate MCF-7 cells that are adapted to grow in acidic conditions using SILAC proteomics and we reveal a significant upregulation of lysosomal proteins. Prominent among these is LAMP2 that functions to protect lysosomal membranes from acid proteolysis. LAMP2 upregulation by acidosis is confirmed both in vitro and in vivo. Furthermore, we show that the depletion of LAMP2 is sufficient to increase acidosis-mediated toxicity. In breast cancer patient samples, there is a high correlation of LAMP2 mRNA and protein expression with progression. We also observe that LAMP2 is located at the plasma membrane in clinical samples and this redistribution is acid-induced in vitro. Our findings suggest a potential adaptive mechanism, wherein cells chronically exposed to an acidic environment translocate lysosomal proteins to their surface, thus protecting the plasmalemma from acid-induced hydrolysis. The protein LAMP2 functions to protect lysosomal membranes from acid proteolysis. In this study, the authors show that malignant cells adapt to the acidic tumour microenvironment by redirecting LAMP2 from lysosomes to the plasma membrane.
Collapse
|
852
|
Wang Z, Dabrosin C, Yin X, Fuster MM, Arreola A, Rathmell WK, Generali D, Nagaraju GP, El-Rayes B, Ribatti D, Chen YC, Honoki K, Fujii H, Georgakilas AG, Nowsheen S, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich B, Yang X, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Halicka D, Mohammed SI, Azmi AS, Bilsland A, Keith WN, Jensen LD. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 2015; 35 Suppl:S224-S243. [PMID: 25600295 PMCID: PMC4737670 DOI: 10.1016/j.semcancer.2015.01.001] [Citation(s) in RCA: 336] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 12/25/2014] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
Deregulation of angiogenesis--the growth of new blood vessels from an existing vasculature--is a main driving force in many severe human diseases including cancer. As such, tumor angiogenesis is important for delivering oxygen and nutrients to growing tumors, and therefore considered an essential pathologic feature of cancer, while also playing a key role in enabling other aspects of tumor pathology such as metabolic deregulation and tumor dissemination/metastasis. Recently, inhibition of tumor angiogenesis has become a clinical anti-cancer strategy in line with chemotherapy, radiotherapy and surgery, which underscore the critical importance of the angiogenic switch during early tumor development. Unfortunately the clinically approved anti-angiogenic drugs in use today are only effective in a subset of the patients, and many who initially respond develop resistance over time. Also, some of the anti-angiogenic drugs are toxic and it would be of great importance to identify alternative compounds, which could overcome these drawbacks and limitations of the currently available therapy. Finding "the most important target" may, however, prove a very challenging approach as the tumor environment is highly diverse, consisting of many different cell types, all of which may contribute to tumor angiogenesis. Furthermore, the tumor cells themselves are genetically unstable, leading to a progressive increase in the number of different angiogenic factors produced as the cancer progresses to advanced stages. As an alternative approach to targeted therapy, options to broadly interfere with angiogenic signals by a mixture of non-toxic natural compound with pleiotropic actions were viewed by this team as an opportunity to develop a complementary anti-angiogenesis treatment option. As a part of the "Halifax Project" within the "Getting to know cancer" framework, we have here, based on a thorough review of the literature, identified 10 important aspects of tumor angiogenesis and the pathological tumor vasculature which would be well suited as targets for anti-angiogenic therapy: (1) endothelial cell migration/tip cell formation, (2) structural abnormalities of tumor vessels, (3) hypoxia, (4) lymphangiogenesis, (5) elevated interstitial fluid pressure, (6) poor perfusion, (7) disrupted circadian rhythms, (8) tumor promoting inflammation, (9) tumor promoting fibroblasts and (10) tumor cell metabolism/acidosis. Following this analysis, we scrutinized the available literature on broadly acting anti-angiogenic natural products, with a focus on finding qualitative information on phytochemicals which could inhibit these targets and came up with 10 prototypical phytochemical compounds: (1) oleanolic acid, (2) tripterine, (3) silibinin, (4) curcumin, (5) epigallocatechin-gallate, (6) kaempferol, (7) melatonin, (8) enterolactone, (9) withaferin A and (10) resveratrol. We suggest that these plant-derived compounds could be combined to constitute a broader acting and more effective inhibitory cocktail at doses that would not be likely to cause excessive toxicity. All the targets and phytochemical approaches were further cross-validated against their effects on other essential tumorigenic pathways (based on the "hallmarks" of cancer) in order to discover possible synergies or potentially harmful interactions, and were found to generally also have positive involvement in/effects on these other aspects of tumor biology. The aim is that this discussion could lead to the selection of combinations of such anti-angiogenic compounds which could be used in potent anti-tumor cocktails, for enhanced therapeutic efficacy, reduced toxicity and circumvention of single-agent anti-angiogenic resistance, as well as for possible use in primary or secondary cancer prevention strategies.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Daniele Generali
- Molecular Therapy and Pharmacogenomics Unit, AO Isituti Ospitalieri di Cremona, Cremona, Italy
| | - Ganji P Nagaraju
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, USA
| | - Kanya Honoki
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirate University, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirate University, United Arab Emirates
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guilford, Surrey, UK
| | | | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Asfar S Azmi
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lasse D Jensen
- Department of Medical, and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
853
|
Amith SR, Wilkinson JM, Baksh S, Fliegel L. The Na⁺/H⁺ exchanger (NHE1) as a novel co-adjuvant target in paclitaxel therapy of triple-negative breast cancer cells. Oncotarget 2015; 6:1262-75. [PMID: 25514463 PMCID: PMC4359231 DOI: 10.18632/oncotarget.2860] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/25/2014] [Indexed: 02/04/2023] Open
Abstract
Dysregulation of Na+ /H+ exchanger isoform one (NHE1) activity is a hallmark of cells undergoing tumorigenesis and metastasis, the leading cause of patient mortality. The acidic tumor microenvironment is thought to facilitate the development of resistance to chemotherapy drugs and to promote extracellular matrix remodeling leading to metastasis. Here, we investigated NHE1 as a co-adjuvant target in paclitaxel chemotherapy of metastatic breast cancer. We generated a stable NHE1-knockout of the highly invasive, triple-negative, MDA-MB-231 breast cancer cells. The NHE1-knockout cells proliferated comparably to parental cells, but had markedly lower rates of migration and invasion in vitro. In vivo xenograft tumor growth in athymic nude mice was also dramatically decreased compared to parental MDA-MB-231 cells. Loss of NHE1 expression also increased the susceptibility of knockout cells to paclitaxel-mediated cell death. NHE1 inhibition, in combination with paclitaxel, resulted in a dramatic decrease in viability, and migratory and invasive potential of triple-negative breast cancer cells, but not in hormone receptor-positive, luminal MCF7 cells. Our data suggest that NHE1 is critical in triple-negative breast cancer metastasis, and its chemical inhibition boosts the efficacy of paclitaxel in vitro, highlighting NHE1 as a novel, potential co-adjuvant target in breast cancer chemotherapy.
Collapse
Affiliation(s)
- Schammim Ray Amith
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Shairaz Baksh
- Department of Pediatrics, Biochemistry and Oncology, Alberta Inflammatory Bowel Disease Consortium, University of Alberta, Edmonton, Alberta, Canada
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
854
|
Chu CY, Jin YT, Zhang W, Yu J, Yang HP, Wang HY, Zhang ZJ, Liu XP, Zou Q. CA IX is upregulated in CoCl2-induced hypoxia and associated with cell invasive potential and a poor prognosis of breast cancer. Int J Oncol 2015; 48:271-80. [PMID: 26648580 DOI: 10.3892/ijo.2015.3253] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022] Open
Abstract
Hypoxia, a common phenomenon during the development of malignant solid tumors including breast cancer, serves to propagate a cascade of molecular pathways triggered by hypoxia-inducible factor-1α (HIF-1α). Carbonic anhydrase IX (CA IX), one of the target genes of HIF-1α, has been reported to be involved in progression of malignant tumors. The objective of this study was to investigate the expression of HIF-1α and CA IX in hypoxia, involvement of CA IX in the regulation of migration and invasion/metastasis and its prognostic significance in breast cancer. We used cobalt chloride (CoCl2) as a hypoxia-mimetic agent and found that the expression of HIF-1α protein, CA IX mRNA and protein, is effectively upregulated, except for HIF-1α mRNA. Data showed that the elevated CA IX expression is closely related to in vitro cell migration and invasion, and the underlying mechanism of this process may be associated with epithelial-mesenchymal transition (EMT). The study of clinical tissue samples also demonstrated that CA IX is an independent prognostic marker that may serve as a useful clinical biomarker for predicting tumor progression and the invasion/metastasis of breast cancer. These results provide further insight into the role of CA IX in tumor progression and put forward further strong evidence as well as new consideration for CA IX target therapy.
Collapse
Affiliation(s)
- Cheng-Yu Chu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yi-Ting Jin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Wei Zhang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Juan Yu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Hai-Ping Yang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Hong-Ying Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zi-Jing Zhang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiu-Ping Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Qiang Zou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
855
|
Polyphenols as Modulator of Oxidative Stress in Cancer Disease: New Therapeutic Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6475624. [PMID: 26649142 PMCID: PMC4663347 DOI: 10.1155/2016/6475624] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/21/2015] [Indexed: 12/19/2022]
Abstract
Cancer onset and progression have been linked to oxidative stress by increasing DNA mutations or inducing DNA damage, genome instability, and cell proliferation and therefore antioxidant agents could interfere with carcinogenesis. It is well known that conventional radio-/chemotherapies influence tumour outcome through ROS modulation. Since these antitumour treatments have important side effects, the challenge is to develop new anticancer therapeutic strategies more effective and less toxic for patients. To this purpose, many natural polyphenols have emerged as very promising anticancer bioactive compounds. Beside their well-known antioxidant activities, several polyphenols target epigenetic processes involved in cancer development through the modulation of oxidative stress. An alternative strategy to the cytotoxic treatment is an approach leading to cytostasis through the induction of therapy-induced senescence. Many anticancer polyphenols cause cellular growth arrest through the induction of a ROS-dependent premature senescence and are considered promising antitumour therapeutic tools. Furthermore, one of the most innovative and interesting topics is the evaluation of efficacy of prooxidant therapies on cancer stem cells (CSCs). Several ROS inducers-polyphenols can impact CSCs metabolisms and self-renewal related pathways. Natural polyphenol roles, mainly in chemoprevention and cancer therapies, are described and discussed in the light of the current literature data.
Collapse
|
856
|
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
|
857
|
Metabolic exchanges within tumor microenvironment. Cancer Lett 2015; 380:272-80. [PMID: 26546872 DOI: 10.1016/j.canlet.2015.10.027] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 12/30/2022]
Abstract
Tumor progression toward malignancy often requires a metabolic rewiring of cancer cells to meet changes in metabolic demand to forefront nutrient and oxygen withdrawal, together with strong anabolic requests to match high proliferation rate. Tumor microenvironment highly contributes to metabolic rewiring of cancer cells, fostering complete nutrient exploitation, favoring OXPHOS of lipids and glutamine at the expense of glycolysis and enhancing exchanges via extracellular microvesicles or exosomes of proteins, lipids and small RNAs among tumor and stromal cells. Noteworthy, the same molecular drivers of metabolic reprogramming within tumor and stroma are also able to elicit motility, survival and self-renewal on cancer cells, thereby sustaining successful escaping strategies to circumvent the hostile hypoxic, acidic and inflammatory environment. This review highlights the emerging role of nutrients and vesicle-mediated exchanges among tumor and stromal cells, defining their molecular pathways and offering new perspectives to develop treatments targeting this complex metabolic rewiring.
Collapse
|
858
|
Gilson RC, Tang R, Som A, Klajer C, Sarder P, Sudlow GP, Akers WJ, Achilefu S. Protonation and Trapping of a Small pH-Sensitive Near-Infrared Fluorescent Molecule in the Acidic Tumor Environment Delineate Diverse Tumors in Vivo. Mol Pharm 2015; 12:4237-46. [PMID: 26488921 DOI: 10.1021/acs.molpharmaceut.5b00430] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enhanced glycolysis and poor perfusion in most solid malignant tumors create an acidic extracellular environment, which enhances tumor growth, invasion, and metastasis. Complex molecular systems have been explored for imaging and treating these tumors. Here, we report the development of a small molecule, LS662, that emits near-infrared (NIR) fluorescence upon protonation by the extracellular acidic pH environment of diverse solid tumors. Protonation of LS662 induces selective internalization into tumor cells and retention in the tumor microenvironment. Noninvasive NIR imaging demonstrates selective retention of the pH sensor in diverse tumors, and two-photon microscopy of ex vivo tumors reveals significant retention of LS662 in tumor cells and the acid tumor microenvironment. Passive and active internalization processes combine to enhance NIR fluorescence in tumors over time. The low background fluorescence allows tumors to be detected with high sensitivity, as well as dead or dying cells to be delineated from healthy cells. In addition to demonstrating the feasibility of using small molecule pH sensors to image multiple aggressive solid tumor types via a protonation-induced internalization and retention pathway, the study reveals the potential of using LS662 to monitor treatment response and tumor-targeted drug delivery.
Collapse
Affiliation(s)
- Rebecca C Gilson
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Rui Tang
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Avik Som
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Chloe Klajer
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Pinaki Sarder
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Gail P Sudlow
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Walter J Akers
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Samuel Achilefu
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| |
Collapse
|
859
|
Nguyen VP, Alves DS, Scott HL, Davis FL, Barrera FN. A Novel Soluble Peptide with pH-Responsive Membrane Insertion. Biochemistry 2015; 54:6567-75. [DOI: 10.1021/acs.biochem.5b00856] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vanessa P. Nguyen
- Department of Biochemistry
and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Daiane S. Alves
- Department of Biochemistry
and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Haden L. Scott
- Department of Biochemistry
and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Forrest L. Davis
- Department of Biochemistry
and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Francisco N. Barrera
- Department of Biochemistry
and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
860
|
Holder AB, Rodrigo MR. Model for acid-mediated tumour invasion with chemotherapy intervention II: Spatially heterogeneous populations. Math Biosci 2015; 270:10-29. [PMID: 26459059 DOI: 10.1016/j.mbs.2015.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 07/15/2015] [Accepted: 09/28/2015] [Indexed: 11/16/2022]
Abstract
This paper extends the model for acid-mediated tumour invasion with chemotherapy intervention examined in part I. The model presented in part I considers the interaction between tumour cells, normal cells, acid and drug in a well mixed (i.e. spatially homogeneous) setting, which is governed by a system of nonlinear differential equations. The model examined here removes the assumption that the populations are spatially homogeneous resulting in a system of nonlinear partial differential equations. Numerical simulations of this model are presented for different treatment methods displaying several possible behaviours. Asymptotic approximations are also derived for a special case of the treatment method and set of parameter values. This analysis then allows us to draw conclusions about the effectiveness of treating acid-mediated tumours with chemotherapy.
Collapse
Affiliation(s)
- Andrew B Holder
- School of Mathematics and Applied Statistics, University of Wollongong, NSW 2522, Australia.
| | - Marianito R Rodrigo
- School of Mathematics and Applied Statistics, University of Wollongong, NSW 2522, Australia.
| |
Collapse
|
861
|
Kimbrough CW, Khanal A, Zeiderman M, Khanal BR, Burton NC, McMasters KM, Vickers SM, Grizzle WE, McNally LR. Targeting Acidity in Pancreatic Adenocarcinoma: Multispectral Optoacoustic Tomography Detects pH-Low Insertion Peptide Probes In Vivo. Clin Cancer Res 2015; 21:4576-85. [PMID: 26124201 PMCID: PMC4609270 DOI: 10.1158/1078-0432.ccr-15-0314] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND pH-low insertion peptides (pHLIP) can serve as a targeting moiety that enables pH-sensitive probes to detect solid tumors. Using these probes in conjunction with multispectral optoacoustic tomography (MSOT) is a promising approach to improve imaging for pancreatic cancer. METHODS A pH-sensitive pHLIP (V7) was conjugated to 750 NIR fluorescent dye and evaluated as a targeted probe for pancreatic adenocarcinoma. The pH-insensitive K7 pHLIP served as an untargeted control. Probe binding was assessed in vitro at pH 7.4, 6.8, and 6.6 using human pancreatic cell lines S2VP10 and S2013. Using MSOT, semiquantitative probe accumulation was then assessed in vivo with a murine orthotopic pancreatic adenocarcinoma model. RESULTS In vitro, the V7-750 probe demonstrated significantly higher fluorescence at pH 6.6 compared with pH 7.4 (S2VP10, P = 0.0119; S2013, P = 0.0160), whereas no difference was observed with the K7-750 control (S2VP10, P = 0.8783; S2013, P = 0.921). In the in vivo S2VP10 model, V7-750 probe resulted in 782.5 MSOT a.u. signal compared with 5.3 MSOT a.u. in K7-750 control in tumor (P = 0.0001). Similarly, V7-750 probe signal was 578.3 MSOT a.u. in the S2013 model compared with K7-750 signal at 5.1 MSOT a.u. (P = 0.0005). There was minimal off-target accumulation of the V7-750 probe within the liver or kidney, and probe distribution was confirmed with ex vivo imaging. CONCLUSIONS Compared with pH-insensitive controls, V7-750 pH-sensitive probe specifically targets pancreatic adenocarcinoma and has minimal off-target accumulation. The noninvasive detection of pH-targeted probes by means of MSOT represents a promising modality to improve the detection and monitoring of pancreatic cancer.
Collapse
Affiliation(s)
- Charles W Kimbrough
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Anil Khanal
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Matthew Zeiderman
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Bigya R Khanal
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | | | - Kelly M McMasters
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | | | | | - Lacey R McNally
- Department of Medicine, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
862
|
Sánchez-Sánchez L, Tapia-Moreno A, Juarez-Moreno K, Patterson DP, Cadena-Nava RD, Douglas T, Vazquez-Duhalt R. Design of a VLP-nanovehicle for CYP450 enzymatic activity delivery. J Nanobiotechnology 2015; 13:66. [PMID: 26452461 PMCID: PMC4599659 DOI: 10.1186/s12951-015-0127-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/24/2015] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The intracellular delivery of enzymes for therapeutic use has a promising future for the treatment of several diseases such as genetic disorders and cancer. Virus-like particles offer an interesting platform for enzymatic delivery to targeted cells because of their great cargo capacity and the enhancement of the biocatalyst stability towards several factors important in the practical application of these nanoparticles. RESULTS We have designed a nano-bioreactor based on the encapsulation of a cytochrome P450 (CYP) inside the capsid derived from the bacteriophage P22. An enhanced peroxigenase, CYPBM3, was selected as a model enzyme because of its potential in enzyme prodrug therapy. A total of 109 enzymes per capsid were encapsulated with a 70 % retention of activity for cytochromes with the correct incorporation of the heme cofactor. Upon encapsulation, the stability of the enzyme towards protease degradation and acidic pH was increased. Cytochrome P450 activity was delivered into Human cervix carcinoma cells via transfecting P22-CYP nanoparticles with lipofectamine. CONCLUSION This work provides a clear demonstration of the potential of biocatalytic virus-like particles as medical relevant enzymatic delivery vehicles for clinical applications.
Collapse
Affiliation(s)
- Lorena Sánchez-Sánchez
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62250, Cuernavaca, Morelos, Mexico.
| | - Alejandro Tapia-Moreno
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| | - Karla Juarez-Moreno
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
- Cátedras CONACyT affiliated to CNyN-UNAM, Ensenada, Mexico.
| | - Dustin P Patterson
- Department of Chemistry and Biochemistry, University of Texas at Tyler, Tyler, 75799, TX, USA.
| | - Ruben D Cadena-Nava
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| | - Trevor Douglas
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| |
Collapse
|
863
|
Reichel D, Rychahou P, Bae Y. Polymer nanoassemblies with solvato- and halo-fluorochromism for drug release monitoring and metastasis imaging. Ther Deliv 2015; 6:1221-37. [PMID: 26446432 PMCID: PMC4977001 DOI: 10.4155/tde.15.59] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Theranostics, an emerging technique that combines therapeutic and diagnostic modalities for various diseases, holds promise to detect cancer in early stages, eradicate metastatic tumors and ultimately reduce cancer mortality. METHODS & RESULTS This study reports unique polymer nanoassemblies that increase fluorescence intensity upon addition of hydrophobic drugs and either increase or decrease fluorescence intensity in acidic environments, depending on nanoparticle core environment properties. Extensive spectroscopic analyses were performed to determine optimal excitation and emission wavelengths, which enabled real time measurement of drugs releasing from the nanoassemblies and ex vivo imaging of acidic liver metastatic tumors from mice. CONCLUSION Polymer nanoassemblies with solvato- and halo-fluorochromic properties are promising platforms to develop novel theranostic tools for the detection and treatment of metastatic tumors.
Collapse
Affiliation(s)
- Derek Reichel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, KY 40536–0596, USA
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, 800 Rose Street, CC140, Lexington, KY 40536, USA
- Department of Surgery, College of Medicine, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, KY 40536–0596, USA
| |
Collapse
|
864
|
Abstract
Nanoparticles represent one of the most widely studied classes of advanced drug delivery platforms in recent years due to a wide range of unique properties and capabilities that can be utilized to improve upon traditional drug administration. Conversely, hydrogel nanoparticles (HNPs) - also called nanogels - represent a unique class of materials that combine the intrinsic advantages of nanotechnology with the inherent capabilities of hydrogels. Responsive hydrogels pose a particularly interesting class of materials that can sense and respond to external stimuli and previous reports of inhalable hydrogel particles have highlighted their potential in pulmonary delivery. Here, we synthesized two different pH-responsive HNPs, designated HNP120 and HNP270, by incorporating functional monomers with a common crosslinker and characterized their physicochemical properties. One of the HNP systems was selected for incorporation into a composite dry powder by spray drying, and the aerodynamic performance of the resulting powder was evaluated. The HNP120s displayed a hydrodynamic diameter of approximately 120 nm in their fully swollen state and a minimal diameter of around 80 nm while the HNP270s were approximately 270 nm and 115 nm, respectively. Electron microscopy confirmed particle size- and morphological uniformity of the HNPs. The HNP120s were spray dried into composite dry powders for inhalation and cascade impaction studies showed good aerosol performance with a mass median aerosol diameter (MMAD) of 4.82 ± 0.37 and a fine particle fraction > 30%. The HNPs released from the spray dried composites retained their responsive behavior thereby illustrating the potential for these materials as intelligent drug delivery systems that combine the advantages of nanotechnology, lung targeting through pulmonary delivery, and stimuli-responsive hydrogels.
Collapse
|
865
|
Keshari KR, Wilson DM, Van Criekinge M, Sriram R, Koelsch BL, Wang ZJ, VanBrocklin HF, Peehl DM, O’Brien T, Sampath D, Carano RAD, Kurhanewicz J. Metabolic response of prostate cancer to nicotinamide phophoribosyltransferase inhibition in a hyperpolarized MR/PET compatible bioreactor. Prostate 2015; 75:1601-9. [PMID: 26177608 PMCID: PMC4537380 DOI: 10.1002/pros.23036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/22/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Metabolic shifts in disease are of great interest for the development of novel therapeutics. In cancer treatment, these therapies exploit the metabolic phenotype associated with oncogenesis and cancer progression. One recent strategy involves the depletion of the cofactors needed to maintain the high rate of glycolysis seen with the Warburg effect. Specifically, blocking nicotinamide adenine dinucleotide (NAD) biosynthesis via nicotinamide phosphoribosyltransferase (NAMPT) inhibition depletes cancer cells of the NAD needed for glycolysis. To characterize this metabolic phenotype in vivo and describe changes in flux with treatment, non-invasive biomarkers are necessary. One such biomarker is hyperpolarized (HP) [1-(13) C] pyruvate, a clinically translatable probe that allows real-time assessment of metabolism. METHODS We therefore developed a cell perfusion system compatible with HP magnetic resonance (MR) and positron emission tomography (PET) to develop translatable biomarkers of response to NAMPT inhibition in reduced volume cell cultures. RESULTS Using this platform, we observed a reduction in pyruvate flux through lactate dehydrogenase with NAMPT inhibition in prostate cancer cells, and showed that both HP lactate and 2-[(18) F] fluoro-2-deoxy-D-glucose (FDG) can be used as biomarkers for treatment response of such targeted agents. Moreover, we observed dynamic flux changes whereby HP pyruvate was re-routed to alanine, providing both positive and negative indicators of treatment response. CONCLUSIONS This study demonstrated the feasibility of a MR/PET compatible bioreactor approach to efficiently explore cell and tissue metabolism, the understanding of which is critical for developing clinically translatable biomarkers of disease states and responses to therapeutics.
Collapse
Affiliation(s)
- Kayvan R. Keshari
- Department of Radiology, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY 10065, USA
- Correspondence and Reprint Request: Kayvan R. Keshari, Ph.D., Assistant Member, Department of Radiology and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, Phone: (646) 888-3631, Fax: (646) 422-0247,
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mark Van Criekinge
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bertram L. Koelsch
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Zhen J. Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Donna M. Peehl
- Department of Urology, Stanford University, Stanford, CA 94305, USA
| | - Tom O’Brien
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Deepak Sampath
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Richard A. D. Carano
- Department of Biomedical Imaging, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
866
|
Gupta SC, Singh R, Pochampally R, Watabe K, Mo YY. Acidosis promotes invasiveness of breast cancer cells through ROS-AKT-NF-κB pathway. Oncotarget 2015; 5:12070-82. [PMID: 25504433 PMCID: PMC4322981 DOI: 10.18632/oncotarget.2514] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/24/2014] [Indexed: 01/24/2023] Open
Abstract
It is well known that acidic microenvironment promotes tumorigenesis, however, the underlying mechanism remains largely unknown. In the present study, we show that acidosis promotes invasiveness of breast cancer cells through a series of signaling events. First, our study indicates that NF-κB is a key factor for acidosis-induced cell invasion. Acidosis activates NF-κB without affecting STAT3 activity; knockdown of NF-κB p65 abrogates the acidosis-induced invasion activity. Next, we show that the activation of NF-κB is mediated through phosphorylation and degradation of IκBα; and phosphorylation and nuclear translocation of p65. Upstream to NF-κB signaling, AKT is activated under acidic conditions. Moreover, acidosis induces generation of reactive oxygen species (ROS) which can be suppressed by ROS scavengers, reversing the acidosis-induced activation of AKT and NF-κB, and invasiveness. As a negative regulator of AKT, PTEN is oxidized and inactivated by the acidosis-induced ROS. Finally, inhibition of NADPH oxidase (NOX) suppresses acidosis-induced ROS production, suggesting involvement of NOX in acidosis-induced signaling cascade. Of considerable interest, acidosis-induced ROS production and activation of AKT and NF-κB can be only detected in cancer cells, but not in non-malignant cells. Together, these results demonstrate a cancer specific acidosis-induced signaling cascade in breast cancer cells, leading to cell invasion.
Collapse
Affiliation(s)
- Subash C Gupta
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS. Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Ramesh Singh
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS. Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS. Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Kounosuke Watabe
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS. Department of Microbiology, University of Mississippi Medical Center, Jackson, MS
| | - Yin-Yuan Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS. Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
867
|
Technical innovation in adjuvant radiotherapy: Evolution and evaluation of new treatments for today and tomorrow. Breast 2015; 24 Suppl 2:S114-9. [PMID: 26429399 DOI: 10.1016/j.breast.2015.07.026] [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: 11/21/2022] Open
Abstract
Recent innovations in breast cancer radiotherapy include intensity modulated radiotherapy, brachytherapy and intraoperative radiotherapy and current trials are seeking to evaluate their value in optimizing local control while maintaining cosmetic effects. Future clinical dividends in local control and survival may come from the identification of molecular signatures of breast cancer radiosensitivity, the development of predictive signatures and identification of immunohistochemical markers of risk of local recurrence. The importance of tumour heterogeneity is being increasingly recognized as an important factor in determining radiotherapy response and an improved understanding of the biology of the tumour microenvironment may identify targets that allow enhanced radiosensitisation or reversal of radioresistance when inhibited. This review describes recent developments in these areas.
Collapse
|
868
|
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
|
869
|
Sriram R, Van Criekinge M, Hansen A, Wang ZJ, Vigneron DB, Wilson DM, Keshari KR, Kurhanewicz J. Real-time measurement of hyperpolarized lactate production and efflux as a biomarker of tumor aggressiveness in an MR compatible 3D cell culture bioreactor. NMR IN BIOMEDICINE 2015; 28:1141-1149. [PMID: 26202449 PMCID: PMC4537810 DOI: 10.1002/nbm.3354] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 05/28/2015] [Accepted: 06/05/2015] [Indexed: 05/19/2023]
Abstract
We have developed a 3D cell/tissue culture bioreactor compatible with hyperpolarized (HP) (13)C MR and interrogated HP [1-(13)C]lactate production and efflux in human renal cell carcinoma (RCC) cells. This platform is capable of resolving intracellular and extracellular HP lactate pools, allowing the kinetic measurement of lactate production and efflux in the context of cancer aggressiveness and response to therapy. HP (13)C MR studies were performed on three immortalized human renal cell lines: HK2, a normal renal proximal tubule cell line from which a majority of RCCs arise, UMRC6, a cell line derived from a localized RCC, and UOK262, an aggressive and metastatic RCC. The intra- (Lacin ) and extracellular (Lacex ) HP lactate signals were robustly resolved in dynamic (13)C spectra of the cell lines due to a very small but reproducible chemical shift difference (0.031 ± 0.0005 ppm). Following HP [1-(13)C]pyruvate delivery, the ratio of HP Lacin /Lacex was significantly lower for UOK262 cells compared with both UMRC6 and HK2 cells due to a significant (p < 0.05) increase in the Lacex pool size. Lacin /Lacex correlated with the MCT4 mRNA expression of the cell lines, and inhibition of MCT4 transport using DIDS resulted in a significant reduction in the HP Lacex pool size. The extension of these studies to living patient-derived RCC tissue slices using HP [1,2-(13)C2]pyruvate demonstrated a similarly split lactate doublet with a high Lacex pool fraction; in contrast, only a single NMR resonance is noted for HP [5-(13)C]glutamate, consistent with intracellular localization. These studies support the importance of lactate efflux as a biomarker of cancer aggressiveness and metastatic potential, and the utility of the MR compatible 3D cell/tissue culture bioreactor to study not only cellular metabolism but also transport. Additionally, this platform offers a sophisticated way to follow therapeutic interventions and screen novel therapies that target lactate export.
Collapse
Affiliation(s)
- Renuka Sriram
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
- Correspondence and Reprint Request: Renuka Sriram, University of California, San Francisco, Byers Hall, Room 201B, 1700 4th Street, MC 2520, San Francisco, CA 94158, Tel: (415) 514-4874, Fax: (415) 514-4714,
| | - Mark Van Criekinge
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Ailin Hansen
- Norwegian University of Science and Technology, Trondheim, Norway
| | - Zhen J. Wang
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Daniel B. Vigneron
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - David M. Wilson
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| | - Kayvan R. Keshari
- Radiology and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - John Kurhanewicz
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
870
|
Kim SE, Wallat JD, Harker EC, Advincula AA, Pokorski JK. Multifunctional and Spatially Controlled Bioconjugation to Melt Coextruded Nanofibers. Polym Chem 2015; 6:5683-5692. [PMID: 26604990 PMCID: PMC4654962 DOI: 10.1039/c5py00282f] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Polymeric fibers have drawn recent interest for uses in biomedical technologies that span drug delivery, regenerative medicine, and wound-healing patches, amongst others. We have recently reported a new class of fibrous biomaterials fabricated using coextrusion and a photochemical modification procedure to introduce functional groups onto the fibers. In this report, we extend our methodology to control surface modification density, describe methods to synthesize multifunctional fibers, and provide methods to spatially control functional group modification. Several different functional fibers are reported for bioconjugation, including propargyl, alkene, alkoxyamine, and ketone modified fibers. The modification scheme allows for control over surface density and provides a handle for downstream functionalization with appropriate bioconjugation chemistries. Through the use of multiple orthogonal chemistries, fiber chemistry could be differentially controlled to append multiple modifications. Spatial control on the fiber surface was also realized, leading to reverse gradients of small molecule dyes. One application is demonstrated for pH-responsive drug delivery of an anti-cancer therapeutics. Finally, the introduction of orthogonal chemical modifications onto these fibers allowed for modification with multiple cell-responsive peptides providing a substrate for osteoblast differentiation.
Collapse
Affiliation(s)
- Si-Eun Kim
- Department of Macromolecular Science & Engineering, Case Western Reserve University, 2100 Adelbert Road, Cleveland, Ohio, 44106
| | - Jaqueline D. Wallat
- Department of Macromolecular Science & Engineering, Case Western Reserve University, 2100 Adelbert Road, Cleveland, Ohio, 44106
| | - Emily C. Harker
- Department of Macromolecular Science & Engineering, Case Western Reserve University, 2100 Adelbert Road, Cleveland, Ohio, 44106
| | - Abigail A. Advincula
- Department of Macromolecular Science & Engineering, Case Western Reserve University, 2100 Adelbert Road, Cleveland, Ohio, 44106
| | - Jonathan K. Pokorski
- Department of Macromolecular Science & Engineering, Case Western Reserve University, 2100 Adelbert Road, Cleveland, Ohio, 44106
| |
Collapse
|
871
|
Wang RA, Lu YY, Fan DM. Reasons for cancer metastasis: A holistic perspective. Mol Clin Oncol 2015; 3:1199-1202. [PMID: 26807220 DOI: 10.3892/mco.2015.623] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 02/26/2015] [Indexed: 12/15/2022] Open
Abstract
Over several years, scientists investigating cancer have focused their efforts on elucidating the mechanisms underlying cancer metastasis, with the aim of finding a way to inhibit this process. These mechanisms, however, only explain the process of cancer metastasis, but do not explain why cancer would metastasize in the first place. Cancer metastasizes due to several factors, namely attack by the immune system, lack of oxygen and necessary nutrients, large amounts of lactic acid produced by glycolysis and increased cell death. Therefore, the majority of the presently available treatments for cancer also bear the potential to induce metastasis. Thus, it is crucial in medical practice to minimize the risk of cancer metastasis during a time when there are no effective means to inhibit this process.
Collapse
Affiliation(s)
- Rui-An Wang
- State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China; Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China; Department of Pathology and Pathophysiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - You-Yong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Dai-Ming Fan
- State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China; Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
872
|
Li Y, Xu G, Huang K, Wang J, Zhang J, Liu J, Wang Z, Chen G. Alteration of ASIC1 expression in clear cell renal cell carcinoma. Onco Targets Ther 2015; 8:2121-7. [PMID: 26316781 PMCID: PMC4542551 DOI: 10.2147/ott.s86927] [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] [Indexed: 11/29/2022] Open
Abstract
Background Acidic extracellular pH is a major feature of tumor tissue. Acid-sensing ion channels (ASICs) represent an H+-gated subgroup of the degenerin/epithelial Na+ channel family and are activated by acidic microenvironment. Little is known about the expression and clinical significance of ASICs in solid tumors. The purpose of this study was to examine the expression of ASIC1 in human clear cell renal cell carcinoma (CCRCC) and to determine if the expression of ASIC1 is associated with clinicopathological features. Methods The expression of ASIC1 in CCRCC tissues at the mRNA and protein levels was determined by real-time quantitative polymerase chain reaction and Western blot analysis, respectively. A tissue microarray was used to assess the expression of ASIC1 protein in tumor tissue and matched adjacent normal tissues from 75 patients with CCRCC. Results ASIC1 expression was detected in normal renal and CCRCC samples. The expressions of ASIC1 protein and mRNA were significantly decreased in the CCRCC tissues compared with matched normal renal tissues (P<0.05). The staining density measurement showed that the expression of ASIC1 was significantly decreased in stage I (P=0.037), stage II (P=0.026), and stage III (P=0.026), grades I–II CCRCC (P=0.004), and CCRCC from male patients (P=0.00002). However, no significant difference was observed for ASIC1 expression between CCRCC and normal tissue in patients with stage IV CCRCC (P=0.236), patients with grades III–IV CCRCC (P=0.314), and female patients (P=0.095). Spearman correlations demonstrated that ASIC1 expression did not correlate to tumor stage (correlation coefficient [CC =0.168], P=0.149) and the age of patients (CC −0.147, P=0.688) but showed a positive correlation to higher tumor grades (CC =0.270, P=0.018). Conclusion ASIC1 is downregulated in CCRCC. ASIC1 expression may be potentially used as a novel biomarker and even a CCRCC therapeutic target. Further efforts will be made to clarify the mechanism of ASIC1 in occurrence, progression, and metastasis of CCRCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Guoxiong Xu
- Central Laboratory, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Kai Huang
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Jun Wang
- Department of Urology, Shanghai First People's Hospital, Medical College of Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jihong Zhang
- Central Laboratory, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jikai Liu
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Zhanyu Wang
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| | - Gang Chen
- Department of Urology, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
873
|
Robertson-Tessi M, Gillies RJ, Gatenby RA, Anderson ARA. Impact of metabolic heterogeneity on tumor growth, invasion, and treatment outcomes. Cancer Res 2015; 75:1567-79. [PMID: 25878146 DOI: 10.1158/0008-5472.can-14-1428] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histopathologic knowledge that extensive heterogeneity exists between and within tumors has been confirmed and deepened recently by molecular studies. However, the impact of tumor heterogeneity on prognosis and treatment remains as poorly understood as ever. Using a hybrid multiscale mathematical model of tumor growth in vascularized tissue, we investigated the selection pressures exerted by spatial and temporal variations in tumor microenvironment and the resulting phenotypic adaptations. A key component of this model is normal and tumor metabolism and its interaction with microenvironmental factors. The metabolic phenotype of tumor cells is plastic, and microenvironmental selection leads to increased tumor glycolysis and decreased pH. Once this phenotype emerges, the tumor dramatically changes its behavior due to acid-mediated invasion, an effect that depends on both variations in the tumor cell phenotypes and their spatial distribution within the tumor. In early stages of growth, tumors are stratified, with the most aggressive cells developing within the interior of the tumor. These cells then grow to the edge of the tumor and invade into the normal tissue using acidosis. Simulations suggest that diffusible cytotoxic treatments, such as chemotherapy, may increase the metabolic aggressiveness of a tumor due to drug-mediated selection. Chemotherapy removes the metabolic stratification of the tumor and allows more aggressive cells to grow toward blood vessels and normal tissue. Antiangiogenic therapy also selects for aggressive phenotypes due to degradation of the tumor microenvironment, ultimately resulting in a more invasive tumor. In contrast, pH buffer therapy slows down the development of aggressive tumors, but only if administered when the tumor is still stratified. Overall, findings from this model highlight the risks of cytotoxic and antiangiogenic treatments in the context of tumor heterogeneity resulting from a selection for more aggressive behaviors.
Collapse
Affiliation(s)
- Mark Robertson-Tessi
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Alexander R A Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
874
|
Tung CH, Qi J, Hu L, Han MS, Kim Y. A Quick Responsive Fluorogenic pH Probe for Ovarian Tumor Imaging. Theranostics 2015; 5:1166-74. [PMID: 26284146 PMCID: PMC4533099 DOI: 10.7150/thno.12813] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/09/2015] [Indexed: 12/25/2022] Open
Abstract
A novel cell-permeable compound, CypH-1, that is non-fluorescent at neutral pH, but fluoresces under mildly acidic conditions with a near infrared maximum emission wavelength was designed for the detection of tumors in the clinical setting. The potential of CypH-1 in ovarian cancer imaging was demonstrated using a murine model. The intraperitoneally administered CypH-1 results in a robust fluorescence signal of discrete neoplastic lesions with millimeter range resolution within few hours. Moreover, fluorescence signal is strikingly enhanced at peripheral regions of tumors at the microscopic level suggesting a sharp physiological difference at the tumor/normal tissue interface. This robust acid-activated imaging agent is expected to have significant impact in broad surgical and diagnostic applications.
Collapse
|
875
|
Shu NS, Chung MS, Yao L, An M, Qiang W. Residue-specific structures and membrane locations of pH-low insertion peptide by solid-state nuclear magnetic resonance. Nat Commun 2015. [PMID: 26195283 PMCID: PMC4518304 DOI: 10.1038/ncomms8787] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pH-low insertion peptide (pHLIP) binds to a membrane at pH 7.4 unstructured but folds across the bilayer as a transmembrane helix at pH∼6. Despite their promising applications as imaging probes and drug carriers that target cancer cells for cytoplasmic cargo delivery, the mechanism of pH modulation on pHLIP-membrane interactions has not been completely understood. Here, we show the first study on membrane-associated pHLIP using solid-state NMR spectroscopy. Data on residue-specific conformation and membrane location describe pHLIP in various surface-bound and membrane-inserted states at pH 7.4, 6.4 and 5.3. The critical membrane-adsorbed state is more complex than previously envisioned. At pH 6.4, for the major unstructured population, the peptide sinks deeper into the membrane in a state II′ that is distinct from the adsorbed state II observed at pH 7.4, which may enable pHLIP to sense slight change in acidity even before insertion. The pH-low insertion peptides (pHLIPs) respond to environmental pH variations by forming transmembrane α-helices. Here, the authors present the residue-specific structures and membrane locations of pHLIPs at different pH levels to probe the mechanism of their pH-dependant membrane insertion.
Collapse
Affiliation(s)
- Nicolas S Shu
- Department of Chemistry, State University of New York, Binghamton, New York 13902, USA
| | - Michael S Chung
- Department of Chemistry, State University of New York, Binghamton, New York 13902, USA
| | - Lan Yao
- Department of Physics, Applied Physics and Astronomy, State University of New York, Binghamton, New York 13902, USA
| | - Ming An
- Department of Chemistry, State University of New York, Binghamton, New York 13902, USA
| | - Wei Qiang
- Department of Chemistry, State University of New York, Binghamton, New York 13902, USA
| |
Collapse
|
876
|
Fais S, Venturi G, Gatenby B. Microenvironmental acidosis in carcinogenesis and metastases: new strategies in prevention and therapy. Cancer Metastasis Rev 2015; 33:1095-108. [PMID: 25376898 PMCID: PMC4244550 DOI: 10.1007/s10555-014-9531-3] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Much effort is currently devoted to developing patient-specific cancer therapy based on molecular characterization of tumors. In particular, this approach seeks to identify driver mutations that can be blocked through small molecular inhibitors. However, this approach is limited by extensive intratumoral genetic heterogeneity, and, not surprisingly, even dramatic initial responses are typically of limited duration as resistant tumor clones rapidly emerge and proliferate. We propose an alternative approach based on observations that while tumor evolution produces genetic divergence, it is also associated with striking phenotypic convergence that loosely correspond to the well-known cancer “hallmarks”. These convergent properties can be described as driver phenotypes and may be more consistently and robustly expressed than genetic targets. To this purpose, it is necessary to identify strategies that are critical for cancer progression and metastases, and it is likely that these driver phenotypes will be closely related to cancer “hallmarks”. It appears that an antiacidic approach, by targetting a driver phenotype in tumors, may be thought as a future strategy against tumors in either preventing the occurrence of cancer or treating tumor patients with multiple aims, including the improvement of efficacy of existing therapies, possibly reducing their systemic side effects, and controlling tumor growth, progression, and metastasis. This may be achieved with existing molecules such as proton pump inhibitors (PPIs) and buffers such as sodium bicarbonate, citrate, or TRIS.
Collapse
Affiliation(s)
- Stefano Fais
- Department of Therapeutic Research and Medicines Evaluation, Unit of Antitumor Drugs, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
- Department of Drug Research and Medicines Evaluation, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena 299, 00161 Rome, Italy
| | - Giulietta Venturi
- Department of Therapeutic Research and Medicines Evaluation, Unit of Antitumor Drugs, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
| | - Bob Gatenby
- Radiology Department, Cancer Biology and Evolution Program Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| |
Collapse
|
877
|
Abstract
A major goal of modern medicine is increasing patient specificity so that the right treatment is administered to the right patient at the right time with the right dose. While current cancer studies have largely focused on identification of genetic or epigenetic properties of tumor cells, emerging evidence has clearly demonstrated substantial genetic heterogeneity between tumors in the same patient and within subclones of a single tumor. Thus, molecular analysis from populations of cells (either a whole tumor or small biopsy of that tumor) is, at best, an incomplete representation of the underlying biology. These observations indicate a significant need to define intratumoral evolutionary dynamics that yield the observed spatial variations in cellular properties. It is generally accepted that genetic heterogeneity among cancer cells is a manifestation of intratumoral evolution, and this is typically viewed as a consequence of random mutations generated by genomic instability within the cancer cells. We suggest that this represents an incomplete view of Darwinian dynamics, which typically are governed by phenotypic variations in response to spatial and temporal heterogeneity in environmental selection forces. We propose that pathologic feature analysis can provide precise information regarding regional variations in environmental selection forces and phenotypic adaptations. These observations can be integrated using quantitative, spatially explicit methods developed in landscape ecology to interrogate heterogenous biological processes in tumors within individual patients. The ability to investigate tumor heterogeneity has been shown to inform physicians regarding critical aspects of cancer progression including invasion, metastasis, drug resistance, and disease relapse.
Collapse
|
878
|
Chung MF, Liu HY, Lin KJ, Chia WT, Sung HW. A pH-Responsive Carrier System that Generates NO Bubbles to Trigger Drug Release and Reverse P-Glycoprotein-Mediated Multidrug Resistance. Angew Chem Int Ed Engl 2015; 54:9890-3. [PMID: 26136242 DOI: 10.1002/anie.201504444] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Indexed: 11/08/2022]
Abstract
Multidrug resistance (MDR) resulting from the overexpression of drug transporters such as P-glycoprotein (Pgp) increases the efflux of drugs and thereby limits the effectiveness of chemotherapy. To address this issue, this work develops an injectable hollow microsphere (HM) system that carries the anticancer agent irinotecan (CPT-11) and a NO-releasing donor (NONOate). Upon injection of this system into acidic tumor tissue, environmental protons infiltrate the shell of the HMs and react with their encapsulated NONOate to form NO bubbles that trigger localized drug release and serve as a Pgp-mediated MDR reversal agent. The site-specific drug release and the NO-reduced Pgp-mediated transport can cause the intracellular accumulation of the drug at a concentration that exceeds the cell-killing threshold, eventually inducing its antitumor activity. These results reveal that this pH-responsive HM carrier system provides a potentially effective method for treating cancers that develop MDR.
Collapse
Affiliation(s)
- Ming-Fan Chung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan (ROC)
| | - Hung-Yi Liu
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan (ROC)
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan (ROC)
| | - Wei-Tso Chia
- Department of Orthopaedics, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan (ROC).
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan (ROC).
| |
Collapse
|
879
|
Chung MF, Liu HY, Lin KJ, Chia WT, Sung HW. A pH-Responsive Carrier System that Generates NO Bubbles to Trigger Drug Release and Reverse P-Glycoprotein-Mediated Multidrug Resistance. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201504444] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
880
|
Huo S, Dong J, Shen S, Ren Y, Song C, Xu J, Shi T. L-selenomethionine reduces platinum(IV) anticancer model compounds at strikingly faster rates than L-methionine. Dalton Trans 2015; 43:15328-36. [PMID: 25075569 DOI: 10.1039/c4dt01528b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
L-Selenomethionine (SeMet), the predominant form of selenium acquired from the diet by humans, has been used as a supplement, and exhibit some important functions like cancer prevention and antioxidative defense. Its interactions with Pt(II) anticancer drugs have been characterized, but its redox reactions with platinum(IV) anticancer prodrugs have not been exploited. In this work, the oxidation of SeMet by Pt(IV) anticancer model compounds trans-[PtX2(CN)4](2-) (X = Cl, Br) was characterized. A stopped-flow spectrometer was used to record the rapid scan spectra and to follow the reaction kinetics over a wide pH range. An overall second-order rate law was derived: -d[Pt(IV)]/dt = k'[Pt(IV)][SeMet], where k' pertains to the observed second-order rate constants. The k'-pH profiles showed that k' increased only about 6 times even though the solution pH was varied from 0.25 to 10.5. The redox stoichiometry was determined as Δ[Pt(IV)]/Δ[SeMet] = 1 : (1.07 ± 0.07), suggesting that SeMet was oxidized to selenomethionine selenoxide. The selenoxide together with its hydrated form was identified explicitly by high resolution mass spectral analysis. A reaction mechanism was proposed which encompassed three parallel rate-determining steps relying on the protolytic species of SeMet. Rate constants of the rate-determining steps were obtained from the simulations of the k'-pH profiles. Activation parameters were determined for the reactions of the zwitterionic form of SeMet with the Pt(IV) complexes. A bridged electron transfer process is delineated in the rate-determining steps and several lines of evidence support the bridged electron transfer mode. Strikingly, reduction of [PtX2(CN)4](2-) by SeMet is 3.7 × 10(3)-5.7 × 10(4) times faster than that by L-methionine. Some potential biological consequences resulting from the strikingly fast reduction are discussed.
Collapse
Affiliation(s)
- Shuying Huo
- College of Chemistry and Environmental Science, and the MOE Key Laboratory of Medicinal Chemistry and Molecular Diagnostics, Hebei University, Baoding 071002, Hebei Province, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
881
|
Extracellular acidification synergizes with PDGF to stimulate migration of mouse embryo fibroblasts through activation of p38MAPK with a PTX-sensitive manner. Biochem Biophys Res Commun 2015; 460:191-7. [DOI: 10.1016/j.bbrc.2015.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/02/2015] [Indexed: 11/19/2022]
|
882
|
Tsai JLL, Zou T, Liu J, Chen T, Chan AOY, Yang C, Lok CN, Che CM. Luminescent platinum(ii) complexes with self-assembly and anti-cancer properties: hydrogel, pH dependent emission color and sustained-release properties under physiological conditions. Chem Sci 2015; 6:3823-3830. [PMID: 29218152 PMCID: PMC5707448 DOI: 10.1039/c4sc03635b] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/13/2015] [Indexed: 12/28/2022] Open
Abstract
Luminescent platinum(ii) complexes show anti-cancer and pH-dependent self-assembly and sustained-release properties under physiological conditions.
Supramolecular interactions are of paramount importance in biology and chemistry, and can be used to develop new vehicles for drug delivery. Recently, there is a surge of interest on self-assembled functional supramolecular structures driven by intermolecular metal–metal interactions in cellular conditions. Herein we report a series of luminescent Pt(ii) complexes [Pt(C^N^Npyr)(C
Created by potrace 1.16, written by Peter Selinger 2001-2019
]]>
NR)]+ [HC^N^Npyr = 2-phenyl-6-(1H-pyrazol-3-yl)-pyridine)] containing pincer type ligands having pyrazole moieties. These Pt(ii) complexes exert potent cytotoxicity to a panel of cancer cell lines including primary bladder cancer cells and display strong phosphorescence that is highly sensitive to the local environment. The self-assembly of these complexes is significantly affected by pH of the solution medium. Based on TEM, SEM, ESI-MS, absorption and emission spectroscopy, and fluorescence microscopy together with cell based assays, [Pt(C^N^Npyr)(C
Created by potrace 1.16, written by Peter Selinger 2001-2019
]]>
NR)]+ complexes were observed to self-assemble into orange phosphorescent polymeric aggregates driven by intermolecular Pt(ii)–Pt(ii) and ligand–ligand interactions in a low-pH physiological medium. Importantly, the intracellular assembly and dis-assembly of [Pt(C^N^Npyr)(C
Created by potrace 1.16, written by Peter Selinger 2001-2019
]]>
NR)]+ are accompanied by change of emission color from orange to green. These [Pt(C^N^Npyr)(C
Created by potrace 1.16, written by Peter Selinger 2001-2019
]]>
NR)]+ complexes accumulated in the lysosomes of cancer cells, increased the lysosomal membrane permeability and induced cell death. One of these platinum(ii) complexes formed hydrogels which displayed pH-responsive and sustained release properties, leading to low-pH-stimulated and time-dependent cytotoxicity towards cancer cells. These hydrogels can function as vehicles to deliver anti-cancer agent cargo, such as the bioactive natural products studied in this work.
Collapse
Affiliation(s)
- Johnson Lui-Lui Tsai
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China .
| | - Taotao Zou
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China . .,HKU Shenzhen Institute of Research and Innovation , Shenzhen 518053 , China
| | - Jia Liu
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China .
| | - Tianfeng Chen
- Department of Chemistry , Jinan University , Guangzhou 510632 , China
| | - Anna On-Yee Chan
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China .
| | - Chen Yang
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China . .,HKU Shenzhen Institute of Research and Innovation , Shenzhen 518053 , China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China .
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry , Institute of Molecular Functional Materials , Chemical Biology Centre and Department of Chemistry , The University of Hong Kong , Pokfulam Road , Hong Kong , China . .,HKU Shenzhen Institute of Research and Innovation , Shenzhen 518053 , China
| |
Collapse
|
883
|
Discovery and characterization of an endogenous CXCR4 antagonist. Cell Rep 2015; 11:737-47. [PMID: 25921529 DOI: 10.1016/j.celrep.2015.03.061] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/10/2015] [Accepted: 03/25/2015] [Indexed: 11/23/2022] Open
Abstract
CXCL12-CXCR4 signaling controls multiple physiological processes and its dysregulation is associated with cancers and inflammatory diseases. To discover as-yet-unknown endogenous ligands of CXCR4, we screened a blood-derived peptide library for inhibitors of CXCR4-tropic HIV-1 strains. This approach identified a 16 amino acid fragment of serum albumin as an effective and highly specific CXCR4 antagonist. The endogenous peptide, termed EPI-X4, is evolutionarily conserved and generated from the highly abundant albumin precursor by pH-regulated proteases. EPI-X4 forms an unusual lasso-like structure and antagonizes CXCL12-induced tumor cell migration, mobilizes stem cells, and suppresses inflammatory responses in mice. Furthermore, the peptide is abundant in the urine of patients with inflammatory kidney diseases and may serve as a biomarker. Our results identify EPI-X4 as a key regulator of CXCR4 signaling and introduce proteolysis of an abundant precursor protein as an alternative concept for chemokine receptor regulation.
Collapse
|
884
|
Bellenghi M, Puglisi R, Pedini F, De Feo A, Felicetti F, Bottero L, Sangaletti S, Errico MC, Petrini M, Gesumundo C, Denaro M, Felli N, Pasquini L, Tripodo C, Colombo MP, Carè A, Mattia G. SCD5-induced oleic acid production reduces melanoma malignancy by intracellular retention of SPARC and cathepsin B. J Pathol 2015; 236:315-25. [PMID: 25802234 DOI: 10.1002/path.4535] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/10/2015] [Accepted: 03/17/2015] [Indexed: 01/17/2023]
Abstract
A proper balance between saturated and unsaturated fatty acids (FAs) is required for maintaining cell homeostasis. The increased demand of FAs to assemble the plasma membranes of continuously dividing cancer cells might unbalance this ratio and critically affect tumour outgrowth. We unveiled the role of the stearoyl-CoA desaturase SCD5 in converting saturated FAs into mono-unsaturated FAs during melanoma progression. SCD5 is down-regulated in advanced melanoma and its restored expression significantly reduced melanoma malignancy, both in vitro and in vivo, through a mechanism governing the secretion of extracellular matrix proteins, such as secreted protein acidic and rich in cysteine (SPARC) and collagen IV and of their proteases, such as cathepsin B. Enforced expression of SCD5 or supplementation of its enzymatic product, oleic acid, reduced the intracellular pH (pHe > pHi) and, in turn, vesicular trafficking across plasma membranes as well as melanoma dissemination. This intracellular acidification appears also to depend on SCD5-induced reduction of the C2 subunit of the vacuolar H(+) -ATPase, a proton pump whose inhibition changes the secretion profile of cancer cells. Our data support a role for SCD5 and its enzymatic product, oleic acid, in protection against malignancy, offering an explanation for the beneficial Mediterranean diet. Furthermore, SCD5 appears to functionally connect tumour cells and the surrounding stroma toward modification of the tumour microenvironment, with consequences on tumour spread and resistance to treatment.
Collapse
Affiliation(s)
- Maria Bellenghi
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rossella Puglisi
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pedini
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra De Feo
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Federica Felicetti
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lisabianca Bottero
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sabina Sangaletti
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Maria Cristina Errico
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Petrini
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Cinzia Gesumundo
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Denaro
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Rome, Italy
| | - Nadia Felli
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Pasquini
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Mario Paolo Colombo
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Alessandra Carè
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Mattia
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
885
|
Rosca EV, Wright M, Gonitel R, Gedroyc W, Miller AD, Thanou M. Thermosensitive, near-infrared-labeled nanoparticles for topotecan delivery to tumors. Mol Pharm 2015; 12:1335-46. [PMID: 25826624 DOI: 10.1021/mp5002679] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Liposomal nanoparticles have proven to be versatile systems for drug delivery. However, the progress in clinic has been slower and less efficient than expected. This suggests a need for further development using carefully designed chemical components to improve usefulness under clinical conditions and maximize therapeutic effect. For cancer chemotherapy, PEGylated liposomes were the first nanomedicine to reach the market and have been used clinically for several years. Approaches toward targeted drug delivery using next generation "thermally triggered" nanoparticles are now in clinical trials. However, clinically tested thermosensitive liposomes (TSLs) lack the markers that allow tumor labeling and improved imaging for tissue specific applied hyperthermia. Here we describe the development of optically labeled TSLs for image guidance drug delivery and proof-of-concept results for their application in the treatment of murine xenograft tumors using the anticancer drug topotecan. These labeled TSLs also allow the simultaneous, real-time diagnostic imaging of nanoparticle biodistribution using a near-infrared (NIR; 750-950 nm) fluorophore coupled to a lipidic component of the lipid bilayer. When combined with multispectral fluorescence analysis, this allows for specific and high sensitivity tracking of the nanoparticles in vivo. The application of NIR fluorescence-labeled TSLs could have a transformative effect on future cancer chemotherapy.
Collapse
Affiliation(s)
- Elena V Rosca
- †Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Michael Wright
- †Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Roman Gonitel
- †Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Wladyslaw Gedroyc
- §Department of Experimental Medicine, Imperial College London, London, U.K
| | - Andrew D Miller
- †Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Maya Thanou
- †Institute of Pharmaceutical Science, King's College London, London, U.K
| |
Collapse
|
886
|
Dhar G, Sen S, Chaudhuri G. Acid gradient across plasma membrane can drive phosphate bond synthesis in cancer cells: acidic tumor milieu as a potential energy source. PLoS One 2015; 10:e0124070. [PMID: 25874623 PMCID: PMC4398327 DOI: 10.1371/journal.pone.0124070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/25/2015] [Indexed: 01/20/2023] Open
Abstract
Aggressive cancers exhibit an efficient conversion of high amounts of glucose to lactate accompanied by acid secretion, a phenomenon popularly known as the Warburg effect. The acidic microenvironment and the alkaline cytosol create a proton-gradient (acid gradient) across the plasma membrane that represents proton-motive energy. Increasing experimental data from physiological relevant models suggest that acid gradient stimulates tumor proliferation, and can also support its energy needs. However, direct biochemical evidence linking extracellular acid gradient to generation of intracellular ATP are missing. In this work, we demonstrate that cancer cells can synthesize significant amounts of phosphate-bonds from phosphate in response to acid gradient across plasma membrane. The noted phenomenon exists in absence of glycolysis and mitochondrial ATP synthesis, and is unique to cancer. Biochemical assays using viable cancer cells, and purified plasma membrane vesicles utilizing radioactive phosphate, confirmed phosphate-bond synthesis from free phosphate (Pi), and also localization of this activity to the plasma membrane. In addition to ATP, predominant formation of pyrophosphate (PPi) from Pi was also observed when plasma membrane vesicles from cancer cells were subjected to trans-membrane acid gradient. Cancer cytosols were found capable of converting PPi to ATP, and also stimulate ATP synthesis from Pi from the vesicles. Acid gradient created through glucose metabolism by cancer cells, as observed in tumors, also proved critical for phosphate-bond synthesis. In brief, these observations reveal a role of acidic tumor milieu as a potential energy source and may offer a novel therapeutic target.
Collapse
Affiliation(s)
- Gautam Dhar
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1740, United States of America
- * E-mail: (GD); (GC)
| | - Suvajit Sen
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1740, United States of America
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States of America
| | - Gautam Chaudhuri
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1740, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1740, United States of America
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States of America
- * E-mail: (GD); (GC)
| |
Collapse
|
887
|
Ban JJ, Lee M, Im W, Kim M. Low pH increases the yield of exosome isolation. Biochem Biophys Res Commun 2015; 461:76-9. [PMID: 25849885 DOI: 10.1016/j.bbrc.2015.03.172] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/29/2015] [Indexed: 12/11/2022]
Abstract
Exosomes are the extracellular vesicles secreted by various cells. Exosomes mediate intercellular communication by delivering a variety of molecules between cells. Cancer cell derived exosomes seem to be related with tumor progression and metastasis. Tumor microenvironment is thought to be acidic and this low pH controls exosome physiology, leading to tumor progression. Despite the importance of microenvironmental pH on exosome, most of exosome studies have been performed without regard to pH. Therefore, the difference of exosome stability and yield of isolation by different pH need to be studied. In this research, we investigated the yield of total exosomal protein and RNA after incubation in acidic, neutral and alkaline conditioned medium. Representative exosome markers were investigated by western blot after incubation of exosomes in different pH. As a result, the concentrations of exosomal protein and nucleic acid were significantly increased after incubation in the acidic medium compared with neutral medium. The higher levels of exosome markers including CD9, CD63 and HSP70 were observed after incubation in an acidic environment. On the other hand, no exosomal protein, exosomal RNA and exosome markers have been detected after incubation in an alkaline condition. In summary, our results indicate that the acidic condition is the favorable environment for existence and isolation of exosomes.
Collapse
Affiliation(s)
- Jae-Jun Ban
- Department of Bioscience and Biotechnology, College of Life Science, Institute of Biotechnology, Sejong University, Seoul, South Korea
| | - Mijung Lee
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Wooseok Im
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea.
| | - Manho Kim
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea; Protein Metabolism Medical Research Center, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
888
|
1,2,3,4,6-Penta-O-galloylglucose within Galla Chinensis Inhibits Human LDH-A and Attenuates Cell Proliferation in MDA-MB-231 Breast Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:276946. [PMID: 25918543 PMCID: PMC4396556 DOI: 10.1155/2015/276946] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/20/2014] [Accepted: 11/20/2014] [Indexed: 12/28/2022]
Abstract
A characteristic feature of aggressive malignancy is the overexpression of lactic acid dehydrogenase- (LDH-) A, concomitant to pericellular accumulation of lactate. In a recent high-throughput screening, we identified Rhus chinensis (Mill.) gallnut (RCG) (also known as Galla Chinensis) extract as a potent (IC50 < 1 µg/mL) inhibitor of human LDH-A (hLDH-A). In this study, through bioactivity guided fractionation of the crude extract, the data demonstrate that penta-1,2,3,4,6-O-galloyl-β-D-glucose (PGG) was a primary constituent responsible for hLDH-A inhibition, present at ~9.95 ± 0.34% dry weight. Theoretical molecular docking studies of hLDH-A indicate that PGG acts through competitive binding at the NADH cofactor site, effects confirmed by functional enzyme studies where the IC50 = 27.32 nM was reversed with increasing concentration of NADH. Moreover, we confirm protein expression of hLDH-A in MDA-231 human breast carcinoma cells and show that PGG was toxic (LC50 = 94.18 µM), parallel to attenuated lactic acid production (IC50 = 97.81 µM). In a 72-hour cell proliferation assay, PGG was found to be a potent cytostatic agent with ability to halt cell division (IC50 = 1.2 µM) relative to paclitaxel (IC50 < 100 nM). In summary, these findings demonstrate that PGG is a potent hLDH-A inhibitor with significant capacity to halt proliferation of human breast cancer cells.
Collapse
|
889
|
Grillo-Hill BK, Choi C, Jimenez-Vidal M, Barber DL. Increased H⁺ efflux is sufficient to induce dysplasia and necessary for viability with oncogene expression. eLife 2015; 4. [PMID: 25793441 PMCID: PMC4392478 DOI: 10.7554/elife.03270] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Intracellular pH (pHi) dynamics is increasingly recognized as an important regulator of a range of normal and pathological cell behaviors. Notably, increased pHi is now acknowledged as a conserved characteristic of cancers and in cell models is confirmed to increase proliferation and migration as well as limit apoptosis. However, the significance of increased pHi for cancer in vivo remains unresolved. Using Drosophila melanogaster, we show that increased pHi is sufficient to induce dysplasia in the absence of other transforming cues and potentiates growth and invasion with oncogenic Ras. Using a genetically encoded biosensor we also confirm increased pHi in situ. Moreover, in Drosophila models and clonal human mammary cells we show that limiting H(+) efflux with oncogenic Raf or Ras induces acidosis and synthetic lethality. Further, we show lethality in invasive primary tumor cell lines with inhibiting H(+) efflux. Synthetic lethality with reduced H(+) efflux and activated oncogene expression could be exploited therapeutically to restrain cancer progression while limiting off-target effects.
Collapse
Affiliation(s)
- Bree K Grillo-Hill
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Changhoon Choi
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Maite Jimenez-Vidal
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
890
|
Molecular Mechanism for the Control of Eukaryotic Elongation Factor 2 Kinase by pH: Role in Cancer Cell Survival. Mol Cell Biol 2015; 35:1805-24. [PMID: 25776553 DOI: 10.1128/mcb.00012-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/03/2015] [Indexed: 01/09/2023] Open
Abstract
Acidification of the extracellular and/or intracellular environment is involved in many aspects of cell physiology and pathology. Eukaryotic elongation factor 2 kinase (eEF2K) is a Ca(2+)/calmodulin-dependent kinase that regulates translation elongation by phosphorylating and inhibiting eEF2. Here we show that extracellular acidosis elicits activation of eEF2K in vivo, leading to enhanced phosphorylation of eEF2. We identify five histidine residues in eEF2K that are crucial for the activation of eEF2K during acidosis. Three of them (H80, H87, and H94) are in its calmodulin-binding site, and their protonation appears to enhance the ability of calmodulin to activate eEF2K. The other two histidines (H227 and H230) lie in the catalytic domain of eEF2K. We also identify His108 in calmodulin as essential for activation of eEF2K. Acidification of cancer cell microenvironments is a hallmark of malignant solid tumors. Knocking down eEF2K in cancer cells attenuated the decrease in global protein synthesis when cells were cultured at acidic pH. Importantly, activation of eEF2K is linked to cancer cell survival under acidic conditions. Inhibition of eEF2K promotes cancer cell death under acidosis.
Collapse
|
891
|
Burns KE, Robinson MK, Thévenin D. Inhibition of cancer cell proliferation and breast tumor targeting of pHLIP-monomethyl auristatin E conjugates. Mol Pharm 2015; 12:1250-8. [PMID: 25741818 DOI: 10.1021/mp500779k] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Localized delivery is vital for the successful development of novel and effective therapeutics for the treatment of cancer. The targeting and delivery described herein is based on the pH (low) insertion peptide (pHLIP), a unique delivery peptide that can selectively target tumors in mice and translocate and release cargo molecules intracellularly based solely on the low extracellular pH intrinsic to cancer cells. In this study, we investigate the efficacy of pHLIP to target and deliver the highly potent and clinically validated microtubule inhibitor monomethyl auristatin E (MMAE) to cancer cells and breast tumors. We show that pHLIP-MMAE conjugates induce a potent cytotoxic effect (>90% inhibition of cell growth) in a concentration- and pH-dependent manner after only 2 h incubation without any apparent disruption of the plasma membrane. pHLIP-MMAE conjugates exhibit between an 11- and 144-fold higher antiproliferative effect at low pH than that at physiological pH and a pronounced pH-dependent cytotoxicity as compared to that of free drug. Furthermore, we demonstrate that a pHLIP-MMAE drug conjugate effectively targets triple-negative breast tumor xenografts in mice. These results indicate that pHLIP-based auristatin conjugates may have an enhanced therapeutic window as compared to that of free drug, providing a targeting mechanism to attenuate systemic toxicity.
Collapse
Affiliation(s)
- Kelly E Burns
- †Department of Chemistry, Lehigh University, 6 East Packer Avenue, Bethlehem, Pennsylvania 18015, United States
| | - Matthew K Robinson
- ‡Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, United States
| | - Damien Thévenin
- †Department of Chemistry, Lehigh University, 6 East Packer Avenue, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
892
|
Abstract
Cancers progress through a series of events that can be characterized as "somatic evolution." A central premise of Darwinian evolutionary theory is that the environment imparts pressure to select for species that are most fit within that particular microenvironmental context. Furthermore, the rate of evolution is proportional to both (1) the strength of the environmental selection and (2) the phenotypic variance of the selected population. It is notable that, during the progression of cancers from carcinogenesis to local invasion to metastasis, the selective landscape continuously changes, and throughout this process, there is increased selection for cells that have altered metabolic phenotypes: implying that these phenotypes impart a selective advantage during the process of environmental selection. One of the most prevalent selected phenotypes is that of aerobic glycolysis, that is, the continued fermentation of glucose even in the presence of adequate oxygen. The mechanisms of this so-called "Warburg effect" have been well studied, and there are multiple models to explain how this occurs at the molecular level. Herein, we propose that unifying insights can be gained by evaluating the environmental context within which this phenotype arises. In other words, we focus not on the "how" but the "why" do cancer cells exhibit high aerobic glycolysis. This is best approached by examining the sequelae of aerobic glycolysis that may impart a selective advantage. Many of these have been considered, including generation of anabolic substrates, response rates of glycolysis vis-à-vis respiration, and generation of antioxidants. A further sequeala considered here is that aerobic glycolysis results in a high rate of lactic acid production; resulting in acidification of the extracellular space. Indeed, it has been shown that a low extracellular pH promotes local invasion, promotes metastasis, and inhibits antitumor immunity. In naturally occurring cancers, low extracellular pH is a strong negative prognostic indicator of metastasis-free survival. Furthermore, it has been shown that inhibition of extracellular acidosis can inhibit metastasis and promote antitumor immunity. Hence, we propose that excess acid production confers a selective advantage for cells during the somatic evolution of cancers.
Collapse
Affiliation(s)
- Robert J Gillies
- From the Departments of Cancer Imaging and Metabolism and Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | |
Collapse
|
893
|
Scott HL, Nguyen VP, Alves DS, Davis FL, Booth KR, Bryner J, Barrera FN. The negative charge of the membrane has opposite effects on the membrane entry and exit of pH-low insertion peptide. Biochemistry 2015; 54:1709-12. [PMID: 25692747 DOI: 10.1021/acs.biochem.5b00069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pH-low insertion peptide (pHLIP) targets acidic diseases such as cancer. The acidity of the environment causes key aspartic acids in pHLIP to become protonated, causing the peptide to insert into membranes. Here we investigate how the negative charge of the membrane influences how pHLIP enters and exits the lipid bilayer. We found that electrostatic repulsion affected differently the membrane entry and exit of pHLIP for negatively charged membranes. As a consequence, a large hysteresis was observed. We propose this is not a consequence of structural changes but results from local changes in the environment of aspartic acids, shifting their pK values.
Collapse
Affiliation(s)
- Haden L Scott
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee , Knoxville, Tennessee 37996, United States
| | | | | | | | | | | | | |
Collapse
|
894
|
Kaur S, Prasad C, Balakrishnan B, Banerjee R. Trigger responsive polymeric nanocarriers for cancer therapy. Biomater Sci 2015. [PMID: 26221933 DOI: 10.1039/c5bm00002e] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Conventional chemotherapy for the treatment of cancer has limited specificity when administered systemically and is often associated with toxicity issues. Enhanced accumulation of polymeric nanocarriers at a tumor site may be achieved by passive and active targeting. Incorporation of trigger responsiveness into these polymeric nanocarriers improves the anticancer efficacy of such systems by modulating the release of the drug according to the tumor environment. Triggers used for tumor targeting include internal triggers such as pH, redox and enzymes and external triggers such as temperature, magnetic field, ultrasound and light. While internal triggers are specific cues of the tumor microenvironment, external triggers are those which are applied externally to control the release. This review highlights the various strategies employed for the preparation of such trigger responsive polymeric nanocarriers for cancer therapy and provides an overview of the state of the art in this field.
Collapse
Affiliation(s)
- Shahdeep Kaur
- Nanomedicine Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India.
| | | | | | | |
Collapse
|
895
|
Kim MG, Park JY, Miao W, Lee J, Oh YK. Polyaptamer DNA nanothread-anchored, reduced graphene oxide nanosheets for targeted delivery. Biomaterials 2015; 48:129-36. [PMID: 25701038 DOI: 10.1016/j.biomaterials.2015.01.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/21/2014] [Accepted: 01/20/2015] [Indexed: 10/24/2022]
Abstract
Here, we report reduced graphene oxide (rGO) nanosheets anchoring receptor-specific polyaptamer nanothreads for targeted drug delivery. DNA polyaptamer nanothreads of protein tyrosine kinase 7 receptor (PTK7) were synthesized by rolling cycle amplification. To strengthen the anchoring of polyaptamer nanothreads onto rGO, oligoT bridge domain was introduced between each repeating PTK7 aptamer sequence. As compared to PTK7 polyaptamer nanothreads alone, PTK7 polyaptamer nanothreads with 22-mer oligoT bridges (PNT) showed higher anchoring capacity onto rGO nanosheets. Nanothread-coated surface morphology of PNTrGO was observed. Coating of PNT did not affect the sizes of rGO, but reduced the zeta potential. In PTK7-negative Ramos cells, the uptake of PNT-anchored rGO (PNTrGO) did not differ from that of oligoT-bridged scrambled polyaptamer-anchored rGO (SNTrGO). However, in CCRF-CEM leukemia cells overexpressing PTK7, the uptake of PNTrGO was 2.1-fold higher than that of SNTrGO after 15 min pulse. In vivo distribution to CCRF-CEM tumor tissues was 2.8-fold higher in PNTrGO than in SNTrGO at 48 h post-injection. In CCRF-CEM xenografted mice, intravenously administered doxorubicin (Dox)-loaded PNTrGO showed the higher antitumor activity than other groups, reducing the tumor weight down to 12% of tumor weights of untreated mice. These results suggest the potential of PNTrGO for target-specific drug delivery nanoplatform.
Collapse
Affiliation(s)
- Mi-Gyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Joo Yeon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Wenjun Miao
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jaiwoo Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
896
|
Lozupone F, Borghi M, Marzoli F, Azzarito T, Matarrese P, Iessi E, Venturi G, Meschini S, Canitano A, Bona R, Cara A, Fais S. TM9SF4 is a novel V-ATPase-interacting protein that modulates tumor pH alterations associated with drug resistance and invasiveness of colon cancer cells. Oncogene 2015; 34:5163-74. [PMID: 25659576 DOI: 10.1038/onc.2014.437] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 12/04/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022]
Abstract
An inverted pH gradient across the cell membranes is a typical feature of malignant cancer cells that are characterized by extracellular acidosis and cytosol alkalization. These dysregulations are able to create a unique milieu that favors tumor progression, metastasis and chemo/immune-resistance traits of solid tumors. A key event mediating tumor cell pH alterations is an aberrant activation of ion channels and proton pumps such as (H+)-vacuolar-ATPase (V-ATPase). TM9SF4 is a poorly characterized transmembrane protein that we have recently shown to be related to cannibal behavior of metastatic melanoma cells. Here, we demonstrate that TM9SF4 represents a novel V-ATPase-associated protein involved in V-ATPase activation. We have observed in HCT116 and SW480 colon cancer cell lines that TM9SF4 interacts with the ATP6V1H subunit of the V-ATPase V1 sector. Suppression of TM9SF4 with small interfering RNAs strongly reduces assembly of V-ATPase V0/V1 sectors, thus reversing tumor pH gradient with a decrease of cytosolic pH, alkalization of intracellular vesicles and a reduction of extracellular acidity. Such effects are associated with a significant inhibition of the invasive behavior of colon cancer cells and with an increased sensitivity to the cytotoxic effects of 5-fluorouracil. Our study shows for the first time the important role of TM9SF4 in the aberrant constitutive activation of the V-ATPase, and the development of a malignant phenotype, supporting the potential use of TM9SF4 as a target for future anticancer therapies.
Collapse
Affiliation(s)
- F Lozupone
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - M Borghi
- Infectious, Parasitic and Immune-Mediated Diseases Department, Istituto Superiore di Sanità, Rome, Italy
| | - F Marzoli
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - T Azzarito
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - P Matarrese
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - E Iessi
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - G Venturi
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - S Meschini
- Technology and Health Department, Istituto Superiore di Sanità, Rome, Italy
| | - A Canitano
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - R Bona
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - A Cara
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| | - S Fais
- Therapeutic Research and Medicines Evaluation Department, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
897
|
Abstract
Cancer research has shifted in recent years from studying intracellular processes (identification of damaged genes and signaling pathways) to extracellular (hierarchy of tumor cells, cell transitions, clone competition) and tissue (interactions of a tumor with its environment) research. But then the next step seems to be logical: studying biochemistry of tumor-bearing organisms (namely, cancer-induced changes in cellular and tissue metabolism leading to the organism's death). These data can help to develop new methods of cancer treatment. This article discusses some of the challenges of contemporary oncology and possible ways to overcome them.
Collapse
Affiliation(s)
- A V Lichtenstein
- Blokhin Cancer Research Center, Institute of Carcinogenesis, Moscow, 115478, Russia.
| |
Collapse
|
898
|
Onyango JO, Chung MS, Eng CH, Klees LM, Langenbacher R, Yao L, An M. Noncanonical amino acids to improve the pH response of pHLIP insertion at tumor acidity. Angew Chem Int Ed Engl 2015; 54:3658-3663. [PMID: 25650762 DOI: 10.1002/anie.201409770] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/27/2014] [Indexed: 12/17/2022]
Abstract
The pH low insertion peptide (pHLIP) offers the potential to deliver drugs selectively to the cytoplasm of cancer cells based on tumor acidosis. The WT pHLIP inserts into membranes with a pH50 of 6.1, while most solid tumors have extracellular pH (pH(e)) of 6.5-7.0. To close this gap, a SAR study was carried out to search for pHLIP variants with improved pH response. Replacing Asp25 with α-aminoadipic acid (Aad) adjusts the pH50 to 6.74, matching average tumor acidity, and replacing Asp14 with γ-carboxyglutamic acid (Gla) increases the sharpness of pH response (transition over 0.5 instead of 1 pH unit). These effects are additive: the Asp14Gla/Asp25Aad double variant shows a pH50 of 6.79, with sharper transition than Asp25Aad. Furthermore, the advantage of the double variant over WT pHLIP in terms of cargo delivery was demonstrated in turn-on fluorescence assays and anti-proliferation studies (using paclitaxel as cargo) in A549 lung cancer cells at pH 6.6.
Collapse
Affiliation(s)
- Joab O Onyango
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Michael S Chung
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Chee-Huat Eng
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Lukas M Klees
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Rachel Langenbacher
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Lan Yao
- Department of Physics, Applied Physics and Astronomy State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| | - Ming An
- Department of Chemistry, State University of New York (SUNY), Binghamton University P. O. Box 6000, Binghamton, NY 13902 (USA)
| |
Collapse
|
899
|
Onyango JO, Chung MS, Eng CH, Klees LM, Langenbacher R, Yao L, An M. Noncanonical Amino Acids to Improve the pH Response of pHLIP Insertion at Tumor Acidity. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201409770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
900
|
Graham RM, Thompson JW, Webster KA. Inhibition of the vacuolar ATPase induces Bnip3-dependent death of cancer cells and a reduction in tumor burden and metastasis. Oncotarget 2015; 5:1162-73. [PMID: 24811485 PMCID: PMC4012732 DOI: 10.18632/oncotarget.1699] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The pro-apoptotic protein Bnip3 is induced by hypoxia and is present in the core regions of most solid tumors. Bnip3 induces programmed necrosis by an intrinsic caspase independent mitochondrial pathway. Many tumor cells have evolved pathways to evade Bnip3-mediated death attesting to the physiological relevance of the survival threat imposed by Bnip3. We have reported that acidosis can trigger the Bnip3 death pathway in hypoxic cells therefore we hypothesized that manipulation of intracellular pH by pharmacological inhibition of the vacuolar (v)ATPase proton pump, a significant pH control pathway, may activate Bnip3 and promote death of hypoxic cells within the tumor. Here we confirm that bafilomycin A1 (BafA1), a selective vATPase inhibitor, significantly increased death of breast cancer cells in a hypoxia and Bnip3-dependent manner and significantly reduced tumor growth in MCF7 and MDA-MB-231 mouse xenografts. Combined treatment of cells with BafA1 and the ERK1/2 inhibitor U0126 further augmented cell death. Combined treatment of mice containing MDA-MB-231 xenografts with BafA1 and the ERK1/2 inhibitor sorafenib was superior to either treatment alone and supported tumor regression. BafA1 and sorafenib treatments alone reduced MDA-MB-231 cell metastasis and again the combination was significantly more effective than either treatment alone and was without apparent side effects. These results present a novel mechanism to destroy hypoxic tumor cells that may help reverse the resistance of hypoxic tumors to radiation and chemotherapy and perhaps target tumor stem cells.
Collapse
|