1
|
Zhang Y, Song D, Han X, Liu H, Wang Y, Wang X, Dou C. Unlocking the potential of melanotransferrin (CD228): implications for targeted drug development and novel therapeutic avenues. Expert Opin Ther Targets 2024; 28:1117-1129. [PMID: 39676256 DOI: 10.1080/14728222.2024.2441705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/05/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION Melanotransferrin (CD228), a cell membrane-anchored protein, has emerged as a significant cancer antigen due to its high expression in various solid tumors. This review synthesizes the current understanding and therapeutic potential of CD228. AREAS COVERED We conducted a literature search using PubMed, Web of Science, and ClinicalTrials.gov with the keywords 'melanotransferrin' and 'CD228.' Our comprehensive review examines CD228 and its isoforms, membrane-bound CD228 (mMFI2) and soluble CD228 (sMFI2), their roles in tumorigenesis, angiogenesis, endothelial cell migration, plasminogen activation, and transendothelial transport across the BBB, as well as the current state of drug development efforts targeting CD228. EXPERT OPINION Targeting CD228 represents a promising therapeutic strategy in oncology, with mMFI2 as a potential target for solid tumors and sMFI2 valuable for disease diagnosis in malignant tumors, Alzheimer's disease, and arthritis, and facilitating macromolecular drug delivery across the blood-brain barrier (BBB). Despite its potential to transform the treatment landscape for numerous solid cancers, further research into the precise mechanisms and clinical translation of CD228-directed treatments is needed to maximize its therapeutic utility.
Collapse
Affiliation(s)
- Yanan Zhang
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Deyong Song
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Xiaolei Han
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Hong Liu
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Yunfan Wang
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Xianju Wang
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Changlin Dou
- Research and Development Center, Shandong Boan Biotechnology Co., Ltd, Yantai, Shandong, China
| |
Collapse
|
2
|
Singh CSB, Eyford BA, Abraham T, Munro L, Choi KB, Okon M, Vitalis TZ, Gabathuler R, Lu CJ, Pfeifer CG, Tian MM, Jefferies WA. Discovery of a Highly Conserved Peptide in the Iron Transporter Melanotransferrin that Traverses an Intact Blood Brain Barrier and Localizes in Neural Cells. Front Neurosci 2021; 15:596976. [PMID: 34149342 PMCID: PMC8212695 DOI: 10.3389/fnins.2021.596976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/01/2021] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) hinders the distribution of therapeutics intended for treatment of diseases of the brain. Our previous studies demonstrated that that a soluble form of melanotransferrin (MTf; Uniprot P08582; also known as p97, MFI2, and CD228), a mammalian iron-transport protein, is an effective carrier for delivery of drug conjugates across the BBB into the brain and was the first BBB targeting delivery system to demonstrate therapeutic efficacy within the brain. Here, we performed a screen to identify peptides from MTf capable of traversing the BBB. We identified a highly conserved 12-amino acid peptide, termed MTfp, that retains the ability to cross the intact BBB intact, distributes throughout the parenchyma, and enter endosomes and lysosomes within neurons, astrocytes and microglia in the brain. This peptide may provide a platform for the transport of therapeutics to the CNS, and thereby offers new avenues for potential treatments of neuropathologies that are currently refractory to existing therapies.
Collapse
Affiliation(s)
- Chaahat S B Singh
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Brett A Eyford
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Abraham
- Department of Neural and Behavioral Sciences and Microscopy Imaging Core Lab, Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kyung Bok Choi
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Mark Okon
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | | | - Reinhard Gabathuler
- Bioasis Technologies Inc., Guilford, CT, United States.,King's College London, London, United Kingdom
| | - Chieh-Ju Lu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Mei Mei Tian
- Bioasis Technologies Inc., Guilford, CT, United States
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Eyford BA, Singh CSB, Abraham T, Munro L, Choi KB, Hill T, Hildebrandt R, Welch I, Vitalis TZ, Gabathuler R, Gordon JA, Adomat H, Guns ES, Lu CJ, Pfeifer CG, Tian MM, Jefferies WA. A Nanomule Peptide Carrier Delivers siRNA Across the Intact Blood-Brain Barrier to Attenuate Ischemic Stroke. Front Mol Biosci 2021; 8:611367. [PMID: 33869275 PMCID: PMC8044710 DOI: 10.3389/fmolb.2021.611367] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) hinders the distribution of therapeutics intended for treatment of neuroinflammation (NI) of the central nervous system. A twelve-amino acid peptide that transcytoses the BBB, termed MTfp, was chemically conjugated to siRNA to create a novel peptide-oligonucleotide conjugate (POC), directed to downregulate NOX4, a gene thought responsible for oxidative stress in ischemic stroke. The MTfp-NOX4 POC has the ability to cross the intact BBB and knockdown NOX4 expression in the brain. Following induction of ischemic stroke, animals pretreated with the POC exhibited significantly smaller infarcts; accompanied by increased protection against neurological deterioration and improved recovery. The data demonstrates that the MTfp can act as a nanomule to facilitate BBB transcytosis of siRNAs; where the NOX-4 specific siRNA moiety can elicit effective therapeutic knockdown of a gene responsible for oxidative stress in the central nervous system. This study is the first to conclusively demonstrate both siRNA-carrier delivery and therapeutic efficacy in any CNS disease model where the BBB remains intact and thus offers new avenues for potential treatments of oxidative stress underlying neuroinflammation in a variety of neuropathologies that are currently refractory to existing therapies.
Collapse
Affiliation(s)
- Brett A. Eyford
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Chaahat S. B. Singh
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Abraham
- Department of Neural and Behavioral Sciences and Microscopy Imaging Core Lab, Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Kyung Bok Choi
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Tracy Hill
- Centre for Comparative Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rhonda Hildebrandt
- Centre for Comparative Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ian Welch
- Centre for Comparative Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Reinhard Gabathuler
- Bioasis Technologies Inc., Guilford, CT, United States
- King’s College London, London, United Kingdom
| | - Jacob A. Gordon
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hans Adomat
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Emma S.T. Guns
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chieh-Ju Lu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl G. Pfeifer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Mei Mei Tian
- Bioasis Technologies Inc., Guilford, CT, United States
| | - Wilfred A. Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
The membrane-bound and soluble form of melanotransferrin function independently in the diagnosis and targeted therapy of lung cancer. Cell Death Dis 2020; 11:933. [PMID: 33127882 PMCID: PMC7599248 DOI: 10.1038/s41419-020-03124-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 01/28/2023]
Abstract
Melanotransferrin (MFI2) is a newly identified tumor-associated protein, which consists of two forms of proteins, membrane-bound (mMFI2) and secretory (sMFI2). However, little is known about the expression pattern and their relevance in lung cancer. Here, we found that both two forms of MFI2 are highly expressed in lung cancer. The expression of MFI2 in lung cancer was detected by using the public database and qRT-PCR. Overexpression and knockdown cell lines and recombinant sMFI2 protein were used to study the function of mMFI2 and sMFI2. RNA-seq, protein chip, ChIP assay, Immunoprecipitation, ELISA, and immunofluorescence were used to study the molecular biological mechanism of mMFI2 and sMFI2. We found that mMFI2 promoted the expression of EMT’s common marker N-cadherin by downregulating the transcription factor KLI4, which in turn promoted tumor metastasis; sMFI2 could promote the metastasis of autologous tumor cells in an autocrine manner but the mechanism is different from that of mMFI2. In addition, sMFI2 was proved could inhibit the migration of vascular endothelial cells and subsequently enhance angiogenic responses in a paracrine manner. We propose that the expressions and functions of the two forms of MFI2 in lung cancer are relatively independent. Specifically, mMFI2 was a potential lung cancer therapeutic target, while sMFI2 was highly enriched in advanced lung cancer, and could be used as a tumor staging index.
Collapse
|
5
|
Vidal M. Exosomes and GPI-anchored proteins: Judicious pairs for investigating biomarkers from body fluids. Adv Drug Deliv Rev 2020; 161-162:110-123. [PMID: 32828789 DOI: 10.1016/j.addr.2020.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/27/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
Exosomes are 50-100 nm membranous vesicles actively released by cells which can be indicative of a diseased cell status. They contain various kinds of molecule - proteins, mRNA, miRNA, lipids - that are actively being studied as potential biomarkers. Hereafter I put forward several arguments in favor of the potential use of glycosylphosphatidylinositol-anchored proteins (GPI-APs) as biomarkers especially of cancerous diseases. I will briefly update readers on the exosome field and review various features of GPI-APs, before further discussing the advantages of this class of proteins as potential exosomal biomarkers. I will finish with a few examples of exosomal GPI-APs that have already been demonstrated to be good prognostic markers, as well as innovative approaches developed to quantify these exosomal biomarkers.
Collapse
|
6
|
Thom G, Tian MM, Hatcher JP, Rodrigo N, Burrell M, Gurrell I, Vitalis TZ, Abraham T, Jefferies WA, Webster CI, Gabathuler R. A peptide derived from melanotransferrin delivers a protein-based interleukin 1 receptor antagonist across the BBB and ameliorates neuropathic pain in a preclinical model. J Cereb Blood Flow Metab 2019; 39:2074-2088. [PMID: 29845881 PMCID: PMC6775589 DOI: 10.1177/0271678x18772998] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Delivery of biologic drugs across the blood-brain barrier is becoming a reality. However, the solutions often involve the assembly of complex multi-specific antibody molecules. Here we utilize a simple 12 amino-acid peptide originating from the melanotransferrin (MTf) protein that has shown improved brain delivery properties. 3D confocal fluorescence microscopic analysis demonstrated brain parenchymal localisation of a fluorescently labelled antibody (NIP228) when chemically conjugated to either the MTf peptide or full-length MTf protein. Measurement of plasma kinetics demonstrated the MTf peptide fusions had very similar kinetics to an unmodified NIP228 control antibody, whereas the fusion to MTf protein had significantly reduced plasma exposure most likely due to a higher tissue distribution in the periphery. Brain exposure for the MTf peptide fusions was significantly increased for the duration of the study, exceeding that of the fusions to full length MTf protein. Using a neuropathic pain model, we have demonstrated that fusions to interleukin-1 receptor antagonist (IL-1RA) are able to induce significant and durable analgesia following peripheral administration. These data demonstrate that recombinant and chemically conjugated MTf-based brain delivery vectors can deliver therapeutic levels of drug to the central nervous system.
Collapse
Affiliation(s)
| | | | - Jon P Hatcher
- Neuroscience IMED Biotech Unit, AstraZeneca, AKB, Cambridge, UK
| | | | | | - Ian Gurrell
- Neuroscience IMED Biotech Unit, AstraZeneca, AKB, Cambridge, UK
| | | | - Thomas Abraham
- Neural and Behavioural Sciences & Microscopy Imaging Core Lab, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Wilfred A Jefferies
- The Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Centre for Blood Research and Departments of Microbiology and Immunology, Medical Genetics, and Zoology University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
7
|
Ashraf A, Alepuz Guillen JA, Aljuhani M, Hubens C, So PW. Low Cerebrospinal Fluid Levels of Melanotransferrin Are Associated With Conversion of Mild Cognitively Impaired Subjects to Alzheimer's Disease. Front Neurosci 2019; 13:181. [PMID: 30906244 PMCID: PMC6419538 DOI: 10.3389/fnins.2019.00181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
The disruption of iron metabolism and iron transport proteins have been implicated in the pathogenesis of Alzheimer's disease (AD). Serum melanotransferrin (MTf), a transferrin homolog capable of reversibly binding iron, has been proposed as a biochemical marker of AD. MTf has also been shown to be elevated in iron-rich reactive microglia near amyloid plaques in AD. We examined the association of CSF MTf to hippocampal volumes and cognitive tests in 86 cognitively normal, 135 mild cognitive impairment (MCI) and 66 AD subjects. CSF was collected at baseline for MTf, Aβ, total-tau and phosphorylated-tau measurements. Serial cognitive testing with ADAS-Cog13, Rey's auditory visual learning test (RAVLT), mini-mental state examination (MMSE) were performed alongside hippocampal MRI volumetric analysis for up to 10 years after baseline measurements. High levels of baseline CSF MTf were positively associated with baseline hippocampal volume (R 2 = 22%, β = 0.202, and p = 0.017) and RAVLT scores (R 2 = 7.30%, β = -0.178, and p = 0.043) and negatively correlated to ADAS-Cog13 (R 2 = 17.3%, β = 0.247, and p = 0.003) scores in MCI subjects. Interestingly, MCI subjects that converted to AD demonstrated significantly lower levels of CSF MTf (p = 0.020) compared to MCI non-converters at baseline. We suggest the diminished CSF MTf observed in MCI-converters to AD may arise from impaired transport of MTf from blood into the brain tissue/CSF and/or increased MTf export from the CSF into the blood arising from attenuated competition with reduced levels of CSF Aβ. Further investigations are required to determine the source of CSF MTf and how brain MTf is regulated by cellular barriers, Aβ and activated microglia that surround plaques in AD pathophysiology. In conclusion, low CSF MTf may identify those MCI individuals at risk of converting to AD.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jose Andres Alepuz Guillen
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Manal Aljuhani
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Chantal Hubens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
8
|
Lentiviral transfer of an inducible transgene expressing a soluble form of Gas1 causes glioma cell arrest, apoptosis and inhibits tumor growth. Cancer Gene Ther 2010; 18:87-99. [DOI: 10.1038/cgt.2010.54] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
9
|
Karkan D, Pfeifer C, Vitalis TZ, Arthur G, Ujiie M, Chen Q, Tsai S, Koliatis G, Gabathuler R, Jefferies WA. A unique carrier for delivery of therapeutic compounds beyond the blood-brain barrier. PLoS One 2008; 3:e2469. [PMID: 18575595 PMCID: PMC2424243 DOI: 10.1371/journal.pone.0002469] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 04/24/2008] [Indexed: 02/01/2023] Open
Abstract
Background Therapeutic intervention in many neurological diseases is thwarted by the physical obstacle formed by the blood-brain barrier (BBB) that excludes most drugs from entering the brain from the blood. Thus, identifying efficacious modes of drug delivery to the brain remains a “holy grail” in molecular medicine and nanobiotechnology. Brain capillaries, that comprise the BBB, possess an endogenous receptor that ferries an iron-transport protein, termed p97 (melanotransferrin), across the BBB. Here, we explored the hypothesis that therapeutic drugs “piggybacked” as conjugates of p97 can be shuttled across the BBB for treatment of otherwise inoperable brain tumors. Approach Human p97 was covalently linked with the chemotherapeutic agents paclitaxel (PTAX) or adriamycin (ADR) and following intravenous injection, measured their penetration into brain tissue and other organs using radiolabeled and fluorescent derivatives of the drugs. In order to establish efficacy of the conjugates, we used nude mouse models to assess p97-drug conjugate activity towards glioma and mammary tumors growing subcutaneously compared to those growing intracranially. Principal Findings Bolus-injected p97-drug conjugates and unconjugated p97 traversed brain capillary endothelium within a few minutes and accumulated to 1–2% of the injected by 24 hours. Brain delivery with p97-drug conjugates was quantitatively 10 fold higher than with free drug controls. Furthermore, both free-ADR and p97-ADR conjugates equally inhibited the subcutaneous growth of gliomas growing outside the brain. Evocatively, only p97-ADR conjugates significantly prolonged the survival of animals bearing intracranial gliomas or mammary tumors when compared to similar cumulated doses of free-ADR. Significance This study provides the initial proof of concept for p97 as a carrier capable of shuttling therapeutic levels of drugs from the blood to the brain for the treatment of neurological disorders, including classes of resident and metastatic brain tumors. It may be prudent, therefore, to consider implementation of this novel delivery platform in various clinical settings for therapeutic intervention in acute and chronic neurological diseases.
Collapse
Affiliation(s)
| | - Cheryl Pfeifer
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| | - Timothy Z. Vitalis
- BioMarin Pharmaceutical Inc., Vancouver, Canada
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gavin Arthur
- BioMarin Pharmaceutical Inc., Vancouver, Canada
- * E-mail:
| | - Maki Ujiie
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| | - Qingqi Chen
- BioMarin Pharmaceutical Inc., Vancouver, Canada
| | - Sam Tsai
- BioMarin Pharmaceutical Inc., Vancouver, Canada
| | - Gerrasimo Koliatis
- BioMarin Pharmaceutical Inc., Vancouver, Canada
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reinhard Gabathuler
- BioMarin Pharmaceutical Inc., Vancouver, Canada
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wilfred A. Jefferies
- Department of Medical Genetics, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, the Michael Smith Laboratories and the Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
10
|
Michaud-Levesque J, Demeule M, Béliveau R. Plasminogen-dependent internalization of soluble melanotransferrin involves the low-density lipoprotein receptor-related protein and annexin II. Biol Chem 2007; 388:747-54. [PMID: 17570828 DOI: 10.1515/bc.2007.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractWe investigated the effect of plasminogen (Plg) on the internalization of recombinant soluble melanotransferrin (sMTf) using U87 human glioblastoma cells and murine embryonic fibroblasts (MEF) deficient in the low-density lipoprotein receptor-related protein (LRP). Using biospecific interaction analysis, both Glu- and Lys-Plg were shown to interact with immobilized sMTf. The binding of sMTf at the cell surface increased in the presence of both forms of Plg in control and in LRP-deficient MEF cells, whereas the uptake was strongly stimulated only by Lys-Plg in control MEF and U87 cells. In addition, in the presence of Lys-Plg, the internalization of sMTf was a saturable process, sensitive to temperature and dependent on the integrity of lysine residues. The addition of the receptor-associated protein, lactoferrin and aprotinin, as well as a monoclonal antibody (mAb) directed against LRP, inhibited the Lys-Plg-dependent uptake of sMTf. These results suggest an important role for LRP in this process. In addition, using binding and uptake assays in the presence of anti-annexin II mAb, we showed that annexin II might be responsible for the initial binding of sMTf in the presence of Plg. Our results suggest a Plg-mediated internalization mechanism for the clearance of sMTf via annexin II and LRP.
Collapse
Affiliation(s)
- Jonathan Michaud-Levesque
- Laboratoire de Médecine Moléculaire, Service d'Hémato-Oncologie, Hôpital Ste-Justine, Université du Québec à Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
11
|
Tang Y, Han T, Everts M, Zhu ZB, Gillespie GY, Curiel DT, Wu H. Directing adenovirus across the blood–brain barrier via melanotransferrin (P97) transcytosis pathway in an in vitro model. Gene Ther 2006; 14:523-32. [PMID: 17167498 DOI: 10.1038/sj.gt.3302888] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adenovirus serotype 5 (Ad5) is widely used in the development of gene therapy protocols. However, current gene therapy strategies involving brain are mostly based on intra-cranial injection. A major obstacle for systemically administered vectors to infect brain tissue is the blood-brain barrier (BBB). One strategy to cross the BBB is transcytosis, a transcellular transport process that shuttles a molecule from one side of the cell to the other side. Recently, melanotransferrin (MTf)/P97 was found to be able to cross the BBB and accumulate in brain. We thus hypothesize that re-directing Ad5 vectors to the MTf transcytosis pathway may facilitate Ad5 vectors to cross the BBB. To test this hypothesis, we constructed a bi-specific adaptor protein containing the extracellular domain of the coxsackie-adenovirus receptor (CAR) and the full-length melanotransferrin (sCAR-MTf), and investigated its ability to re-direct Ad5 vectors to the MTf transcytosis pathway. We found this adaptor protein could re-direct Ad5 to the MTf transcytosis pathway in an in vitro BBB model, and the transcytosed Ad5 viral particles retained their native infectivity. The sCAR-MTf-mediated Ad5 transcytosis was temperature- and dose dependent. In addition, we examined the directionality of sCAR-MTf-mediated Ad5 transcytosis, and found the efficiency of apical-to-basal transcytosis was much higher than that of basal-to-apical direction, supporting a role of this strategy in transporting Ad5 vectors towards the brain. Taken together, our study demonstrated that re-directing Ad5 to the MTf transcytosis pathway could facilitate gene delivery across the BBB.
Collapse
Affiliation(s)
- Y Tang
- Division of Human Gene Therapy, Department of Medicine, The Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Bertrand Y, Demeule M, Rivard GE, Béliveau R. Stimulation of tPA-dependent provisional extracellular fibrin matrix degradation by human recombinant soluble melanotransferrin. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:1024-30. [PMID: 16979249 DOI: 10.1016/j.bbamcr.2006.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2006] [Revised: 08/08/2006] [Accepted: 08/09/2006] [Indexed: 11/15/2022]
Abstract
Tissue-type plasminogen activator (tPA) and its substrate plasminogen (Plg) are key components in the fibrinolytic system. We have recently demonstrated, that truncated human recombinant soluble melanotransferrin (sMTf) could stimulate the activation of Plg by urokinase plasminogen activator and inhibit angiogenesis. Since various angiogenesis inhibitors were shown to stimulate tPA-mediated plasminogen activation, we examined the effects of sMTf on tPA-dependent fibrinolysis. This study demonstrated that sMTf enhanced tPA-activation of Plg by 6-fold. sMTf also increased the release of [125I]-fibrin fragments by tPA-activated plasmin. Moreover, we observed that the interaction of sMTf with Plg provoked a change in the fibrin clot structure by cleaving the fibrin alpha and beta chains. Overall, the present study shows that sMTf modulates tPA-dependent fibrinolysis by modifying the clot structure. These results also suggest that sMTf properties could involve enhanced dissolution of the provisional extracellular fibrin matrix.
Collapse
Affiliation(s)
- Y Bertrand
- Laboratoire de Médecine Moléculaire, Service d'Hématologie-Oncologie, Hôpital Ste-Justine-UQAM, C.P. 8888, Succursale Centre-ville, Montréal, Québec, Canada H3C 3P8
| | | | | | | |
Collapse
|
13
|
Gibbons R, Adeoya-Osiguwa SA, Fraser LR. A mouse sperm decapacitation factor receptor is phosphatidylethanolamine-binding protein 1. Reproduction 2005; 130:497-508. [PMID: 16183867 DOI: 10.1530/rep.1.00792] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Capacitation is a pivotal event for mammalian spermatozoa, involving the loss of surface proteins known as decapacitation factors (DF) and consequent acquisition of fertilizing ability. Earlier studies showed that a mouse sperm DF binds to a receptor, DF-R, whose attachment to the sperm plasma membrane appears to involve a glycosylphosphatidylinositol (GPI) anchor. In the present study, purification and subsequent sequencing of DF-R has identified this ~23 kDa protein as phosphatidyletha-nolamine-binding protein 1 (PEBP 1). To obtain functional evidence that supports sequence homology data, purified recombinant PEBP 1 and PEBP 2 were evaluated for biological activity. While PEBP 1 was able to remove DF activity in solution at concentrations above ~1 nmol/l, PEBP 2 was ineffective, even at 600 nmol/l; this confirmed that DF-R is PEBP 1. Anti-PEBP 1 antiserum recognized recombinant PEBP 1 and a ~23 kDa protein in both mouse and human sperm lysates. Immunolocalization studies revealed that DF-R/PEBP 1 is located on the acrosomal cap, the post-acrosomal region and the flagellum of both mouse and human spermatozoa, with epitope accessibility being capacitation state-dependent and reversible. Treatment of cells with a phospholipase able to cleave GPI anchors essentially abolished immunostaining, thus confirming the extracellular location of DF-R/PEBP 1. We suggest that DF-R/PEBP 1 plays its fundamental role in capacitation by causing alterations in the sperm plasma membrane in both head and flagellum, with functional consequences for membrane-associated proteins. Obtaining more detail about DF ↔ DF-R interactions could lead to useful applications in both fertility treatments and new contraceptive approaches.
Collapse
Affiliation(s)
- Rachel Gibbons
- Reproduction and Rhythms Group, School of Biomedical Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | | | | |
Collapse
|
14
|
Michaud-Levesque J, Demeule M, Béliveau R. Stimulation of cell surface plasminogen activation by membrane-bound melanotransferrin: a key phenomenon for cell invasion. Exp Cell Res 2005; 308:479-90. [PMID: 15936756 DOI: 10.1016/j.yexcr.2005.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Revised: 04/11/2005] [Accepted: 05/05/2005] [Indexed: 10/25/2022]
Abstract
The activation of plasminogen at the cell surface is a crucial step in cell migration and invasion. In the present study, the effect of membrane-bound melanotransferrin (mMTf), also known as human melanoma antigen p97, on cell surface plasminogen binding and activation was investigated by using Chinese Hamster Ovary (CHO) cells transfected with full-length melanotransferrin (MTf) cDNA and SK-MeL-28 melanoma cells. The expression of mMTf in CHO increased cell surface plasminogen binding by about 2-fold. In addition, application of the monoclonal antibody L235 against MTf as well as truncated, soluble MTf (sMTf) abolished plasminogen binding to MTf-transfected and SK-MeL-28 cells, indicating that mMTf is a potential cell surface plasminogen receptor. Moreover, mMTf expression in CHO cells stimulates plasminogen activation at the cell surface by about 2.5-fold. In addition to the induced binding and activation of plasminogen, cell motility, migration and invasion were about 3-fold higher in CHO cells expressing mMTf. Both monoclonal antibody L235 and truncated sMTf inhibited mMTf-stimulated CHO cell motility, migration and invasion. Overall, our results indicate a key role for mMTf in cell surface plasminogen binding and in activation processes involved during cell migration and invasion.
Collapse
Affiliation(s)
- Jonathan Michaud-Levesque
- Laboratoire de Médecine Moléculaire, Service d'Hémato-Oncologie, Hôpital Ste-Justine-Université du Québec à Montréal (UQAM), C.P. 8888, Succursale Centre-ville, Montréal, Québec, Canada H3C 3P8
| | | | | |
Collapse
|
15
|
Raggo C, Ruhl R, McAllister S, Koon H, Dezube BJ, Früh K, Moses AV. Novel cellular genes essential for transformation of endothelial cells by Kaposi's sarcoma-associated herpesvirus. Cancer Res 2005; 65:5084-95. [PMID: 15958552 DOI: 10.1158/0008-5472.can-04-2822] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is involved in the development of lymphoproliferative diseases and Kaposi's sarcoma. The oncogenicity of this virus is reflected in vitro by its ability to transform B cells and endothelial cells. Infection of dermal microvascular endothelial cells (DMVEC) transforms the cells from a cobblestone-like monolayer to foci-forming spindle cells. This transformation is accompanied by dramatic changes in the cellular transcriptome. Known oncogenes, such as c-Kit, are among the KSHV-induced host genes. We previously showed that c-Kit is an essential cellular component of the KSHV-mediated transformation of DMVEC. Here, we test the hypothesis that the transformation process can be used to discover novel oncogenes. When expression of a panel of KSHV-induced cellular transcripts was inhibited with antisense oligomers, we observed inhibition of DMVEC proliferation and foci formation using antisense molecules to RDC1 and Neuritin. We further showed that transformation of KSHV-infected DMVEC was inhibited by small interfering RNA directed at RDC1 or Neuritin. Ectopic expression of Neuritin in NIH 3T3 cells resulted in changes in cell morphology and anchorage-independent growth, whereas RDC1 ectopic expression significantly increased cell proliferation. In addition, both RDC1- and Neuritin-expressing cells formed tumors in nude mice. RDC1 is an orphan G protein-coupled receptor, whereas Neuritin is a growth-promoting protein known to mediate neurite outgrowth. Neither gene has been previously implicated in tumorigenesis. Our data suggest that KSHV-mediated transformation involves exploitation of the hitherto unrealized oncogenic properties of RDC1 and Neuritin.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Cell Transformation, Viral/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Endothelial Cells/cytology
- Endothelial Cells/virology
- GPI-Linked Proteins
- Gene Expression Profiling
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/physiology
- Humans
- LIM Domain Proteins
- Metalloproteins/biosynthesis
- Metalloproteins/genetics
- Mice
- Mice, Nude
- NIH 3T3 Cells
- Neuropeptides/biosynthesis
- Neuropeptides/genetics
- Oligonucleotide Array Sequence Analysis
- Oligonucleotides, Antisense/genetics
- Oncogenes/physiology
- Osteopontin
- Proto-Oncogene Proteins
- RNA, Small Interfering/genetics
- Receptors, CXCR
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, G-Protein-Coupled/genetics
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/virology
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/genetics
Collapse
Affiliation(s)
- Camilo Raggo
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | | | | | | | | | | |
Collapse
|