Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ruben J. Boado is active.

Publication


Featured researches published by Ruben J. Boado.


Clinical Cancer Research | 2004

Intravenous RNA Interference Gene Therapy Targeting the Human Epidermal Growth Factor Receptor Prolongs Survival in Intracranial Brain Cancer

Yun Zhang; Yufeng Zhang; Joshua A. Bryant; Andrew Charles; Ruben J. Boado; William M. Pardridge

Purpose: The human epidermal growth factor receptor (EGFR) plays an oncogenic role in solid cancer, including brain cancer. The present study was designed to prolong survival in mice with intracranial human brain cancer with the weekly i.v. injection of nonviral gene therapy causing RNA interference (RNAi) of EGFR gene expression. Experimental Design: Human U87 gliomas were implanted in the brain of adult scid mice, and weekly i.v. gene therapy was started at day 5 after implantation of 500,000 cells. An expression plasmid encoding a short hairpin RNA directed at nucleotides 2529–2557 within the human EGFR mRNA was encapsulated in pegylated immunoliposomes. The pegylated immunoliposome was targeted to brain cancer with 2 receptor-specific monoclonal antibodies (MAb), the murine 83–14 MAb to the human insulin receptor and the rat 8D3 MAb to the mouse transferrin receptor. Results: In cultured glioma cells, the delivery of the RNAi expression plasmid resulted in a 95% suppression of EGFR function, based on measurement of thymidine incorporation or intracellular calcium signaling. Weekly i.v. RNAi gene therapy caused reduced tumor expression of immunoreactive EGFR and an 88% increase in survival time of mice with advanced intracranial brain cancer. Conclusions: Weekly i.v. nonviral RNAi gene therapy directed against the human EGFR is a new therapeutic approach to silencing oncogenic genes in solid cancers. This is enabled with a nonviral gene transfer technology that delivers liposome-encapsulated plasmid DNA across cellular barriers with receptor-specific targeting ligands.


Human Gene Therapy | 2003

Intravenous nonviral gene therapy causes normalization of striatal tyrosine hydroxylase and reversal of motor impairment in experimental parkinsonism.

Yun Zhang; Frédéric Calon; Chunni Zhu; Ruben J. Boado; William M. Pardridge

Brain gene-targeting technology is used to reversibly normalize tyrosine hydroxylase (TH) activity in the striatum of adult rats, using the experimental 6-hydroxydopamine model of Parkinsons disease. The TH expression plasmid is encapsulated inside an 85-nm PEGylated immunoliposome (PIL) that is targeted with either the OX26 murine monoclonal antibody (MAb) to the rat transferrin receptor (TfR) or with the mouse IgG2a isotype control antibody. TfRMAb-PIL, or mIgG2a-PIL, is injected intravenously at a dose of 10 microg of plasmid DNA per rat. TfRMAb-PIL, but not mIgG2a-PIL, enters the brain via the transvascular route. The targeting TfRMAb enables the nanocontainer carrying the gene to undergo both receptor-mediated transcytosis across the blood-brain barrier (BBB) and receptor-mediated endocytosis into neurons behind the BBB by accessing the TfR. With this approach, the striatal TH activity ipsilateral to the intracerebral injection of the neurotoxin was normalized and increased from 738 +/- 179 to 5486 +/- 899 pmol/hr per milligram of protein. The TH enzyme activity measurements were corroborated by TH immunocytochemistry, which showed that the entire striatum was immunoreactive for TH after intravenous gene therapy. The normalization of striatal biochemistry was associated with a reversal of apomorphine-induced rotation behavior. Lesioned animals treated with the apomorphine exhibited 20 +/- 5 and 6 +/- 2 rotations/min, respectively, after intravenous administration of the TH plasmid encapsulated in mIgG2a-PIL and TfRMAb-PIL. These studies demonstrate that it is possible to normalize brain enzyme activity by intravenous administration and nonviral gene transfer.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Brain-specific expression of an exogenous gene after i.v. administration

Ningya Shi; Yun Zhang; Chunni Zhu; Ruben J. Boado; William M. Pardridge

The treatment of brain diseases with gene therapy requires the gene to be expressed throughout the central nervous system, and this is possible by using gene targeting technology that delivers the gene across the blood–brain barrier after i.v. administration of a nonviral formulation of the gene. The plasmid DNA is targeted to brain with pegylated immunoliposomes (PILs) using a targeting ligand such as a peptidomimetic mAb, which binds to a transporting receptor on the blood–brain barrier. The present studies adapt the PIL gene targeting technology to the mouse by using the rat 8D3 mAb to the mouse transferrin receptor. Tissue-specific expression in brain and peripheral organs of different exogenous genes (β-galactosidase, luciferase) is examined at 1–3 days after i.v. injection in adult mice of the exogenous gene packaged in the interior of 8D3-PIL. The expression plasmid is driven either by a broadly expressed promoter, simian virus 40, or by a brain-specific promoter taken from the 5′ flanking sequence of the human glial fibrillary acidic protein (GFAP) gene. The transgene is expressed in both brain and peripheral tissues when the simian virus 40 promoter is used, but the expression of the exogenous gene is confined to the brain when the transgene is under the influence of the brain-specific GFAP promoter. Confocal microscopy colocalizes immunoreactive bacterial β-galactosidase with immunoreactive GFAP in brain astrocytes. These studies indicate that tissue-specific gene expression in brain is possible after the i.v. administration of a nonviral vector with the combined use of gene targeting technology and tissue-specific gene promoters.


Pharmaceutical Research | 2000

Transport Across the Primate Blood-Brain Barrier of a Genetically Engineered Chimeric Monoclonal Antibody to the Human Insulin Receptor

M. Josephina Coloma; Hwa Jeong Lee; Atsushi Kurihara; Elliot M. Landaw; Ruben J. Boado; Sherie L. Morrison; William M. Pardridge

AbstractPurpose. Brain drug targeting may be achieved by conjugating drugs,that normally do not cross the blood-brain barrier (BBB), to brain drugdelivery vectors. The murine 83-14 MAb to the human insulin receptor(HIR) is a potential brain drug targeting vector that could be used inhumans, if this MAb was genetically engineered to form a chimericantibody, where most of the immunogenic murine sequences arereplaced by human antibody sequence. Methods. The present studies describe the production of the gene forthe chimeric HIRMAb, expression and characterization of the protein,radiolabeling of the chimeric HIRMAb with 111-indium and125-iodine, and quantitative autoradiography of living primate brain taken 2hours after intravenous administration of the [111In]chimeric HIRMAb. Results. The chimeric HIRMAb had identical affinity to the targetantigen as the murine HIRMAb based on Western blotting andimmunoradiometric assay using partially purified HIR affinity purified fromserum free conditioned media produced by a CHO cell line secretingsoluble HIR. The [125I]chimeric HIRMAb was avidly bound to isolatedhuman brain capillaries, and this binding was blocked by the murineHIRMAb. The [111In]chimeric HIRMAb was administeredintravenously to an anesthetized Rhesus monkey, and the 2 hour brain scanshowed robust uptake of the chimeric antibody by the living primatebrain. Conclusions. A genetically engineered chimeric HIRMAb has beenproduced, and the chimeric antibody has identical reactivity to thehuman and primate BBB HIR as the original murine antibody. Thischimeric HIRMAb may be used in humans for drug targeting throughthe BBB of neurodiagnostic or neurotherapeutic drugs that normallydo not cross the BBB.


Pharmaceutical Research | 2001

Receptor-mediated gene targeting to tissues in vivo following intravenous administration of pegylated immunoliposomes

Ningya Shi; Ruben J. Boado; William M. Pardridge

AbstractPurpose. Gene therapy has been limited by the immunogenicity of viral vectors, by the inefficiency of cationic liposomes, and by the rapid degradation in vivofollowing the injection of naked DNA. The present work describes a new approach that enables the non-invasive, non-viral gene therapy of the brain and peripheral organs following an intravenous injection. Methods. The plasmid DNA encoding β-galactosidase is packaged in the interior of neutral liposomes, which are stabilized for in vivo use by surface conjugation with polyethyleglycol (PEG). The tips of about 1% of the PEG strands are attached to a targeting monoclonal antibody (MAb), which acts as a “molecular Trojan Horse” to ferry the liposome carrying the gene across the biological barriers of the brain and other organs. The MAb targets the transferrin receptor, which is enriched at both the blood-brain barrier (BBB), and in peripheral tissues, such as liver and spleen. Results. Expression of the exogenous gene in brain, liver, and spleen was demonstrated with β-galactosidase histochemistry, which showed persistence of gene expression for at least 6 days after a single intravenous injection of the pegylated immunoliposomes. The persistence of the transgene was confirmed by Southern blot analysis. Conclusions. Widespread expression of an exogenous gene in brain and peripheral tissues is induced with a single intravenous administration of plasmid DNA packaged in the interior of pegylated im- munoliposomes. The liposomes are formulated to target specific receptor systems that enable receptor-mediated endocytosis of the complex into cells in vivo. This approach allows for non-invasive, non-viral gene therapy of the brain.


Diabetes | 1995

Upregulation of Blood-Brain Barrier GLUT1 Glucose Transporter Protein and mRNA in Experimental Chronic Hypoglycemia

Arno K. Kumagai; Young Sook Kang; Ruben J. Boado; William M. Pardridge

An in vivo model of chronic hypoglycemia was used to investigate changes in blood-brain barrier(BBB) glucose transport activity and changes in the expression of GLUT1 mRNA and protein in brain microvasculature occurring as an adaptive response to low circulating glucose levels. Chronic hypoglycemia was induced in rats by constant infusion of insulin via osmotic minipumps; control animals received infusions of saline. The criterion for chronic hypoglycemia was an average blood glucose concentration of <2.3 mmol/l (42 mg/dl) after 5 days. The average blood glucose concentration at the end of the experimental period in the rats selected for study was 2.0 ± 0.1 mmol/l (36 ± 1 mg/dl) vs. 4.9 ± 0.1 mmol/l (88 ± 1 mg/dl) in the controls. Internal carotid artery perfusion studies demonstrated an increase in the BBB permeability-surface area (PS) product of 40% (P < 0.0005) in the chronically hypoglycemic animals as compared with controls. Western blotting of solubilized isolated brain capillaries demonstrated a 51% increase (P < 0.05) in immunoreactive BBB GLUT1 in the chronically hypoglycemic rats, and Northern blotting of whole-brain poly(A+) mRNA revealed a 50% increase in the GLUT1-to-actin ratio in the insulin-treated group (P < 0.05). Northern blotting analysis of microvessel-depleted total brain poly(A+) showed that the increase in GLUT1 mRNA in the chronically hypoglycemic rats was restricted to the BBB. The present study demonstrates increased expression of GLUT1 mRNA and protein at the BBB in chronic hypoglycemia and suggests that this increase is responsible for the compensatory increase in BBB glucose transport activity that occurs with chronically low circulating blood glucose levels.


Journal of Cerebral Blood Flow and Metabolism | 2001

Blood—Brain Barrier Genomics

Jian Yi Li; Ruben J. Boado; William M. Pardridge

The blood–brain barrier (BBB) is formed by the brain microvascular endothelium, and the unique transport properties of the BBB are derived from tissue-specific gene expression within this cell. The current studies developed a gene microarray approach specific for the BBB by purifying the initial mRNA from isolated rat brain capillaries to generate tester cDNA. A polymerase chain reaction–based subtraction cloning method, suppression subtractive hybridization (SSH), was used, and the BBB cDNA was subtracted with driver cDNA produced from mRNA isolated from rat liver and kidney. Screening 5% of the subtracted tester cDNA resulted in identification of 50 gene products and more than 80% of those were selectively expressed at the BBB; these included novel gene sequences not found in existing databases, ESTs, and known genes that were not known to be selectively expressed at the BBB. Genes in the latter category include tissue plasminogen activator, insulin-like growth factor-2, PC-3 gene product, myelin basic protein, regulator of G protein signaling 5, utrophin, IκB, connexin-45, the class I major histocompatibility complex, the rat homologue of the transcription factors hbrm or EZH1, and organic anion transporting polypeptide type 2. Knowledge of tissue-specific gene expression at the BBB could lead to new targets for brain drug delivery and could elucidate mechanisms of brain pathology at the microvascular level.


Journal of Cerebral Blood Flow and Metabolism | 2013

Immunologic privilege in the central nervous system and the blood-brain barrier.

Leslie L. Muldoon; Jorge Ivan Alvarez; David J. Begley; Ruben J. Boado; Gregory J. del Zoppo; Nancy D. Doolittle; Britta Engelhardt; John M. Hallenbeck; Russell R. Lonser; John R. Ohlfest; Alexandre Prat; Maurizio Scarpa; Richard J. Smeyne; Lester R. Drewes; Edward A. Neuwelt

The brain is in many ways an immunologically and pharmacologically privileged site. The blood–brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.


Journal of Gene Medicine | 2003

In vivo knockdown of gene expression in brain cancer with intravenous RNAi in adult rats

Yun Zhang; Ruben J. Boado; William M. Pardridge

Gene silencing in mammalian cells is possible with RNA interference (RNAi) with expression plasmids that encode for short hairpin RNAs (shRNA) that hybridize to a specific sequence within a target mRNA. The limiting factor in developing RNAi therapeutics in mammals is the gene delivery system.


Molecular Brain Research | 1994

Gene expression of GLUT3 and GLUT1 glucose transporters in human brain tumors

Ruben J. Boado; Keith L. Black; William M. Pardridge

GLUT3 glucose transporter gene expression is confined to neurons, while GLUT1 gene expression is limited to endothelial cells in normal brain. Thus far, neither of the GLUT genes has been shown to be consistently expressed in glial cells in adult brain in vivo under normal conditions. However, GLUT gene expression may be aberrant in human brain glial tumors. The present investigation shows that the GLUT1 and GLUT3 transcripts are differentially expressed in a series of 20 human brain tumors. The GLUT1/actin mRNA ratio increased in parallel to the astrocytoma grade, compared to a control human brain cortex, although no change in this ratio was seen in 5 meningiomas. Immunoreactive GLUT1 protein was not detectable in human brain tumors, including high-grade gliomas. Both 4.2 or 2.7 kb GLUT3/actin mRNA ratios showed a linear correlation with the glioma grade (P < 0.025), and the GLUT3-immunoreactive protein was also expressed in high grade gliomas. These studies provide evidence for induction of GLUT1 and GLUT3 gene expression in malignant glial cells, and the mRNA levels correlate with the biologic aggressiveness of the tumor. The detection of immunoreactive GLUT3, but not GLUT1, in the high grade gliomas suggest the GLUT3 isoform may be the predominant glucose transporter in highly malignant glial cells of human brain.

Collaboration


Dive into the Ruben J. Boado's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yun Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Jian Yi Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Qing-Hui Zhou

University of California

View shared research outputs
Top Co-Authors

Avatar

Yufeng Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Rachita K. Sumbria

Keck Graduate Institute of Applied Life Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chun-Fang Xia

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuntao Wang

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge