Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Front Behav Neurosci ; 15: 661973, 2021.
Article in English | MEDLINE | ID: mdl-34366802

ABSTRACT

Historically, many investigations into neurodegenerative diseases have focused on alterations in specific neuronal populations such as, for example, the loss of midbrain dopaminergic neurons in Parkinson's disease (PD) and loss of cholinergic transmission in Alzheimer's disease (AD). However, it has become increasingly clear that mammalian brain activities, from executive and motor functioning to memory and emotional responses, are strictly regulated by the integrity of multiple interdependent neuronal circuits. Among subcortical structures, the dopaminergic nigrostriatal and mesolimbic pathways as well as cholinergic innervation from basal forebrain and brainstem, play pivotal roles in orchestrating cognitive and non-cognitive symptoms in PD and AD. Understanding the functional interactions of these circuits and the consequent neurological changes that occur during degeneration provides new opportunities to understand the fundamental inter-workings of the human brain as well as develop new potential treatments for patients with dysfunctional neuronal circuits. Here, excerpted from a session of the European Behavioral Pharmacology Society meeting (Braga, Portugal, August 2019), we provide an update on our recent work in behavioral and cellular neuroscience that primarily focuses on interactions between cholinergic and dopaminergic systems in PD models, as well as stress in AD. These brief discussions include descriptions of (1) striatal cholinergic interneurons (CINs) and PD, (2) dopaminergic and cholinergic modulation of impulse control, and (3) the use of an implantable cell-based system for drug delivery directly the into brain and (4) the mechanisms through which day life stress, a risk factor for AD, damage protein and RNA homeostasis leading to AD neuronal malfunction.

2.
Neural Plast ; 2020: 8814028, 2020.
Article in English | MEDLINE | ID: mdl-33293946

ABSTRACT

Neuroinflammation and cytokine-dependent neurotoxicity appear to be major contributors to the neuropathology in Parkinson's disease (PD). While pharmacological advancements have been a mainstay in the treatment of PD for decades, it is becoming increasingly clear that nonpharmacological approaches including traditional and nontraditional forms of exercise and physical rehabilitation can be critical adjunctive or even primary treatment avenues. Here, we provide an overview of preclinical and clinical research detailing the biological role of proinflammatory molecules in PD and how motor rehabilitation can be used to therapeutically modulate neuroinflammation, restore neural plasticity, and improve motor function in PD.


Subject(s)
Cytokines/metabolism , Exercise Therapy , Exercise/physiology , Nerve Growth Factors/metabolism , Parkinson Disease/rehabilitation , Exercise Therapy/methods , Humans , Neuronal Plasticity/physiology , Parkinson Disease/physiopathology
3.
Curr Res Pharmacol Drug Discov ; 1: 19-29, 2020 Apr.
Article in English | MEDLINE | ID: mdl-34909639

ABSTRACT

Delivering glial cell line-derived neurotrophic factor (GDNF) to the brain is a potential treatment for Parkinson's Disease (PD). Here we use an implantable encapsulated cell technology that uses modified human clonal ARPE-19 â€‹cells to deliver of GDNF to the brain. In vivo studies demonstrated sustained delivery of GDNF to the rat striatum over 6 months. Anatomical benefits and behavioral efficacy were shown in 6-OHDA lesioned rats where nigral dopaminergic neurons were preserved in neuroprotection studies and dopaminergic fibers were restored in neurorecovery studies. When larger, clinical-sized devices were implanted for 3 months into the putamen of Göttingen minipigs, GDNF was widely distributed throughout the putamen and caudate producing a significant upregulation of tyrosine hydroxylase immunohistochemistry. These results are the first to provide clear evidence that implantation of encapsulated GDNF-secreting cells deliver efficacious and biologically relevant amounts of GDNF in a sustained and targeted manner that is scalable to treat the large putamen in patients with Parkinson's disease.

4.
Neural Plast ; 2019: 6286197, 2019.
Article in English | MEDLINE | ID: mdl-30984255

ABSTRACT

Methods: Human ARPE-19 cells engineered to secrete high levels of the glial cell line-derived neurotrophic factor (GDNF) were encapsulated into hollow fiber membranes. The devices were implanted into the rat striatum 1 week prior to striatal quinolinic acid injections. Animals were evaluated using a battery of validated motor tests, and histology was performed to determine the extent of GDNF diffusion and associated prevention of neuronal cell loss and behavioral deficits. Results: Encapsulated cell-based delivery of GDNF produced widespread distribution of GDNF throughout the entire implanted striatum. Stereological estimates of striatal neuron number and volume of lesion size revealed that GDNF delivery resulted in near complete neuroprotection. Conclusions: Delivery of neurotrophic molecules such as GDNF using encapsulated cells has reached a technological point where clinical evaluation is justified. Because GDNF has been effective in animal models of Parkinson's disease, stroke, epilepsy, and Huntington's disease, among other debilitating neurodegenerative diseases, encapsulated cell-based delivery of GDNF might represent one innovative means of slowing the neural degeneration seen in a myriad of currently untreatable neurological diseases.


Subject(s)
Corpus Striatum/drug effects , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Quinolinic Acid/toxicity , Animals , Cell Encapsulation , Cell Line , Drug Delivery Systems , Humans , LLC-PK1 Cells , Male , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Rats, Sprague-Dawley , Swine
5.
J Neurosci ; 39(11): 2144-2156, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30665947

ABSTRACT

Neurotrophic factors are candidates for treating epilepsy, but their development has been hampered by difficulties in achieving stable and targeted delivery of efficacious concentrations within the desired brain region. We have developed an encapsulated cell technology that overcomes these obstacles by providing a targeted, continuous, de novo synthesized source of high levels of neurotrophic molecules from human clonal ARPE-19 cells encapsulated into hollow fiber membranes. Here we illustrate the potential of this approach for delivering glial cell line-derived neurotrophic factor (GDNF) directly to the hippocampus of epileptic rats. In vivo studies demonstrated that bilateral intrahippocampal implants continued to secrete GDNF that produced high hippocampal GDNF tissue levels in a long-term manner. Identical implants robustly reduced seizure frequency in the pilocarpine model. Seizures were reduced rapidly, and this effect increased in magnitude over 3 months, ultimately leading to a reduction of seizures by 93%. This effect persisted even after device removal, suggesting potential disease-modifying benefits. Importantly, seizure reduction was associated with normalized changes in anxiety and improved cognitive performance. Immunohistochemical analyses revealed that the neurological benefits of GDNF were associated with the normalization of anatomical alterations accompanying chronic epilepsy, including hippocampal atrophy, cell degeneration, loss of parvalbumin-positive interneurons, and abnormal neurogenesis. These effects were associated with the activation of GDNF receptors. All in all, these results support the concept that the implantation of encapsulated GDNF-secreting cells can deliver GDNF in a sustained, targeted, and efficacious manner, paving the way for continuing preclinical evaluation and eventual clinical translation of this approach for epilepsy.SIGNIFICANCE STATEMENT Epilepsy is one of the most common neurological conditions, affecting millions of individuals of all ages. These patients experience debilitating seizures that frequently increase over time and can associate with significant cognitive decline and psychiatric disorders that are generally poorly controlled by pharmacotherapy. We have developed a clinically validated, implantable cell encapsulation system that delivers high and consistent levels of GDNF directly to the brain. In epileptic animals, this system produced a progressive and permanent reduction (>90%) in seizure frequency. These benefits were accompanied by improvements in cognitive and anxiolytic behavior and the normalization of changes in CNS anatomy that underlie chronic epilepsy. Together, these data suggest a novel means of tackling the frequently intractable neurological consequences of this devastating disorder.


Subject(s)
Epilepsy/drug therapy , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Seizures/drug therapy , Animals , Cell Encapsulation , Cell Line , Drug Delivery Systems/methods , Epilepsy/chemically induced , Humans , Male , Pilocarpine/administration & dosage , Rats, Sprague-Dawley , Seizures/chemically induced
6.
Prog Retin Eye Res ; 68: 67-82, 2019 01.
Article in English | MEDLINE | ID: mdl-30342088

ABSTRACT

Millions of people worldwide suffer from debilitating, progressive, and often permanent loss of vision without any viable treatment options. The complex physiological barriers of the eye contribute to the difficulty in developing novel therapies by limiting our ability to deliver therapeutics in a sustained and controlled manner; especially when attempting to deliver drugs to the posterior eye or trying to regenerate the diseased retina. Cell-based therapies offer a significant potential advancement in these situations. In particular, encapsulating, or immunoisolating, cells within implantable, semi-permeable membranes has emerged as a clinically viable means of delivering therapeutic molecules to the eye for indefinite periods of time. The optimization of encapsulation device designs is occurring together with refinements in biomaterials, genetic engineering, and stem-cell production, yielding, for the first time, the possibility of widespread therapeutic use of this technology. Here, we highlight the status of the most advanced and widely explored iteration of cell encapsulation with an eye toward translating the potential of this technological approach to the medical reality.


Subject(s)
Biological Products/administration & dosage , Cell- and Tissue-Based Therapy/methods , Drug Delivery Systems , Eye Diseases/therapy , Polymers/administration & dosage , Humans
7.
Mol Ther Methods Clin Dev ; 9: 211-224, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29766029

ABSTRACT

Brain-derived neurotrophic factor (BDNF) may represent a therapeutic for chronic epilepsy, but evaluating its potential is complicated by difficulties in its delivery to the brain. Here, we describe the effects on epileptic seizures of encapsulated cell biodelivery (ECB) devices filled with genetically modified human cells engineered to release BDNF. These devices, implanted into the hippocampus of pilocarpine-treated rats, highly decreased the frequency of spontaneous seizures by more than 80%. These benefits were associated with improved cognitive performance, as epileptic rats treated with BDNF performed significantly better on a novel object recognition test. Importantly, long-term BDNF delivery did not alter normal behaviors such as general activity or sleep/wake patterns. Detailed immunohistochemical analyses revealed that the neurological benefits of BDNF were associated with several anatomical changes, including reduction in degenerating cells and normalization of hippocampal volume, neuronal counts (including parvalbumin-positive interneurons), and neurogenesis. In conclusion, the present data suggest that BDNF, when continuously released in the epileptic hippocampus, reduces the frequency of generalized seizures, improves cognitive performance, and reverts many histological alterations associated with chronic epilepsy. Thus, ECB device-mediated long-term supplementation of BDNF in the epileptic tissue may represent a valid therapeutic strategy against epilepsy and some of its co-morbidities.

8.
J Comp Neurol ; 525(3): 553-573, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27490949

ABSTRACT

Central cholinergic structures within the brain of the even-toed hoofed Goettingen miniature domestic pig (Sus scrofa domesticus) were evaluated by immunohistochemical visualization of choline acetyltransferase (ChAT) and the low-affinity neurotrophin receptor, p75NTR . ChAT-immunoreactive (-ir) perikarya were seen in the olfactory tubercle, striatum, medial septal nucleus, vertical and horizontal limbs of the diagonal band of Broca, and the nucleus basalis of Meynert, medial habenular nucleus, zona incerta, neurosecretory arcuate nucleus, cranial motor nuclei III and IV, Edinger-Westphal nucleus, parabigeminal nucleus, pedunculopontine nucleus, and laterodorsal tegmental nucleus. Cholinergic ChAT-ir neurons were also found within transitional cortical areas (insular, cingulate, and piriform cortices) and hippocampus proper. ChAT-ir fibers were seen throughout the dentate gyrus and hippocampus, in the mediodorsal, laterodorsal, anteroventral, and parateanial thalamic nuclei, the fasciculus retroflexus of Meynert, basolateral and basomedial amygdaloid nuclei, anterior pretectal and interpeduncular nuclei, as well as select laminae of the superior colliculus. Double immunofluorescence demonstrated that virtually all ChAT-ir basal forebrain neurons were also p75NTR -positive. The present findings indicate that the central cholinergic system in the miniature pig is similar to other mammalian species. Therefore, the miniature pig may be an appropriate animal model for preclinical studies of neurodegenerative diseases where the cholinergic system is compromised. J. Comp. Neurol. 525:553-573, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/metabolism , Choline O-Acetyltransferase/metabolism , Receptors, Nerve Growth Factor/metabolism , Swine, Miniature/metabolism , Animals , Brain/anatomy & histology , Female , Immunohistochemistry , Models, Animal , Swine , Swine, Miniature/anatomy & histology
9.
Adv Drug Deliv Rev ; 67-68: 131-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23880505

ABSTRACT

Delivering therapeutic molecules, including trophic factor proteins, across the blood brain barrier to the brain parenchyma to treat chronic neurodegenerative diseases remains one of the great challenges in biology. To be effective, delivery needs to occur in a long-term and stable manner at sufficient quantities directly to the target region in a manner that is selective but yet covers enough of the target site to be efficacious. One promising approach uses cellular implants that produce and deliver therapeutic molecules directly to the brain region of interest. Implanted cells can be precisely positioned into the desired region and can be protected from host immunological attack by encapsulating them and by surrounding them within an immunoisolatory, semipermeable capsule. In this approach, cells are enclosed within a semiporous capsule with a perm selective membrane barrier that admits oxygen and required nutrients and releases bioactive cell secretions while restricting passage of larger cytotoxic agents from the host immune defense system. Recent advances in human cell line development have increased the levels of secreted therapeutic molecules from encapsulated cells, and membrane extrusion techniques have led to the first ever clinical demonstrations of long-term survival and function of encapsulated cells in the brain parenchyma. As such, cell encapsulation is capable of providing a targeted, continuous, de novo synthesized source of very high levels of therapeutic molecules that can be distributed over significant portions of the brain.


Subject(s)
Cell Transplantation/methods , Neurodegenerative Diseases/therapy , Alzheimer Disease/therapy , Animals , Cell Transplantation/instrumentation , Humans , Huntington Disease/therapy , Nerve Growth Factor/metabolism , Parkinson Disease/therapy , Tissue Scaffolds
10.
PLoS One ; 8(10): e78490, 2013.
Article in English | MEDLINE | ID: mdl-24205243

ABSTRACT

BACKGROUND: This study aims to create a convenient reference for both clinicians and researchers so that vis-à-vis comparisons between brain disorders can be made quickly and accurately. We report here the incidence and prevalence of the major adult-onset brain disorders in the United States using a meta-analysis approach. MATERIAL AND METHODS: Epidemiological figures were collected from the most recent, reliable data available in the research literature. Population statistics were based on the most recent census from the US Census Bureau. Extrapolations were made only when necessary. The most current epidemiological studies for each disorder were chosen. All effort was made to use studies based on national cohorts. Studies reviewed were conducted between 1950 and 2009. The data of the leading studies for several neurological studies was compiled in order to obtain the most accurate extrapolations. Results were compared to commonly accepted values in order to evaluate validity. RESULTS: It was found that 6.75% of the American adult population is afflicted with brain disorders. This number was eclipsed by the 8.02% of Floridians with brain disorders, which is due to the large aged population residing in the state. CONCLUSIONS: There was a noticeable lack of epidemiological data concerning adult-onset brain disorders. Since approximately 1 out of every 7 households is affected by brain disorders, increased research into this arena is warranted.


Subject(s)
Nervous System Diseases/epidemiology , Brain Diseases/epidemiology , Data Collection , Humans , Incidence , Prevalence , United States
11.
Restor Neurol Neurosci ; 30(3): 225-36, 2012.
Article in English | MEDLINE | ID: mdl-22426041

ABSTRACT

PURPOSE: Encapsulated cell (EC) biodelivery is a promising, clinically relevant technology platform to safely target the delivery of therapeutic proteins to the central nervous system. The purpose of this study was to evaluate EC biodelivery of the novel neurotrophic factor, Meteorin, to the striatum of rats and to investigate its neuroprotective effects against quinolinic acid (QA)-induced excitotoxicity. METHODS: Meteorin-producing ARPE-19 cells were loaded into EC biodelivery devices and implanted into the striatum of rats. Two weeks after implantation, QA was injected into the ipsilateral striatum followed by assessment of neurological performance two and four weeks after QA administration. RESULTS: Implant-delivered Meteorin effectively protected against QA-induced toxicity, as manifested by both near-normal neurological performance and reduction of brain cell death. Morphological analysis of the Meteorin-treated brains showed a markedly reduced striatal lesion size. The EC biodelivery devices produced stable or even increasing levels of Meteorin throughout the study over 6 weeks. CONCLUSIONS: Stereotactically implanted EC biodelivery devices releasing Meteorin could offer a feasible strategy in the treatment of neurological diseases with an excitotoxic component such as Huntington's disease. In a broader sense, the EC biodelivery technology is a promising therapeutic protein delivery platform for the treatment of a wide range of diseases of the central nervous system.


Subject(s)
Absorbable Implants/standards , Cytoprotection/drug effects , Disease Models, Animal , Huntington Disease/drug therapy , Nerve Tissue Proteins/administration & dosage , Quinolines/toxicity , Animals , Brain Tissue Transplantation/methods , Capsules/administration & dosage , Cell Line , Cytoprotection/physiology , Humans , Huntington Disease/chemically induced , Huntington Disease/genetics , Male , Mice , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Rats , Rats, Sprague-Dawley
12.
J Control Release ; 154(3): 249-57, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21704093

ABSTRACT

We previously engineered a macroporous, polymer-based vaccine that initially produces GM-CSF gradients to recruit local dendritic cells and subsequently presents CpG oligonucleotides, and tumor lysate to cell infiltrates to induce immune cell activation and immunity against tumor cells in peripheral tumor models. Here, we demonstrate that this system eradicates established intracranial glioma following implantation into brain tissue, whereas implantation in resection cavities obviates vaccine efficacy. Rats bearing seven-day old, intracranial glioma tumors were treated with PLG vaccines implanted into the tumor bed, resulting in retention of contralateral forelimb function (day 17) that is compromised by tumor formation in control animals, and 90% long-term survival (>100 days). Similar benefits were observed in animals receiving tumor resection plus vaccine implants into the adjacent parenchyma, but direct implantation of PLG vaccines into the resection cavity conferred no benefit. This dissociation of efficacy was likely related to GM-CSF distribution, as implantation of PLG vaccines within brain tissue produced significant GM-CSF gradients for prolonged periods, which was not detected after implantation in resection cavities. These studies demonstrate that PLG vaccine efficacy is correlated to GM-CSF gradient formation, which requires direct implantation into brain tissue, and justify further exploration of this approach for glioma treatment.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Glioma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Polyglactin 910/chemistry , Adjuvants, Immunologic/therapeutic use , Animals , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Cancer Vaccines/therapeutic use , Glioma/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Male , Oligodeoxyribonucleotides/therapeutic use , Porosity , Prostheses and Implants , Rats , Rats, Sprague-Dawley
13.
Curr Stem Cell Res Ther ; 6(3): 171-89, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21476979

ABSTRACT

Current therapies have limited or no capacity to restore lost function, slow ongoing neurodegeneration, or promote regeneration following damage to the brain. Biomaterials are playing an increasingly important role in the development of novel, potentially efficacious approaches to brain treatment and repair. Programmable biomaterials enable and augment the targeted delivery of drugs into the brain and allow cell/tissue transplants to be effectively delivered and integrate into the brain, to serve as delivery vehicles for therapeutic proteins, and rebuild damaged circuits. Similarly, biomaterials are being increasingly used to recapitulate specific aspects of brain niches to promote regeneration and/or repair damaged neuronal pathways with stem cell therapies. Many of these approaches are gaining momentum because nanotechnology allows greater control over material-cell interactions that induce specific developmental processes and cellular responses including differentiation, migration, and outgrowth. This review discusses the state of the art and new directions in the convergence of biomaterial science, drug delivery, and stem cell biology in the treatment of degenerative and malignant brain diseases.


Subject(s)
Biocompatible Materials/therapeutic use , Brain Diseases/therapy , Neural Stem Cells/transplantation , Animals , Brain Diseases/epidemiology , Chronic Pain/therapy , Drug Delivery Systems , Humans , Liposomes , Magnetic Resonance Imaging/methods , Nanoparticles , Nerve Regeneration , Tissue Scaffolds
14.
Pharm Res ; 28(5): 1074-80, 2011 May.
Article in English | MEDLINE | ID: mdl-21225320

ABSTRACT

PURPOSE: The prognosis for glioma patients is poor, and development of new treatments is critical. Previously, we engineered polymer-based vaccines that control GM-CSF, CpG-oligonucleotide, and tumor-lysate presentation to regulate immune cell trafficking and activation, which promoted potent immune responses against peripheral tumors. Here, we extend the use of this system to glioma. METHODS: Rats were challenged with an intracranial injection of glioma cells followed (1 week) by administration of the polymeric vaccine (containing GM-CSF, CpG, and tumor-lysate) in the tumor bed. Control rats were treated with blank matrices, matrices with GM-CSF and CpG, or intra-tumoral bolus injections of GM-CSF, CpG, and tumor lysate. Rats were monitored for survival and tested for neurological function. RESULTS: Survival studies confirmed a benefit of the polymeric vaccine as 90% of vaccinated rats survived for > 100 days. Control rats exhibited minimal benefit. Motor tests revealed that vaccination protected against the loss of forelimb use produced by glioma growth. Histological analysis quantitatively confirmed a robust and rapid reduction in tumor size. Long-term immunity was confirmed when 67% of survivors also survived a second glioma challenge. CONCLUSIONS: These studies extend previous reports regarding this approach to tumor therapy and justify further development for glioma treatment.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Glioma/therapy , Immunotherapy , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Glioma/immunology , Glioma/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/therapeutic use , Rats , Rats, Sprague-Dawley
15.
Neurobiol Dis ; 41(1): 160-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20840868

ABSTRACT

Meteorin is a newly discovered secreted protein involved in both glial and neuronal cell differentiation, as well as in cerebral angiogenesis during development; but effects in the adult nervous system are unknown. The growth factor-like properties and expression of Meteorin during the development of the nervous system raises the possibility that it might possess important neuroprotective or regenerative capabilities. This report is the first demonstration that Meteorin has potent neuroprotective effects in vivo. Lentiviral-mediated striatal delivery of Meteorin to rats two weeks prior to injections of quinolinic acid (QA) dramatically reduced the loss of striatal neurons. The cellular protection afforded by Meteorin was associated with normalization of neurological performance on spontaneous forelimb placing and cylinder behavioral tests and a complete protection against QA-induced weight loss. These benefits were comparable in magnitude to those obtained with lentiviral-mediated delivery of ciliary neurotrophic factor (CNTF), a protein with known neuroprotective properties in the same model system. In naive animals, endogenous levels of both Meteorin and CNTF were increased in glial cells in response to QA lesion indicating that Meteorin may exert its protective effects as part of the reactive gliosis cascade in the injured brain. In summary, these data demonstrate that Meteorin strongly protects striatal neurons and deserves additional evaluation as a novel therapeutic for the treatment of neurological disorders with an excitotoxic component such as Huntington's Disease.


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , Huntington Disease/therapy , Lentivirus/genetics , Movement Disorders/therapy , Nerve Tissue Proteins/genetics , Neurons/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Corpus Striatum/pathology , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , HEK293 Cells , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Movement Disorders/genetics , Movement Disorders/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley
16.
Article in English | MEDLINE | ID: mdl-21158722

ABSTRACT

Treating malignant brain tumors is one of the most formidable challenges in oncology. Contemporary treatments are hampered, in large part, by limited drug delivery across the blood-brain barrier (BBB) to the tumor bed. Biomaterials are playing an increasingly important role in developing more effective brain tumor treatments. Specifically, polymer (nano)particles can provide prolonged drug delivery directly to the tumor following direct intracerebral injection, by making them physiochemically able to cross the BBB to the tumor, or by functionalizing the material surface with peptides and ligands allowing the drug-loaded material to be systemically administered but still specifically target the tumor cells or tumor endothelium. As such, biomaterials can serve as targeted delivery devices for both conventional and novel therapies including gene therapy, photodynamic therapy, anti-angiogenic and thermotherapy. Nanoparticles also have the potential to play key roles in the diagnosis and imaging of brain tumors by revolutionizing both preoperative and intraoperative brain tumor delineation, allowing early detection of pre-cancerous cells, and providing real-time, longitudinal, non-invasive monitoring/imaging of the effects of treatment. The continued translation of current research into clinical practice will rely on solving challenges relating to the pharmacology of nanoparticles but it is envisioned that novel biomaterials will ultimately allow clinicians to target tumors and introduce multiple, pharmaceutically relevant entities for simultaneous targeting, imaging, and therapy in a unique and unprecedented manner.

17.
Adv Exp Med Biol ; 670: 80-91, 2010.
Article in English | MEDLINE | ID: mdl-20384220

ABSTRACT

The choroid plexuses (CPs) play pivotal roles in basic aspects of neural function including maintaining the extracellular milieu of the brain by actively modulating chemical exchange between the CSF and brain parenchyma, surveying the chemical and immunological status of the brain, detoxifying the brain, secreting a nutritive "cocktail" of polypeptides and participating in repair processes following trauma. Even modest changes in the CP can have far reaching effects and changes in the anatomy and physiology of the CP have been linked to several CNS diseases. It is also possible that replacing diseased or transplanting healthy CP might be useful for treating acute and chronic brain diseases. Here we describe the wide-ranging functions of the CP, alterations of these functions in aging and neurodegeneration and recent demonstrations of the therapeutic potential of transplanted microencapsulated CP for neural trauma.


Subject(s)
Brain Tissue Transplantation , Brain/pathology , Brain/physiology , Choroid Plexus/cytology , Epithelial Cells/transplantation , Regeneration , Aging/physiology , Alginates/chemistry , Alginates/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Animals , Brain/cytology , Choroid Plexus/physiology , Disease Models, Animal , Drug Compounding , Epithelial Cells/cytology , Gene Expression Profiling , Humans , Huntington Disease/pathology , Huntington Disease/physiopathology , Huntington Disease/therapy , Microarray Analysis , Stroke/pathology , Stroke/physiopathology , Stroke/therapy , Transplantation, Heterologous
18.
Cell Transplant ; 19(9): 1063-71, 2010.
Article in English | MEDLINE | ID: mdl-20412616

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent proangiogenic peptide and its administration has been considered as a potential neuroprotective strategy following cerebral stroke. Because VEGF has a short half-life and limited access to the brain parenchyma following systemic administration, approaches are being developed to deliver it directly to the site of infarction. In the present study, VEGF was incorporated into a sustained release hydrogel delivery system to examine its potential benefits in a rat model of cerebral ischemia. The hydrogel loaded with VEGF (1 µg) was stereotaxically injected into the striatum of adult rats 15 min prior to a 1-h occlusion of the middle cerebral artery. Two days after surgery, animals were tested for motor function using the elevated bias swing test (EBST) and Bederson neurological battery. Control animals received either stroke alone, stroke plus injections of a blank gel, or a single bolus injection of VEGF (1 µg). Behavioral testing confirmed that the MCA occlusion resulted in significant deficits in the the EBST and Bederson tests. In contrast, the performance of animals receiving VEGF gels was significantly improved relative to controls, with only modest impairments observed. Cerebral infarction analyzed using 2,3,5-triphenyl-tetrazolium chloride staining confirmed that the VEGF gels significantly and potently reduced the lesion volume. No neurological or histological benefits were conferred by either blank gel or bolus VEGF injections. These data demonstrate that VEGF, delivered from a hydrogel directly to the brain, can induce significant functional and structural protection from ischemic damage in a rat model of stroke.


Subject(s)
Brain Ischemia/drug therapy , Hydrogels/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Brain Ischemia/pathology , Disease Models, Animal , Injections , Male , Rats , Rats, Sprague-Dawley , Stroke/drug therapy , Stroke/pathology
19.
Neurotox Res ; 17(1): 66-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19588214

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent peptide with well-documented pro-angiogenic effects. Recently, it has also become clear that exogenous administration of VEGF is neuroprotective in animal models of central nervous system diseases. In the present study, VEGF was incorporated into a sustained release hydrogel delivery system to examine its potential benefits in a rat model of Huntington's disease (HD). The VEGF-containing hydrogel was stereotaxically injected into the striatum of adult rats. Three days later, quinolinic acid (QA; 225 nmol) was injected into the ipsilateral striatum to produce neuronal loss and behavioral deficits that mimic those observed in HD. Two weeks after surgery, animals were tested for motor function using the placement and cylinder tests. Control animals received either QA alone or QA plus empty hydrogel implants. Behavioral testing confirmed that the QA lesion resulted in significant deficits in the ability of the control animals to use their contralateral forelimb. In contrast, the performance of those animals receiving VEGF was significantly improved relative to controls with only modest motor impairments observed. Stereological counts of NeuN-positive neurons throughout the striatum demonstrated that VEGF implants significantly protected against the loss of striatal neurons induced by QA. These data are the first to demonstrate that VEGF can be used to protect striatal neurons from excitotoxic damage in a rat model of HD.


Subject(s)
Huntington Disease/drug therapy , Hydrogels/administration & dosage , Neuroprotective Agents/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiology , Disease Models, Animal , Functional Laterality , Huntington Disease/chemically induced , Huntington Disease/physiopathology , Male , Neurons/drug effects , Neurons/metabolism , Phosphopyruvate Hydratase/metabolism , Psychomotor Performance/drug effects , Quinolinic Acid , Rats , Rats, Sprague-Dawley , Time Factors
20.
Expert Opin Biol Ther ; 9(12): 1453-61, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19916730

ABSTRACT

This perspective is part of an ongoing series of articles that describe the drugs that were dropped from clinical development during the previous year. This paper focuses on those drugs that were discontinued in 2008. The drugs represent a diverse collection of compounds targeted at diseases including cancer, diabetes, infections, arthritis, HIV, restenosis, allergies, anaemia, osteoporosis, wound healing, and growth deficiency. No consistent theme is apparent that accounts for the diversity of discontinued drugs. Still, this diversity, coupled with the numbers of novel drugs that were approved in 2008 suggests a healthy ongoing drug development process.


Subject(s)
Biological Products/therapeutic use , Drug Discovery , Animals , Anti-Infective Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Antirheumatic Agents/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Treatment Failure , Vaccines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...