Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Mol Sci ; 25(7)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38612514

ABSTRACT

Alzheimer's disease (AD) is the fifth leading cause of death among adults aged 65 and older, yet the onset and progression of the disease is poorly understood. What is known is that the presence of amyloid, particularly polymerized Aß42, defines when people are on the AD continuum. Interestingly, as AD progresses, less Aß42 is detectable in the plasma, a phenomenon thought to result from Aß becoming more aggregated in the brain and less Aß42 and Aß40 being transported from the brain to the plasma via the CSF. We propose that extracellular vesicles (EVs) play a role in this transport. EVs are found in bodily fluids such as blood, urine, and cerebrospinal fluid and carry diverse "cargos" of bioactive molecules (e.g., proteins, nucleic acids, lipids, metabolites) that dynamically reflect changes in the cells from which they are secreted. While Aß42 and Aß40 have been reported to be present in EVs, it is not known whether this interaction is specific for these peptides and thus whether amyloid-carrying EVs play a role in AD and/or serve as brain-specific biomarkers of the AD process. To determine if there is a specific interaction between Aß and EVs, we used isothermal titration calorimetry (ITC) and discovered that Aß42 and Aß40 bind to EVs in a manner that is sequence specific, saturable, and endothermic. In addition, Aß incubation with EVs overnight yielded larger amounts of bound Aß peptide that was fibrillar in structure. These findings point to a specific amyloid-EV interaction, a potential role for EVs in the transport of amyloid from the brain to the blood, and a role for this amyloid pool in the AD process.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Adult , Humans , Peptides , Amyloidogenic Proteins , Plasma
2.
J Infect Dis ; 223(7): 1284-1294, 2021 04 08.
Article in English | MEDLINE | ID: mdl-32809013

ABSTRACT

BACKGROUND: Varicella zoster virus (VZV) vasculopathy is characterized by persistent arterial inflammation leading to stroke. Studies show that VZV induces amyloid formation that may aggravate vasculitis. Thus, we determined if VZV central nervous system infection produces amyloid. METHODS: Aß peptides, amylin, and amyloid were measured in cerebrospinal fluid (CSF) from 16 VZV vasculopathy subjects and 36 stroke controls. To determine if infection induced amyloid deposition, mock- and VZV-infected quiescent primary human perineurial cells (qHPNCs), present in vasculature, were analyzed for intracellular amyloidogenic transcripts/proteins and amyloid. Supernatants were assayed for amyloidogenic peptides and ability to induce amyloid formation. To determine amylin's function during infection, amylin was knocked down with small interfering RNA and viral complementary DNA (cDNA) was quantitated. RESULTS: Compared to controls, VZV vasculopathy CSF had increased amyloid that positively correlated with amylin and anti-VZV antibody levels; Aß40 was reduced and Aß42 unchanged. Intracellular amylin, Aß42, and amyloid were seen only in VZV-infected qHPNCs. VZV-infected supernatant formed amyloid fibrils following addition of amyloidogenic peptides. Amylin knockdown decreased viral cDNA. CONCLUSIONS: VZV infection increased levels of amyloidogenic peptides and amyloid in CSF and qHPNCs, indicating that VZV-induced amyloid deposition may contribute to persistent arterial inflammation in VZV vasculopathy. In addition, we identified a novel proviral function of amylin.


Subject(s)
Amyloid beta-Peptides , Amyloid , Arteritis , Herpes Zoster , Islet Amyloid Polypeptide , Peptide Fragments , Amyloid/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Arteritis/cerebrospinal fluid , Arteritis/diagnosis , Arteritis/virology , DNA, Complementary , DNA, Viral , Herpes Zoster/cerebrospinal fluid , Herpes Zoster/diagnosis , Herpesvirus 3, Human , Humans , Islet Amyloid Polypeptide/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Stroke
3.
J Infect Dis ; 221(7): 1088-1097, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31665341

ABSTRACT

BACKGROUND: Herpes zoster is linked to amyloid-associated diseases, including dementia, macular degeneration, and diabetes mellitus, in epidemiological studies. Thus, we examined whether varicella-zoster virus (VZV)-infected cells produce amyloid. METHODS: Production of intracellular amyloidogenic proteins (amylin, amyloid precursor protein [APP], and amyloid-ß [Aß]) and amyloid, as well as extracellular amylin, Aß, and amyloid, was compared between mock- and VZV-infected quiescent primary human spinal astrocytes (qHA-sps). The ability of supernatant from infected cells to induce amylin or Aß42 aggregation was quantitated. Finally, the amyloidogenic activity of viral peptides was examined. RESULTS: VZV-infected qHA-sps, but not mock-infected qHA-sps, contained intracellular amylin, APP, and/or Aß, and amyloid. No differences in extracellular amylin, Aß40, or Aß42 were detected, yet only supernatant from VZV-infected cells induced amylin aggregation and, to a lesser extent, Aß42 aggregation into amyloid fibrils. VZV glycoprotein B (gB) peptides assembled into fibrils and catalyzed amylin and Aß42 aggregation. CONCLUSIONS: VZV-infected qHA-sps produced intracellular amyloid and their extracellular environment promoted aggregation of cellular peptides into amyloid fibrils that may be due, in part, to VZV gB peptides. These findings suggest that together with host and other environmental factors, VZV infection may increase the toxic amyloid burden and contribute to amyloid-associated disease progression.


Subject(s)
Amyloid beta-Peptides , Astrocytes , Islet Amyloid Polypeptide , Varicella Zoster Virus Infection/metabolism , Acyclovir/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Antiviral Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/virology , Cells, Cultured , Extracellular Space/metabolism , Humans , Intracellular Space/metabolism , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 114(51): E11029-E11036, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203673

ABSTRACT

Misfolding of tau proteins into prions and their propagation along neural circuits are thought to result in neurodegeneration causing Alzheimer's disease, progressive supranuclear palsy, chronic traumatic encephalopathy, and other tauopathies. Little is known about the molecular processes mediating tau prion replication and spreading in different brain regions. Using transgenic (Tg) mice with a neuronal promoter driving expression of human mutant (P301S) tau, we found that tau prion formation and histopathologic deposition is largely restricted to the hindbrain. Unexpectedly, tau mRNA and protein levels did not differ between the forebrain and hindbrain, suggesting that other factors modulating the conversion of tau into a prion exist and are region specific. Using a cell-based prion propagation assay, we discovered that tau prion replication is suppressed by forebrain-derived inhibitors, one of which is sortilin, a lysosomal sorting receptor. We also show that sortilin expression is higher in the forebrain than the hindbrain across the life span of the Tg mice, suggesting that sortilin, at least in part, inhibits forebrain tau prion replication in vivo. Our findings provide evidence for selective vulnerability in mice resulting in highly regulated levels of tau prion propagation, thus affording a model for identification of additional molecules that could mitigate the levels of tau prions in human tauopathies.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Protein Binding , Rhombencephalon/metabolism , Rhombencephalon/pathology
6.
Wound Repair Regen ; 23(4): 591-600, 2015.
Article in English | MEDLINE | ID: mdl-26032846

ABSTRACT

Chronic wounds such as diabetic ulcers pose a significant challenge as a number of underlying deficiencies prevent natural healing. In pursuit of a regenerative wound therapy, we developed a heparin-based coacervate delivery system that provides controlled release of heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF) within the wound bed. In this study, we used a polygenic type 2 diabetic mouse model to evaluate the capacity of HB-EGF coacervate to overcome the deficiencies of diabetic wound healing. In full-thickness excisional wounds on NONcNZO10 diabetic mice, HB-EGF coacervate enhanced the proliferation and migration of epidermal keratinocytes, leading to accelerated epithelialization. Furthermore, increased collagen deposition within the wound bed led to faster wound contraction and greater wound vascularization. Additionally, in vitro assays demonstrated that HB-EGF released from the coacervate successfully increased migration of diabetic human keratinocytes. The multifunctional role of HB-EGF in the healing process and its enhanced efficacy when delivered by the coacervate make it a promising therapy for diabetic wounds.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/metabolism , Heparin-binding EGF-like Growth Factor/administration & dosage , Keratinocytes/pathology , Skin/injuries , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Animals , Cell Movement , Cell Proliferation , Diabetes Mellitus, Type 2/pathology , Drug Delivery Systems , Keratinocytes/metabolism , Male , Mice , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
7.
Front Cell Neurosci ; 18: 1340448, 2024.
Article in English | MEDLINE | ID: mdl-38323188

ABSTRACT

Alzheimer's disease (AD), characterized by memory loss and cognitive decline, affects nearly 50 million people worldwide. Amyloid beta (Aß) plaques and intracellular neurofibrillary tangles (NFTs) of phosphorylated Tau protein (pTau) are key histopathological features of the disease in the brain, and recent advances have also identified AD histopathology in the retina. Thus, the retina represents a central nervous system (CNS) tissue highly amenable to non-invasive diagnostic imaging that shows promise as a biomarker for early AD. Given the devastating effects of AD on patients, their families, and society, new treatment modalities that can significantly alter the disease course are urgently needed. In this study, we have developed and characterized a novel human retinal organoid (RO) model derived from induced pluripotent stem cells (iPSCs) from patients with familial AD due to mutations in the amyloid precursor protein gene (APP). Using immunofluorescence and histological staining, we evaluated the cellular composition and AD histopathological features of AD-ROs compared to control ROs from healthy individuals. We found that AD-ROs largely resemble their healthy control counterparts in cellular composition but display increased levels of Aß and pTau. We also present proof of principle of an assay to quantify amyloid levels in whole ROs. This in vitro model of the human AD retina constitutes a new tool for drug screening, biomarker discovery, and pathophysiological studies.

8.
Nat Commun ; 14(1): 118, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36624100

ABSTRACT

Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.


Subject(s)
Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Tauopathies , Animals , Female , Male , Mice , Brain/metabolism , Disease Models, Animal , Microglia/metabolism , Phenotype , Receptors, Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tauopathies/metabolism
10.
ACS Biomater Sci Eng ; 8(9): 3883-3898, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-35950643

ABSTRACT

Myocardial infarction causes cardiomyocyte death and persistent inflammatory responses, which generate adverse pathological remodeling. Delivering therapeutic proteins from injectable materials in a controlled-release manner may present an effective biomedical approach for treating this disease. A thermoresponsive injectable gel composed of chitosan, conjugated with poly(N-isopropylacrylamide) and sulfonate groups, was developed for spatiotemporal protein delivery to protect cardiac function after myocardial infarction. The thermoresponsive gel delivered vascular endothelial growth factor (VEGF), interleukin-10 (IL-10), and platelet-derived growth factor (PDGF) in a sequential and sustained manner in vitro. An acute myocardial infarction mouse model was used to evaluate polymer biocompatibility and to determine therapeutic effects from the delivery system on cardiac function. Immunohistochemistry showed biocompatibility of the hydrogel, while the controlled delivery of the proteins reduced macrophage infiltration and increased vascularization. Echocardiography showed an improvement in ejection fraction and fractional shortening after injecting the thermal gel and proteins. A factorial design of experimental study was implemented to optimize the delivery system for the best combination and doses of proteins for further increasing stable vascularization and reducing inflammation using a subcutaneous injection mouse model. The results showed that VEGF, IL-10, and FGF-2 demonstrated significant contributions toward promoting long-term vascularization, while PDGF's effect was minimal.


Subject(s)
Myocardial Infarction , Vascular Endothelial Growth Factor A , Animals , Hydrogels/pharmacology , Hydrogels/therapeutic use , Interleukin-10/therapeutic use , Mice , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/therapeutic use
11.
Alzheimers Res Ther ; 14(1): 88, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768831

ABSTRACT

BACKGROUND: The apolipoprotein E (APOE) ε4 allele confers the strongest risk for late-onset Alzheimer's disease (AD) besides age itself, but the mechanisms underlying this risk are debated. One hypothesis supported by evidence from multiple labs is that apoE4 binds to the amyloid-ß (Aß) peptide and catalyzes its polymerization into neurotoxic oligomers and fibrils. Inhibiting this early step in the amyloid cascade may thereby reduce or prevent neurodegeneration and AD. METHODS: Using a design of experiments (DOE) approach, we developed a high-throughput assay to identify inhibitors of apoE4-catalyzed polymerization of Aß into oligomers and fibrils. We used it to screen the NIH Clinical Collection of small molecule drugs tested previously in human clinical trials. We then evaluated the efficacy and cytotoxicity of the hit compounds in primary neuron models of apoE4-induced Aß and phosphorylated tau aggregation. Finally, we performed retrospective analyses of the National Alzheimer's Coordinating Center (NACC) clinical dataset, using Cox regression and Cox proportional hazards models to determine if the use of two FDA-approved hit compounds was associated with better cognitive scores (Mini-Mental State Exam), or improved AD clinical diagnosis, when compared with other medications of the same clinical indication. RESULTS: Our high-throughput screen identified eight blood-brain barrier (BBB)-permeable hit compounds that reduced apoE4-catalyzed Aß oligomer and fibril formation in a dose-dependent manner. Five hit compounds were non-toxic toward cultured neurons and also reduced apoE4-promoted Aß and tau neuropathology in a dose-dependent manner. Three of the five compounds were determined to be specific inhibitors of apoE4, whereas the other two compounds were Aß or tau aggregation inhibitors. When prescribed to AD patients for their normal clinical indications, two of the apoE4 inhibitors, imipramine and olanzapine, but not other (non-hit) antipsychotic or antidepressant medications, were associated with improvements in cognition and clinical diagnosis, especially among APOE4 carriers. CONCLUSIONS: The critical test of any proposed AD mechanism is whether it leads to effective treatments. Our high-throughput screen identified two promising FDA-approved drugs, imipramine and olanzapine, which have no structural, functional, or clinical similarities other than their shared ability to inhibit apoE4-catalyzed Aß polymerization, thus identifying this mechanism as an essential contribution of apoE4 to AD.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Catalysis , Cognition , Humans , Imipramine/therapeutic use , Olanzapine/therapeutic use , Polymerization , Retrospective Studies
12.
iScience ; 25(11): 105288, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36304124

ABSTRACT

Previously, we found that amyloid-beta (Aß) competitively inhibits the kinesin motor protein KIF11 (Kinesin-5/Eg5), leading to defects in the microtubule network and in neurotransmitter and neurotrophin receptor localization and function. These biochemical and cell biological mechanisms for Aß-induced neuronal dysfunction may underlie learning and memory defects in Alzheimer's disease (AD). Here, we show that KIF11 overexpression rescues Aß-mediated decreases in dendritic spine density in cultured neurons and in long-term potentiation in hippocampal slices. Furthermore, Kif11 overexpression from a transgene prevented spatial learning deficits in the 5xFAD mouse model of AD. Finally, increased KIF11 expression in neuritic plaque-positive AD patients' brains was associated with better cognitive performance and higher expression of synaptic protein mRNAs. Taken together, these mechanistic biochemical, cell biological, electrophysiological, animal model, and human data identify KIF11 as a key target of Aß-mediated toxicity in AD, which damages synaptic structures and functions critical for learning and memory in AD.

13.
Front Aging Neurosci ; 13: 718426, 2021.
Article in English | MEDLINE | ID: mdl-34603007

ABSTRACT

Innate immune system activation and inflammation are associated with and may contribute to clinical outcomes in people with Down syndrome (DS), neurodegenerative diseases such as Alzheimer's disease (AD), and normal aging. In addition to serving as potential diagnostic biomarkers, innate immune system activation and inflammation may play a contributing or causal role in these conditions, leading to the hypothesis that effective therapies should seek to dampen their effects. However, recent intervention studies with the innate immune system activator granulocyte-macrophage colony-stimulating factor (GM-CSF) in animal models of DS, AD, and normal aging, and in an AD clinical trial suggest that activating the innate immune system and inflammation may instead be therapeutic. We consider evidence that DS, AD, and normal aging are accompanied by innate immune system activation and inflammation and discuss whether and when during the disease process it may be therapeutically beneficial to suppress or promote such activation.

14.
J Control Release ; 253: 73-81, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28315407

ABSTRACT

Platelet-rich plasma (PRP) is widely used for many clinical indications including wound healing due to the high concentrations of growth factors. However, the short half-life of these therapeutic proteins requires multiple large doses, and their efficacy is highly debated among clinicians. Here we report a method of protecting these proteins and releasing them in a controlled manner via a heparin-based coacervate delivery vehicle to improve wound healing in a porcine model. Platelet-derived proteins incorporated into the coacervate were protected and slowly released over 3weeks in vitro. In a porcine model, PRP coacervate significantly accelerated the healing response over 10days, in part by increasing the rate of wound reepithelialization by 35% compared to control. Additionally, PRP coacervate doubled the rate of wound contraction compared to all other treatments, including that of free PRP proteins. Wounds treated with PRP coacervate exhibited increased collagen alignment and an advanced state of vascularity compared to control treatments. These results suggest that this preparation of PRP accelerates healing of cutaneous wounds only as a controlled release formulation. The coacervate delivery vehicle is a simple and effective tool to improve the therapeutic efficacy of platelet-derived proteins for wound healing.


Subject(s)
Platelet-Rich Plasma , Wound Healing/drug effects , Animals , Collagen/metabolism , Delayed-Action Preparations/therapeutic use , Female , Humans , Skin/drug effects , Skin/metabolism , Swine
15.
Aging Dis ; 7(4): 336-449, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27493838

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder with complicated pathophysiologic mechanisms. Endoplasmic reticulum (ER) stress appears to play a critical role in the progression of PD. We demonstrated that basic fibroblast growth factor (bFGF), as a neurotropic factor, inhibited ER stress-induced neuronal cell apoptosis and that 6-hydroxydopamine (6-OHDA)-induced ER stress was involved in the progression of PD in rats. bFGF administration improved motor function recovery, increased tyrosine hydroxylase (TH)-positive neuron survival, and upregulated the levels of neurotransmitters in PD rats. The 6-OHDA-induced ER stress response proteins were inhibited by bFGF treatment. Meanwhile, bFGF also increased expression of TH. The administration of bFGF activated the downstream signals PI3K/Akt and Erk1/2 in vivo and in vitro. Inhibition of the PI3K/Akt and Erk1/2 pathways by specific inhibitors partially reduced the protective effect of bFGF. This study provides new insight towards bFGF translational drug development for PD involving the regulation of ER stress.

16.
Int J Biol Sci ; 12(1): 87-99, 2016.
Article in English | MEDLINE | ID: mdl-26722220

ABSTRACT

Spinal cord injury (SCI) induces the disruption of the blood-spinal cord barrier (BSCB) which leads to infiltration of blood cells, an inflammatory response, and neuronal cell death, resulting spinal cord secondary damage. Retinoic acid (RA) has a neuroprotective effect in both ischemic brain injury and SCI, however the relationship between BSCB disruption and RA in SCI is still unclear. In this study, we demonstrated that autophagy and ER stress are involved in the protective effect of RA on the BSCB. RA attenuated BSCB permeability and decreased the loss of tight junction (TJ) molecules such as P120, ß-catenin, Occludin and Claudin5 after injury in vivo as well as in Brain Microvascular Endothelial Cells (BMECs). Moreover, RA administration improved functional recovery in the rat model of SCI. RA inhibited the expression of CHOP and caspase-12 by induction of autophagic flux. However, RA had no significant effect on protein expression of GRP78 and PDI. Furthermore, combining RA with the autophagy inhibitor chloroquine (CQ) partially abolished its protective effect on the BSCB via exacerbated ER stress and subsequent loss of tight junctions. Taken together, the neuroprotective role of RA in recovery from SCI is related to prevention of of BSCB disruption via the activation of autophagic flux and the inhibition of ER stress-induced cell apoptosis. These findings lay the groundwork for future translational studies of RA for CNS diseases, especially those related to BSCB disruption.


Subject(s)
Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Endoplasmic Reticulum Stress/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Tretinoin/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Cell Survival/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Rats , Rats, Sprague-Dawley
17.
Mol Neurobiol ; 53(10): 7298-7311, 2016 12.
Article in English | MEDLINE | ID: mdl-26687235

ABSTRACT

Many traumatic brain injury (TBI) survivors sustain neurological disability and cognitive impairments due to the lack of defined therapies to reduce TBI-induced blood-brain barrier (BBB) breakdown. Exogenous basic fibroblast growth factor (bFGF) has been shown to have neuroprotective function in brain injury. The present study therefore investigates the beneficial effects of bFGF on the BBB after TBI and the underlying mechanisms. In this study, we demonstrate that bFGF reduces neurofunctional deficits and preserves BBB integrity in a mouse model of TBI. bFGF suppresses RhoA and upregulates tight junction proteins, thereby mitigating BBB breakdown. In vitro, bFGF exerts a protective effect on BBB by upregulating tight junction proteins claudin-5, occludin, zonula occludens-1, p120-catenin, and ß-catenin under oxygen glucose deprivation/reoxygenation (OGD) in human brain microvascular endothelial cells (HBMECs). Both the in vivo and in vitro effects are related to the activation of the downstream signaling pathway, PI3K/Akt/Rac-1. Inhibition of the PI3K/Akt or Rac-1 by specific inhibitors LY294002 or si-Rac-1, respectively, partially reduces the protective effect of bFGF on BBB integrity. Overall, our results indicate that the protective role of bFGF on BBB involves the regulation of tight junction proteins and RhoA in the TBI model and OGD-induced HBMECs injury, and that activation of the PI3K/Akt /Rac-1 signaling pathway underlies these effects.


Subject(s)
Blood-Brain Barrier/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Fibroblast Growth Factor 2/pharmacology , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , Animals , Biomarkers/metabolism , Blood-Brain Barrier/injuries , Brain Injuries, Traumatic/pathology , Endothelial Cells/metabolism , Glucose/deficiency , Guanosine Triphosphate/metabolism , Humans , Intercellular Junctions/drug effects , Male , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
18.
Curr Pharm Biotechnol ; 16(7): 621-9, 2015.
Article in English | MEDLINE | ID: mdl-25658378

ABSTRACT

Protein, gene, and small molecule therapies hold great potential for facilitating comprehensive tissue repair and regeneration. However, their clinical value will rely on effective delivery systems which maximize their therapeutic benefit. Significant advances have been made in recent years towards biomaterial delivery systems to satisfy this clinical need. Here we summarize the most outstanding advances in drug delivery technology for cutaneous wound healing.


Subject(s)
Biocompatible Materials/administration & dosage , Drug Delivery Systems/methods , Wound Healing/drug effects , Wound Healing/physiology , Animals , Drug Delivery Systems/trends , Humans , Regeneration/drug effects , Regeneration/physiology
19.
J Control Release ; 207: 7-17, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25836592

ABSTRACT

Treatment of ischemia through therapeutic angiogenesis faces significant challenges. Growth factor (GF)-based therapies can be more effective when concerns such as GF spatiotemporal presentation, bioactivity, bioavailability, and localization are addressed. During angiogenesis, vascular endothelial GF (VEGF) is required early to initiate neovessel formation while platelet-derived GF (PDGF-BB) is needed later to stabilize the neovessels. The spatiotemporal delivery of multiple bioactive GFs involved in angiogenesis, in a close mimic to physiological cues, holds great potential to treat ischemic diseases. To achieve sequential release of VEGF and PDGF, we embed VEGF in fibrin gel and PDGF in a heparin-based coacervate that is distributed in the same fibrin gel. In vitro, we show the benefits of this controlled delivery approach on cell proliferation, chemotaxis, and capillary formation. A rat myocardial infarction (MI) model demonstrated the effectiveness of this delivery system in improving cardiac function, ventricular wall thickness, angiogenesis, cardiac muscle survival, and reducing fibrosis and inflammation in the infarct zone compared to saline, empty vehicle, and free GFs. Collectively, our results show that this delivery approach mitigated the injury caused by MI and may serve as a new therapy to treat ischemic hearts pending further examination.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-sis/administration & dosage , Vascular Endothelial Growth Factor A/administration & dosage , Ventricular Function, Left/drug effects , Angiogenesis Inducing Agents/chemistry , Animals , Becaplermin , Cell Proliferation/drug effects , Cells, Cultured , Chemistry, Pharmaceutical , Chemotaxis/drug effects , Delayed-Action Preparations , Disease Models, Animal , Drug Carriers , Drug Combinations , Fibrin/chemistry , Fibrosis , Gels , Heparin/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Kinetics , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Smooth Muscle/drug effects , Papio , Proto-Oncogene Proteins c-sis/chemistry , Rats, Sprague-Dawley , Recovery of Function , Solubility , Vascular Endothelial Growth Factor A/chemistry
20.
Int J Clin Exp Pathol ; 8(3): 2928-36, 2015.
Article in English | MEDLINE | ID: mdl-26045801

ABSTRACT

Stem cell transplantation may provide an alternative therapy to promote functional recovery after various neurological disorders including cerebral infarct. Due to the minimal immunogenicity and neuronal differentiation potential of neural stem cells (NSCs), we tested whether intravenous administration of mice-derived C17.2 NSCs could improve neurological function deficit and cerebral infarction volume after ischemic stroke in rats. Additionally, we evaluated the survival, migration, proliferation, and differentiation capacity of transplanted NSCs in the rat brain. Intravenous infusion of NSCs after middle cerebral artery occlusion (MCAO) showed better performance in neurobiological severity scores after MCAO compared to control. However, the volume of cerebral infarction was not different at 7 days after MCAO compared with control. Transplanted NSCs were detected in the ischemic region but not in the contralateral hemisphere. NSCs differentiated into neurons or astrocytes after MCAO. These data suggest that intravenously transplanted NSCs can migrate, proliferate, and differentiate into neurons and astrocytes in the rat brain with focal ischemia and improve functional recovery.


Subject(s)
Ischemic Attack, Transient/surgery , Neural Stem Cells/transplantation , Recovery of Function , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cell Line , Cell Movement , Disease Models, Animal , Fluorescent Antibody Technique , Heterografts , Male , Mice , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL