Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.258
Filter
1.
Int J Nanomedicine ; 19: 4857-4875, 2024.
Article in English | MEDLINE | ID: mdl-38828195

ABSTRACT

Brain diseases are the most devastating problem among the world's increasingly aging population, and the number of patients with neurological diseases is expected to increase in the future. Although methods for delivering drugs to the brain have advanced significantly, none of these approaches provide satisfactory results for the treatment of brain diseases. This remains a challenge due to the unique anatomy and physiology of the brain, including tight regulation and limited access of substances across the blood-brain barrier. Nanoparticles are considered an ideal drug delivery system to hard-to-reach organs such as the brain. The development of new drugs and new nanomaterial-based brain treatments has opened various opportunities for scientists to develop brain-specific delivery systems that could improve treatment outcomes for patients with brain disorders such as Alzheimer's disease, Parkinson's disease, stroke and brain tumors. In this review, we discuss noteworthy literature that examines recent developments in brain-targeted nanomedicines used in the treatment of neurological diseases.


Subject(s)
Blood-Brain Barrier , Brain , Drug Delivery Systems , Nanomedicine , Humans , Nanomedicine/methods , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Drug Delivery Systems/methods , Animals , Nanoparticles/chemistry , Brain Diseases/drug therapy , Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/pharmacokinetics , Parkinson Disease/drug therapy , Alzheimer Disease/drug therapy
2.
Eur J Med Res ; 29(1): 313, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38849950

ABSTRACT

Alzheimer's disease (AD) is a diverse disease with a complex pathophysiology. The presence of extracellular ß-amyloid deposition as neuritic plaques and intracellular accumulation of hyper-phosphorylated tau as neurofibrillary tangles remain the core neuropathologic criteria for diagnosing Alzheimer's disease. Nonetheless, several recent basic discoveries have revealed significant pathogenic roles for other essential cellular and molecular processes. Previously, there were not so many disease-modifying medications (DMT) available as drug distribution through the blood-brain barrier (BBB) is difficult due to its nature, especially drugs of polypeptides nature and proteins. Recently FDA has approved lecanemab as DMT for its proven efficacy. It is also complicated to deliver drugs for diseases like epilepsy or any brain tumor due to the limitations of the BBB. After the advancements in the drug delivery system, different techniques are used to transport the medication across the BBB. Other methods are used, like enhancement of brain blood vessel fluidity by liposomes, infusion of hyperosmotic solutions, and local intracerebral implants, but these are invasive approaches. Non-invasive approaches include the formulation of nanoparticles and their coating with polymers. This review article emphasizes all the above-mentioned techniques, procedures, and challenges to transporting medicines across the BBB. It summarizes the most recent literature dealing with drug delivery across the BBB.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Drug Delivery Systems , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Drug Delivery Systems/methods , Animals , Biological Transport
3.
Theranostics ; 14(8): 3221-3245, 2024.
Article in English | MEDLINE | ID: mdl-38855177

ABSTRACT

The availability of non-invasive drug delivery systems capable of efficiently transporting bioactive molecules across the blood-brain barrier to specific cells at the injury site in the brain is currently limited. Delivering drugs to neurons presents an even more formidable challenge due to their lower numbers and less phagocytic nature compared to other brain cells. Additionally, the diverse types of neurons, each performing specific functions, necessitate precise targeting of those implicated in the disease. Moreover, the complex synthetic design of drug delivery systems often hinders their clinical translation. The production of nanomaterials at an industrial scale with high reproducibility and purity is particularly challenging. However, overcoming this challenge is possible by designing nanomaterials through a straightforward, facile, and easily reproducible synthetic process. Methods: In this study, we have developed a third-generation 2-deoxy-glucose functionalized mixed layer dendrimer (2DG-D) utilizing biocompatible and cost-effective materials via a highly facile convergent approach, employing copper-catalyzed click chemistry. We further evaluated the systemic neuronal targeting and biodistribution of 2DG-D, and brain delivery of a neuroprotective agent pioglitazone (Pio) in a pediatric traumatic brain injury (TBI) model. Results: The 2DG-D exhibits favorable characteristics including high water solubility, biocompatibility, biological stability, nanoscale size, and a substantial number of end groups suitable for drug conjugation. Upon systemic administration in a pediatric mouse model of traumatic brain injury (TBI), the 2DG-D localizes in neurons at the injured brain site, clears rapidly from off-target locations, effectively delivers Pio, ameliorates neuroinflammation, and improves behavioral outcomes. Conclusions: The promising in vivo results coupled with a convenient synthetic approach for the construction of 2DG-D makes it a potential nanoplatform for addressing brain diseases.


Subject(s)
Dendrimers , Deoxyglucose , Drug Delivery Systems , Neurons , Animals , Dendrimers/chemistry , Neurons/drug effects , Neurons/metabolism , Drug Delivery Systems/methods , Deoxyglucose/pharmacology , Deoxyglucose/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Mice , Pioglitazone/pharmacology , Pioglitazone/administration & dosage , Pioglitazone/pharmacokinetics , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain/metabolism , Brain/drug effects , Brain Diseases/drug therapy , Humans , Disease Models, Animal , Tissue Distribution , Male
4.
Nat Commun ; 15(1): 4698, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844770

ABSTRACT

Given the marginal penetration of most drugs across the blood-brain barrier, the efficacy of various agents remains limited for glioblastoma (GBM). Here we employ low-intensity pulsed ultrasound (LIPU) and intravenously administered microbubbles (MB) to open the blood-brain barrier and increase the concentration of liposomal doxorubicin and PD-1 blocking antibodies (aPD-1). We report results on a cohort of 4 GBM patients and preclinical models treated with this approach. LIPU/MB increases the concentration of doxorubicin by 2-fold and 3.9-fold in the human and murine brains two days after sonication, respectively. Similarly, LIPU/MB-mediated blood-brain barrier disruption leads to a 6-fold and a 2-fold increase in aPD-1 concentrations in murine brains and peritumoral brain regions from GBM patients treated with pembrolizumab, respectively. Doxorubicin and aPD-1 delivered with LIPU/MB upregulate major histocompatibility complex (MHC) class I and II in tumor cells. Increased brain concentrations of doxorubicin achieved by LIPU/MB elicit IFN-γ and MHC class I expression in microglia and macrophages. Doxorubicin and aPD-1 delivered with LIPU/MB results in the long-term survival of most glioma-bearing mice, which rely on myeloid cells and lymphocytes for their efficacy. Overall, this translational study supports the utility of LIPU/MB to potentiate the antitumoral activities of doxorubicin and aPD-1 for GBM.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Doxorubicin , Microbubbles , Programmed Cell Death 1 Receptor , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Doxorubicin/analogs & derivatives , Animals , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/drug therapy , Glioma/immunology , Glioma/pathology , Brain/metabolism , Brain/drug effects , Female , Drug Delivery Systems , Ultrasonic Waves , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioblastoma/pathology , Male , Microglia/drug effects , Microglia/metabolism , Mice, Inbred C57BL , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , Polyethylene Glycols
5.
PLoS One ; 19(6): e0297451, 2024.
Article in English | MEDLINE | ID: mdl-38857220

ABSTRACT

Traumatic brain injury has faced numerous challenges in drug development, primarily due to the difficulty of effectively delivering drugs to the brain. However, there is a potential solution in targeted drug delivery methods involving antibody-drug conjugates or nanocarriers conjugated with targeting antibodies. Following a TBI, the blood-brain barrier (BBB) becomes permeable, which can last for years and allow the leakage of harmful plasma proteins. Consequently, an appealing approach for TBI treatment involves using drug delivery systems that utilize targeting antibodies and nanocarriers to help restore BBB integrity. In our investigation of this strategy, we examined the efficacy of free antibodies and nanocarriers targeting a specific endothelial surface marker called vascular cell adhesion molecule-1 (VCAM-1), which is known to be upregulated during inflammation. In a mouse model of TBI utilizing central fluid percussion injury, free VCAM-1 antibody did not demonstrate superior targeting when comparing sham vs. TBI brain. However, the administration of VCAM-1-targeted nanocarriers (liposomes) exhibited a 10-fold higher targeting specificity in TBI brain than in sham control. Flow cytometry and confocal microscopy analysis confirmed that VCAM-1 liposomes were primarily taken up by brain endothelial cells post-TBI. Consequently, VCAM-1 liposomes represent a promising platform for the targeted delivery of therapeutics to the brain following traumatic brain injury.


Subject(s)
Blood-Brain Barrier , Brain Injuries, Traumatic , Nanoparticles , Vascular Cell Adhesion Molecule-1 , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Nanoparticles/chemistry , Liposomes , Male , Drug Delivery Systems , Mice, Inbred C57BL , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/drug effects
6.
Sci Rep ; 14(1): 10573, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38719983

ABSTRACT

Multiple sclerosis (MS) is a chronic neurological disease characterized by inflammatory demyelination that disrupts neuronal transmission resulting in neurodegeneration progressive disability. While current treatments focus on immunosuppression to limit inflammation and further myelin loss, no approved therapies effectively promote remyelination to mitigate the progressive disability associated with chronic demyelination. Lysophosphatidic acid (LPA) is a pro-inflammatory lipid that is upregulated in MS patient plasma and cerebrospinal fluid (CSF). LPA activates the LPA1 receptor, resulting in elevated CNS cytokine and chemokine levels, infiltration of immune cells, and microglial/astrocyte activation. This results in a neuroinflammatory response leading to demyelination and suppressed remyelination. A medicinal chemistry effort identified PIPE-791, an oral, brain-penetrant, LPA1 antagonist. PIPE-791 was characterized in vitro and in vivo and was found to be a potent, selective LPA1 antagonist with slow receptor off-rate kinetics. In vitro, PIPE-791 induced OPC differentiation and promoted remyelination following a demyelinating insult. PIPE-791 further mitigated the macrophage-mediated inhibition of OPC differentiation and inhibited microglial and fibroblast activation. In vivo, the compound readily crossed the blood-brain barrier and blocked LPA1 in the CNS after oral dosing. Direct dosing of PIPE-791 in vivo increased oligodendrocyte number, and in the mouse experimental autoimmune encephalomyelitis (EAE) model of MS, we observed that PIPE-791 promoted myelination, reduced neuroinflammation, and restored visual evoked potential latencies (VEP). These findings support targeting LPA1 for remyelination and encourage development of PIPE-791 for treating MS patients with advantages not seen with current immunosuppressive disease modifying therapies.


Subject(s)
Multiple Sclerosis , Receptors, Lysophosphatidic Acid , Remyelination , Animals , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/metabolism , Remyelination/drug effects , Humans , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Brain/metabolism , Brain/drug effects , Brain/pathology , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice, Inbred C57BL , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Lysophospholipids/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
7.
J Neuroinflammation ; 21(1): 131, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760784

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) causes acute and long-term cognitive deficits. However, information on the prevention and treatment of cognitive dysfunction after sepsis is limited. The neuropeptide orexin-A (OXA) has been shown to play a protective role against neurological diseases by modulating the inflammatory response through the activation of OXR1 and OXR2 receptors. However, the role of OXA in mediating the neuroprotective effects of SAE has not yet been reported. METHODS: A mouse model of SAE was induced using cecal ligation perforation (CLP) and treated via intranasal administration of exogenous OXA after surgery. Mouse survival, in addition to cognitive and anxiety behaviors, were assessed. Changes in neurons, cerebral edema, blood-brain barrier (BBB) permeability, and brain ultrastructure were monitored. Levels of pro-inflammatory factors (IL-1ß, TNF-α) and microglial activation were also measured. The underlying molecular mechanisms were investigated by proteomics analysis and western blotting. RESULTS: Intranasal OXA treatment reduced mortality, ameliorated cognitive and emotional deficits, and attenuated cerebral edema, BBB disruption, and ultrastructural brain damage in mice. In addition, OXA significantly reduced the expression of the pro-inflammatory factors IL-1ß and TNF-α, and inhibited microglial activation. In addition, OXA downregulated the expression of the Rras and RAS proteins, and reduced the phosphorylation of P-38 and JNK, thus inhibiting activation of the MAPK pathway. JNJ-10,397,049 (an OXR2 blocker) reversed the effect of OXA, whereas SB-334,867 (an OXR1 blocker) did not. CONCLUSION: This study demonstrated that the intranasal administration of moderate amounts of OXA protects the BBB and inhibits the activation of the OXR2/RAS/MAPK pathway to attenuate the outcome of SAE, suggesting that OXA may be a promising therapeutic approach for the management of SAE.


Subject(s)
Mice, Inbred C57BL , Orexins , Sepsis-Associated Encephalopathy , Animals , Mice , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/metabolism , Orexins/metabolism , Male , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Disease Models, Animal , Administration, Intranasal
8.
Eur J Pharmacol ; 974: 176631, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38692425

ABSTRACT

OBJECTIVE: Dasatinib and quercetin (D & Q) have demonstrated promise in improving aged-related pathophysiological dysfunctions in humans and mice. Herein we aimed to ascertain whether the heat stress (HS)-induced cognitive deficits in aged or even young adult male mice can be reduced by D & Q therapy. METHODS: Before the onset of HS, animals were pre-treated with D & Q or placebo for 3 consecutive days every 2 weeks over a 10-week period. Cognitive function, intestinal barrier permeability, and blood-brain barrier permeability were assessed. RESULTS: Compared to the non-HS young adult male mice, the HS young adult male mice or the aged male mice had significantly lesser extents of the exacerbated stress reactions, intestinal barrier disruption, endotoxemia, systemic inflammation and oxidative stress, blood-brain barrier disruption, hippocampal inflammation and oxidative stress, and cognitive deficits evaluated at 7 days post-HS. All the cognitive deficits and other syndromes that occurred in young adult HS mice or in aged HS mice were significantly attenuated by D & Q therapy (P < 0.01). Compared to the young adult HS mice, the aged HS mice had significantly (P < 0.01) higher severity of cognitive deficits and other related syndromes. CONCLUSIONS: First, our data show that aged male mice are more vulnerable to HS-induced cognitive deficits than those of the young adult male mice. Second, we demonstrate that a combination of D and Q therapy attenuates cognitive deficits in heat stressed aged or young adult male mice via broad normalization of the brain-gut-endotoxin axis function.


Subject(s)
Blood-Brain Barrier , Dasatinib , Oxidative Stress , Quercetin , Animals , Male , Dasatinib/pharmacology , Dasatinib/therapeutic use , Quercetin/pharmacology , Quercetin/therapeutic use , Mice , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Oxidative Stress/drug effects , Aging/drug effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Heat-Shock Response/drug effects , Permeability/drug effects , Drug Therapy, Combination , Hippocampus/drug effects , Hippocampus/metabolism , Cognition/drug effects
9.
Clin Transl Sci ; 17(5): e13804, 2024 May.
Article in English | MEDLINE | ID: mdl-38700454

ABSTRACT

St. John's wort (SJW) extract, a herbal medicine with antidepressant effects, is a potent inducer of intestinal and/or hepatic cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp), which can cause clinically relevant drug interactions. It is currently not known whether SJW can also induce P-gp activity at the human blood-brain barrier (BBB), which may potentially lead to decreased brain exposure and efficacy of certain central nervous system (CNS)-targeted P-gp substrate drugs. In this study, we used a combination of positron emission tomography (PET) imaging and cocktail phenotyping to gain a comprehensive picture on the effect of SJW on central and peripheral P-gp and CYP activities. Before and after treatment of healthy volunteers (n = 10) with SJW extract with a high hyperforin content (3-6%) for 12-19 days (1800 mg/day), the activity of P-gp at the BBB was assessed by means of PET imaging with the P-gp substrate [11C]metoclopramide and the activity of peripheral P-gp and CYPs was assessed by administering a low-dose phenotyping cocktail (caffeine, omeprazole, dextromethorphan, and midazolam or fexofenadine). SJW significantly increased peripheral P-gp, CYP3A, and CYP2C19 activity. Conversely, no significant changes in the peripheral metabolism, brain distribution, and P-gp-mediated efflux of [11C]metoclopramide across the BBB were observed following the treatment with SJW extract. Our data suggest that SJW does not lead to significant P-gp induction at the human BBB despite its ability to induce peripheral P-gp and CYPs. Simultaneous intake of SJW with CNS-targeted P-gp substrate drugs is not expected to lead to P-gp-mediated drug interactions at the BBB.


Subject(s)
Blood-Brain Barrier , Hypericum , Phloroglucinol , Phloroglucinol/analogs & derivatives , Plant Extracts , Positron-Emission Tomography , Terfenadine/analogs & derivatives , Terpenes , Humans , Hypericum/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Phloroglucinol/pharmacokinetics , Phloroglucinol/pharmacology , Phloroglucinol/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/pharmacokinetics , Male , Adult , Positron-Emission Tomography/methods , Terpenes/pharmacology , Terpenes/pharmacokinetics , Terpenes/metabolism , Female , Young Adult , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds/pharmacokinetics , Bridged Bicyclo Compounds/administration & dosage , Terfenadine/pharmacokinetics , Terfenadine/administration & dosage , Terfenadine/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Healthy Volunteers
10.
Nat Commun ; 15(1): 3987, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734698

ABSTRACT

Despite advances in active drug targeting for blood-brain barrier penetration, two key challenges persist: first, attachment of a targeting ligand to the drug or drug carrier does not enhance its brain biodistribution; and second, many brain diseases are intricately linked to microcirculation disorders that significantly impede drug accumulation within brain lesions even after they cross the barrier. Inspired by the neuroprotective properties of vinpocetine, which regulates cerebral blood flow, we propose a molecular library design centered on this class of cyclic tertiary amine compounds and develop a self-enhanced brain-targeted nucleic acid delivery system. Our findings reveal that: (i) vinpocetine-derived ionizable-lipidoid nanoparticles efficiently breach the blood-brain barrier; (ii) they have high gene-loading capacity, facilitating endosomal escape and intracellular transport; (iii) their administration is safe with minimal immunogenicity even with prolonged use; and (iv) they have potent pharmacologic brain-protective activity and may synergize with treatments for brain disorders as demonstrated in male APP/PS1 mice.


Subject(s)
Blood-Brain Barrier , Brain , Cerebrovascular Circulation , Nanoparticles , Vinca Alkaloids , Animals , Vinca Alkaloids/pharmacology , Vinca Alkaloids/pharmacokinetics , Vinca Alkaloids/administration & dosage , Vinca Alkaloids/chemistry , Nanoparticles/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice , Cerebrovascular Circulation/drug effects , Male , Brain/metabolism , Brain/drug effects , Brain/blood supply , Humans , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Tissue Distribution , Drug Delivery Systems , Mice, Transgenic
11.
J Neurosci Res ; 102(5): e25340, 2024 May.
Article in English | MEDLINE | ID: mdl-38745527

ABSTRACT

The ubiquity of nanoparticles, sourced from both natural environments and human activities, presents critical challenges for public health. While offering significant potential for innovative biomedical applications-especially in enhancing drug transport across the blood-brain barrier-these particles also introduce possible hazards due to inadvertent exposure. This concise review explores the paradoxical nature of nanoparticles, emphasizing their promising applications in healthcare juxtaposed with their potential neurotoxic consequences. Through a detailed examination, we delineate the pathways through which nanoparticles can reach the brain and the subsequent health implications. There is growing evidence of a disturbing association between nanoparticle exposure and the onset of neurodegenerative conditions, highlighting the imperative for comprehensive research and strategic interventions. Gaining a deep understanding of these mechanisms and enacting protective policies are crucial steps toward reducing the health threats of nanoparticles, thereby maximizing their therapeutic advantages.


Subject(s)
Nanoparticles , Neurodegenerative Diseases , Humans , Nanoparticles/toxicity , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Environmental Exposure/adverse effects
12.
J Nanobiotechnology ; 22(1): 251, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750597

ABSTRACT

BACKGROUND: Hypothermia is a promising therapy for traumatic brain injury (TBI) in the clinic. However, the neuroprotective outcomes of hypothermia-treated TBI patients in clinical studies are inconsistent due to several severe side effects. Here, an injectable refrigerated hydrogel was designed to deliver 3-iodothyronamine (T1AM) to achieve a longer period of local hypothermia for TBI treatment. Hydrogel has four advantages: (1) It can be injected into injured sites after TBI, where it forms a hydrogel and avoids the side effects of whole-body cooling. (2) Hydrogels can biodegrade and be used for controlled drug release. (3) Released T1AM can induce hypothermia. (4) This hydrogel has increased medical value given its simple operation and ability to achieve timely treatment. METHODS: Pol/T hydrogels were prepared by a low-temperature mixing method and characterized. The effect of the Pol/T hydrogel on traumatic brain injury in mice was studied. The degradation of the hydrogel at the body level was observed with a small animal imager. Brain temperature and body temperature were measured by brain thermometer and body thermometer, respectively. The apoptosis of peripheral nerve cells was detected by immunohistochemical staining. The protective effect of the hydrogels on the blood-brain barrier (BBB) after TBI was evaluated by the Evans blue penetration test. The protective effect of hydrogel on brain edema after injury in mice was detected by Magnetic resonance (MR) in small animals. The enzyme linked immunosorbent assay (ELISA) method was used to measure the levels of inflammatory factors. The effects of behavioral tests on the learning ability and exercise ability of mice after injury were evaluated. RESULTS: This hydrogel was able to cool the brain to hypothermia for 12 h while maintaining body temperature within the normal range after TBI in mice. More importantly, hypothermia induced by this hydrogel leads to the maintenance of BBB integrity, the prevention of cell death, the reduction of the inflammatory response and brain edema, and the promotion of functional recovery after TBI in mice. This cooling method could be developed as a new approach for hypothermia treatment in TBI patients. CONCLUSION: Our study showed that injectable and biodegradable frozen Pol/T hydrogels to induce local hypothermia in TBI mice can be used for the treatment of traumatic brain injury.


Subject(s)
Blood-Brain Barrier , Brain Injuries, Traumatic , Hydrogels , Hypothermia, Induced , Animals , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/drug therapy , Mice , Hydrogels/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Male , Hypothermia, Induced/methods , Neuroprotection/drug effects , Brain/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Body Temperature , Mice, Inbred C57BL
13.
Front Immunol ; 15: 1386780, 2024.
Article in English | MEDLINE | ID: mdl-38756773

ABSTRACT

Introduction: Intracerebral hemorrhage (ICH) often triggers oxidative stress through reactive oxygen species (ROS). Transforming growth factor-ß-activated kinase 1 (TAK1) plays a pivotal role in regulating oxidative stress and inflammation across various diseases. 5Z-7-Oxozeaenol (OZ), a specific inhibitor of TAK1, has exhibited therapeutic effects in various conditions. However, the impact of OZ following ICH and its underlying molecular mechanisms remain elusive. This study aimed to explore the possible role of OZ in ICH and its underlying mechanisms by inhibiting oxidative stress-mediated pyroptosis. Methods: Adult male Sprague-Dawley rats were subjected to an ICH model, followed by treatment with OZ. Neurobehavioral function, blood-brain barrier integrity, neuronal pyroptosis, and oxidative stress markers were assessed using various techniques including behavioral tests, immunofluorescence staining, western blotting, transmission electron microscopy, and biochemical assays. Results: Our study revealed that OZ administration significantly inhibited phosphorylated TAK1 expression post-ICH. Furthermore, TAK1 blockade by OZ attenuated blood-brain barrier (BBB) disruption, neuroinflammation, and oxidative damage while enhancing neurobehavioral function. Mechanistically, OZ administration markedly reduced ROS production and oxidative stress by facilitating nuclear factor-erythroid 2-related factor 2 (NRF2) nuclear translocation. This was accompanied by a subsequent suppression of the NOD-like receptor protein 3 (NLRP3) activation-mediated inflammatory cascade and neuronal pyroptosis. Discussion: Our findings highlight that OZ alleviates brain injury and oxidative stress-mediated pyroptosis via the NRF2 pathway. Inhibition of TAK1 emerges as a promising approach for managing ICH.


Subject(s)
Cerebral Hemorrhage , MAP Kinase Kinase Kinases , NF-E2-Related Factor 2 , Neurons , Oxidative Stress , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Animals , Pyroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Male , Rats , Signal Transduction/drug effects , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Disease Models, Animal , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/drug therapy , Reactive Oxygen Species/metabolism , Lactones , Resorcinols , Zearalenone/administration & dosage
14.
Proc Natl Acad Sci U S A ; 121(20): e2318119121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709930

ABSTRACT

Brain metastasis of advanced breast cancer often results in deleterious consequences. Metastases to the brain lead to significant challenges in treatment options, as the blood-brain barrier (BBB) prevents conventional therapy. Thus, we hypothesized that creation of a nanoparticle (NP) that distributes to both primary tumor site and across the BBB for secondary brain tumor can be extremely beneficial. Here, we report a simple targeting strategy to attack both the primary breast and secondary brain tumors utilizing a single NP platform. The nature of these mitochondrion-targeted, BBB-penetrating NPs allow for simultaneous targeting and drug delivery to the hyperpolarized mitochondrial membrane of the extracranial primary tumor site in addition to tumors at the brain. By utilizing a combination of such dual anatomical distributing NPs loaded with therapeutics, we demonstrate a proof-of-concept idea to combat the increased metabolic plasticity of brain metastases by lowering two major energy sources, oxidative phosphorylation (OXPHOS) and glycolysis. By utilizing complementary studies and genomic analyses, we demonstrate the utility of a chemotherapeutic prodrug to decrease OXPHOS and glycolysis by pairing with a NP loaded with pyruvate dehydrogenase kinase 1 inhibitor. Decreasing glycolysis aims to combat the metabolic flexibility of both primary and secondary tumors for therapeutic outcome. We also address the in vivo safety parameters by addressing peripheral neuropathy and neurobehavior outcomes. Our results also demonstrate that this combination therapeutic approach utilizes mitochondrial genome targeting strategy to overcome DNA repair-based chemoresistance mechanisms.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Breast Neoplasms , Nanoparticles , Oxidative Phosphorylation , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain Neoplasms/pathology , Animals , Humans , Female , Nanoparticles/chemistry , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Oxidative Phosphorylation/drug effects , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/drug effects , Drug Delivery Systems/methods , Glycolysis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use
15.
ACS Chem Neurosci ; 15(10): 2028-2041, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38710594

ABSTRACT

Chronic cerebral hypoperfusion (CCH)-triggered blood-brain barrier (BBB) dysfunction is a core pathological change occurring in vascular dementia (VD). Despite the recent advances in the exploration of the structural basis of BBB impairment and the routes of entry of harmful compounds after a BBB leakage, the molecular mechanisms inducing BBB impairment remain largely unknown in terms of VD. Here, we employed a CCH-induced VD model and discovered increased vascular cell adhesion molecule 1 (VCAM1) expression on the brain endothelial cells (ECs). The expression of VCAM1 was directly correlated with the severity of BBB impairment. Moreover, the VCAM1 expression was associated with different regional white matter lesions. Furthermore, a compound that could block VCAM1 activation, K-7174, was also found to alleviate BBB leakage and protect the white matter integrity, whereas pharmacological manipulation of the BBB leakage did not affect the VCAM1 expression. Thus, our results demonstrated that VCAM1 is an important regulator that leads to BBB dysfunction following CCH. Blocking VCAM1-mediated BBB impairment may thus offer a new strategy to treat CCH-related neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Vascular Cell Adhesion Molecule-1 , Vascular Cell Adhesion Molecule-1/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Animals , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Brain/metabolism , Brain/pathology , Dementia, Vascular/metabolism , Dementia, Vascular/pathology , Humans , Brain Ischemia/metabolism , Brain Ischemia/pathology , Mice
16.
Sci Total Environ ; 935: 173249, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38754502

ABSTRACT

Selenium (Se), a highly beneficial animal feed additive, exhibits remarkable antioxidant and anti-inflammatory properties. Nano­selenium (Nano-Se) is an advanced formulation of Se featuring a specialized drug delivery vehicle, with good bioavailability, higher efficacy, and lower toxicity compared to the traditional form of Se. With the advancement of industry, cadmium (Cd) contamination occurs in different countries and regions and thereby contaminating different food crops, and the degree of pollution is degree increasing year by year. The present investigation entailed the oral administration of CdCl2 and/or Nano-Se to male chickens of the Hy-Line Variety White breed, which are one day old, subsequent to a 7-day adaptive feeding period, for a duration of 90 days. The study aimed to elucidate the potential protective impact of Nano-Se on Cd exposure. The study found that Nano-Se demonstrates potential in mitigating the blood-brain barrier (BBB) dysfunction characterized by impairment of adherens junctions (AJS) and tight junctions (TJS) by inhibiting reactive oxygen species (ROS) overproduction. In addition, the data uncovered that Nano-Se demonstrates a proficient ability in alleviating BBB impairment and inflammatory reactions caused by Cd through the modulation of the Wnt7A/ß-catenin pathway, highlights its potential to maintain brain homeostasis. Hence, this research anticipates that the utilization of Nano-Se effectively mitigate the detrimental impacts associated with Cd exposure on the BBB.


Subject(s)
Blood-Brain Barrier , Cadmium , Chickens , Selenium , Animals , Cadmium/toxicity , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Male , beta Catenin/metabolism , Wnt Signaling Pathway/drug effects
17.
Eur J Med Chem ; 272: 116463, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38704944

ABSTRACT

Butyrylcholinesterase (BChE) has attracted wide interest as a promising target in Alzheimer's disease (AD) investigation. BChE is considered to play a compensable role of hydrolyzing acetylcholine (ACh), and its positive correlation with ß-amyloid (Aß) deposition also promotes disease progression. Herein, we uncovered a selective potent BChE inhibitor S21-1011 (eqBChE IC50 = 0.059 ± 0.006 µM, hBChE IC50 = 0.162 ± 0.069 µM), which presented satisfactory druggability and therapeutic efficacy in AD models. In pharmacokinetics (PK) studies, S21-1011 showed excellent blood-brain barrier (BBB) permeability, metabolism stability and high oral-bioavailability. In pharmacodynamic (PD) studies, it protected neural cells from toxicity and inflammation stimulation in vitro. Besides, it also exerted anti-inflammatory effect and alleviated cognitive impairment in mice models induced by lipopolysaccharides (LPS) and Aß. Generally, this compound has been confirmed to function as a neuroprotector and cognition improver in various AD pathology-like models. Therefore, S21-1011, a novel potent BChE inhibitor, could be considered as a potential anti-AD candidate worthy of more profound investigation.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Cholinesterase Inhibitors , Quinolines , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/chemical synthesis , Mice , Humans , Structure-Activity Relationship , Quinolines/chemistry , Quinolines/pharmacology , Quinolines/chemical synthesis , Drug Discovery , Molecular Structure , Male , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Dose-Response Relationship, Drug , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/chemical synthesis , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Inflammation/drug therapy , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
18.
Eur J Med Chem ; 272: 116457, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38704941

ABSTRACT

It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.


Subject(s)
Brain Ischemia , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Brain Ischemia/drug therapy , Animals , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Molecular Structure
19.
ACS Nano ; 18(22): 14469-14486, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38770948

ABSTRACT

Glioblastoma (GBM) is a lethal brain tumor with high levels of malignancy. Most chemotherapy agents show serious systemic cytotoxicity and restricted delivery effectiveness due to the impediments of the blood-brain barrier (BBB). Immunotherapy has developed great potential for aggressive tumor treatments. Disappointingly, its efficacy against GBM is hindered by the immunosuppressive tumor microenvironment (TME) and BBB. Herein, a multiple synergistic immunotherapeutic strategy against GBM was developed based on the nanomaterial-biology interaction. We have demonstrated that this BM@MnP-BSA-aPD-1 can transverse the BBB and target the TME, resulting in amplified synergetic effects of metalloimmunotherapy and photothermal immunotherapy (PTT). The journey of this nanoformulation within the TME contributed to the activation of the stimulator of the interferon gene pathway, the initiation of the immunogenic cell death effect, and the inhibition of the programmed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1) signaling axis. This nanomedicine revitalizes the immunosuppressive TME and evokes the cascade effect of antitumor immunity. Therefore, the combination of BM@MnP-BSA-aPD-1 and PTT without chemotherapeutics presents favorable benefits in anti-GBM immunotherapy and exhibits immense potential for clinical translational applications.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunotherapy , Microglia , Tumor Microenvironment , Glioblastoma/therapy , Glioblastoma/pathology , Glioblastoma/immunology , Glioblastoma/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Humans , Animals , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Photothermal Therapy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism
20.
Neurosci Lett ; 833: 137828, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38772437

ABSTRACT

There is a critical need for safer and better-tolerated alternatives to address the current limitations of antidepressant treatments for major depressive disorder. Recently, drugs targeting the GABA system via α5-containing GABAA receptors (α5-GABAAR) as negative allosteric modulators (α5-NAMs) have shown promise in alleviating stress-related behaviors in preclinical studies, suggesting that α5-NAMs may have translational relevance as novel antidepressant medications. Here, we evaluated the efficacy of Basmisanil, an α5-NAM that has been evaluated in Phase 2 clinical studies as a cognitive enhancer, in a battery of behavioral tests relevant to coping strategies, motivation, and aversion in male mice, along with plasma and brain pharmacokinetic measurements. Our findings reveal that Basmisanil induces dose-dependent rapid antidepressant-like responses in the forced swim test and sucrose splash test without promoting locomotor stimulating effects. Furthermore, Basmisanil elicits sustained behavioral responses in the female urine sniffing test and sucrose splash test, observed 24 h and 48 h post-treatment, respectively. Bioanalysis of plasma and brain samples confirms effective blood-brain barrier penetration by Basmisanil and extrapolation to previously published data suggest that effects were observed at doses (10 and 30 mg/kg i.p.) corresponding to relatively modest levels of α5-GABAAR occupancy (40-65 %). These results suggest that Basmisanil exhibits a combination of rapid and sustained antidepressant-like effects highlighting the potential of α5-NAMs as a novel therapeutic strategy for depression.


Subject(s)
Antidepressive Agents , Receptors, GABA-A , Animals , Male , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/pharmacokinetics , Mice , Brain/metabolism , Brain/drug effects , Female , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...