Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Control Release ; 371: 216-236, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38810705

ABSTRACT

Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.

2.
J Neurotrauma ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38818807

ABSTRACT

The kallikrein-kinin system is one of the first inflammatory pathways to be activated following traumatic brain injury (TBI) and has been shown to exacerbate brain edema formation in the acute phase through activation of Bradykinin-2-receptors (B2R). However, the influence of B2 receptors on chronic posttraumatic damage and outcome is unclear. In the current study we assessed long term effects of B2R-knockout after experimental traumatic brain injury. B2R knockout mice (heterozygous, homozygous) and wildtype littermates (n=10/group) were subjected to controlled cortical impact TBI. Lesion size was evaluated by MRI up to 90 days after CCI. Motor and memory function were regularly assessed by Neurological severity Score (NSS), Beam Walk (BW), and Barnes Maze test. 90 days after TBI, brains were harvested for immunohistochemical analysis. There was no difference in cortical lesion size between B2R deficient and wildtype animals three months after injury, however, hippocampal damage was reduced in B2R KO mice (p=0.03). Protection of hippocampal tissue was accompanied by a significant improvement of learning and memory function three months after TBI (p=0.02 WT vs. KO), whereas motor function was not influenced. Scar formation and astrogliosis were unaffected, but bradykinin-2-receptor deficiency led to a gene-dose dependent attenuation of microglial activation and a reduction of CD45+ cells three months after TBI in cortex (p=0.0003) and hippocampus (p< 0.0001). These results suggest that chronic hippocampal neurodegeneration and subsequent cognitive impairment is mediated by prolonged neuroinflammation and bradykinin-2-receptors. Inhibition of B2-receptors may therefore represent a novel strategy to reduce long-term neurocognitive deficits after TBI.

3.
Brain ; 147(3): 1057-1074, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38153327

ABSTRACT

Incomplete reperfusion of the microvasculature ('no-reflow') after ischaemic stroke damages salvageable brain tissue. Previous ex vivo studies suggest pericytes are vulnerable to ischaemia and may exacerbate no-reflow, but the viability of pericytes and their association with no-reflow remains under-explored in vivo. Using longitudinal in vivo two-photon single-cell imaging over 7 days, we showed that 87% of pericytes constrict during cerebral ischaemia and remain constricted post reperfusion, and 50% of the pericyte population are acutely damaged. Moreover, we revealed ischaemic pericytes to be fundamentally implicated in capillary no-reflow by limiting and arresting blood flow within the first 24 h post stroke. Despite sustaining acute membrane damage, we observed that over half of all cortical pericytes survived ischaemia and responded to vasoactive stimuli, upregulated unique transcriptomic profiles and replicated. Finally, we demonstrated the delayed recovery of capillary diameter by ischaemic pericytes after reperfusion predicted vessel reconstriction in the subacute phase of stroke. Cumulatively, these findings demonstrate that surviving cortical pericytes remain both viable and promising therapeutic targets to counteract no-reflow after ischaemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Humans , Pericytes/physiology , Cerebral Infarction
4.
Cell ; 186(17): 3706-3725.e29, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37562402

ABSTRACT

The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.


Subject(s)
Bone Marrow , Nervous System Diseases , Skull , Animals , Humans , Mice , Bone Marrow/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carrier Proteins/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Positron-Emission Tomography/methods , Receptors, GABA/metabolism , Skull/cytology , Skull/diagnostic imaging
5.
Stroke ; 54(8): 2126-2134, 2023 08.
Article in English | MEDLINE | ID: mdl-37325921

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) is characterized by acute and delayed reductions of cerebral blood flow (CBF) caused, among others, by spasms of cerebral arteries and arterioles. Recently, the inactivation of perivascular macrophages (PVM) has been demonstrated to improve neurological outcomes after experimental SAH, but the underlying mechanisms of protection remain unclear. The aim of our exploratory study was, therefore, to investigate the role of PVM in the formation of acute microvasospasms after experimental SAH. METHODS: PVMs were depleted in 8- to 10-week-old male C57BL/6 mice (n=8/group) by intracerebroventricular application of clodronate-loaded liposomes and compared with mice with vehicle liposome injections. Seven days later, SAH was induced by filament perforation under continuous monitoring of CBF and intracranial pressure. Results were compared with sham-operated animals and animals who underwent SAH induction but no liposome injection (n=4/group each). Six hours after SAH induction or sham surgery, numbers of microvasospasms per volume of interest and % of affected pial and penetrating arterioles were examined in 9 standardized regions of interest per animal by in vivo 2-photon microscopy. Depletion of PVMs was proven by quantification of PVMs/mm3 identified by immunohistochemical staining for CD206 and Collagen IV. Statistical significance was tested with t tests for parametric data and Mann-Whitney U test for nonparametric data. RESULTS: PVMs were located around pial and intraparenchymal arterioles and were effectively depleted by clodronate from 671±28 to 46±14 PVMs/mm3 (P<0.001). After SAH, microvasospasms was observed in pial arteries and penetrating and precapillary arterioles and were accompanied by an increase to 1405±142 PVMs/mm3. PVM depletion significantly reduced the number of microvasospasms from 9 IQR 5 to 3 IQR 3 (P<0.001). CONCLUSIONS: Our results suggest that PVMs contribute to the formation of microvasospasms after experimental SAH.


Subject(s)
Subarachnoid Hemorrhage , Mice , Male , Animals , Subarachnoid Hemorrhage/complications , Clodronic Acid , Mice, Inbred C57BL , Arterioles , Cerebrovascular Circulation/physiology , Disease Models, Animal
6.
Small ; 18(18): e2200302, 2022 05.
Article in English | MEDLINE | ID: mdl-35384294

ABSTRACT

The current lack of understanding about how nanocarriers cross the blood-brain barrier (BBB) in the healthy and injured brain is hindering the clinical translation of nanoscale brain-targeted drug-delivery systems. Here, the bio-distribution of lipid nano-emulsion droplets (LNDs) of two sizes (30 and 80 nm) in the mouse brain after traumatic brain injury (TBI) is investigated. The highly fluorescent LNDs are prepared by loading them with octadecyl rhodamine B and a bulky hydrophobic counter-ion, tetraphenylborate. Using in vivo two-photon and confocal imaging, the circulation kinetics and bio-distribution of LNDs in the healthy and injured mouse brain are studied. It is found that after TBI, LNDs of both sizes accumulate at vascular occlusions, where specifically 30 nm LNDs extravasate into the brain parenchyma and reach neurons. The vascular occlusions are not associated with bleedings, but instead are surrounded by processes of activated microglia, suggesting a specific opening of the BBB. Finally, correlative light-electron microscopy reveals 30 nm LNDs in endothelial vesicles, while 80 nm particles remain in the vessel lumen, indicating size-selective vesicular transport across the BBB via vascular occlusions. The data suggest that microvascular occlusions serve as "gates" for the transport of nanocarriers across the BBB.


Subject(s)
Brain Injuries, Traumatic , Nanoparticles , Animals , Blood-Brain Barrier , Brain , Drug Carriers/chemistry , Liposomes , Mice , Nanoparticles/chemistry
7.
Nanomedicine ; 40: 102511, 2022 02.
Article in English | MEDLINE | ID: mdl-34915181

ABSTRACT

The potential of poly(lactic-co-glycolic acid) (PLGA) to design nanoparticles (NPs) and target the central nervous system remains to be exploited. In the current study we designed fluorescent 70-nm PLGA NPs, loaded with bulky fluorophores, thereby making them significantly brighter than quantum dots in single-particle fluorescence measurements. The high brightness of NPs enabled their visualization by intravital real-time 2-photon microscopy. Subsequently, we found that PLGA NPs coated with pluronic F-68 circulated in the blood substantially longer than uncoated NPs and were taken up by cerebro-vascular endothelial cells. Additionally, confocal microscopy revealed that coated PLGA NPs were present in late endothelial endosomes of cerebral vessels within 1 h after systemic injection and were more readily taken up by endothelial cells in peripheral organs. The combination of ultra-bright NPs and in vivo imaging may thus represent a promising approach to reduce the gap between development and clinical application of nanoparticle-based drug carriers.


Subject(s)
Nanoparticles , Poloxamer , Drug Carriers , Endothelial Cells , Glycols , Microscopy , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer
8.
Acta Neuropathol Commun ; 9(1): 138, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404478

ABSTRACT

Traumatic brain injury (TBI) causes acute and subacute tissue damage, but is also associated with chronic inflammation and progressive loss of brain tissue months and years after the initial event. The trigger and the subsequent molecular mechanisms causing chronic brain injury after TBI are not well understood. The aim of the current study was therefore to investigate the hypothesis that necroptosis, a form a programmed cell death mediated by the interaction of Receptor Interacting Protein Kinases (RIPK) 1 and 3, is involved in this process. Neuron-specific RIPK1- or RIPK3-deficient mice and their wild-type littermates were subjected to experimental TBI by controlled cortical impact. Posttraumatic brain damage and functional outcome were assessed longitudinally by repetitive magnetic resonance imaging (MRI) and behavioral tests (beam walk, Barnes maze, and tail suspension), respectively, for up to three months after injury. Thereafter, brains were investigated by immunohistochemistry for the necroptotic marker phosphorylated mixed lineage kinase like protein(pMLKL) and activation of astrocytes and microglia. WT mice showed progressive chronic brain damage in cortex and hippocampus and increased levels of pMLKL after TBI. Chronic brain damage occurred almost exclusively in areas with iron deposits and was significantly reduced in RIPK1- or RIPK3-deficient mice by up to 80%. Neuroprotection was accompanied by a reduction of astrocyte and microglia activation and improved memory function. The data of the current study suggest that progressive chronic brain damage and cognitive decline after TBI depend on the expression of RIPK1/3 in neurons. Hence, inhibition of necroptosis signaling may represent a novel therapeutic target for the prevention of chronic post-traumatic brain damage.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/genetics , Brain/metabolism , Microglia/metabolism , Necroptosis/genetics , Neurons/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Brain/diagnostic imaging , Brain/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Brain Injury, Chronic/genetics , Brain Injury, Chronic/metabolism , Brain Injury, Chronic/pathology , Brain Injury, Chronic/physiopathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hindlimb Suspension , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/pathology , Magnetic Resonance Imaging , Maze Learning , Memory , Mice , Mice, Knockout , Neurons/pathology , Protein Kinases/metabolism
9.
J Neurotrauma ; 38(11): 1572-1584, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33779289

ABSTRACT

Traumatic brain injury (TBI) causes long-lasting neurodegeneration and cognitive impairments; however, the underlying mechanisms of these processes are not fully understood. Acid-sensing ion channels 1a (ASIC1a) are voltage-gated Na+- and Ca2+-channels shown to be involved in neuronal cell death; however, their role for chronic post-traumatic brain damage is largely unknown. To address this issue, we used ASIC1a-deficient mice and investigated their outcome up to 6 months after TBI. ASIC1a-deficient mice and their wild-type (WT) littermates were subjected to controlled cortical impact (CCI) or sham surgery. Brain water content was analyzed 24 h and behavioral outcome up to 6 months after CCI. Lesion volume was assessed longitudinally by magnetic resonance imaging and 6 months after injury by histology. Brain water content was significantly reduced in ASIC1a-/- animals compared to WT controls. Over time, ASIC1a-/- mice showed significantly reduced lesion volume and reduced hippocampal damage. This translated into improved cognitive function and reduced depression-like behavior. Microglial activation was significantly reduced in ASIC1a-/- mice. In conclusion, ASIC1a deficiency resulted in reduced edema formation acutely after TBI and less brain damage, functional impairments, and neuroinflammation up to 6 months after injury. Hence, ASIC1a seems to be involved in chronic neurodegeneration after TBI.


Subject(s)
Acid Sensing Ion Channels/deficiency , Brain Damage, Chronic/etiology , Brain Damage, Chronic/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Animals , Brain Damage, Chronic/psychology , Brain Injuries, Traumatic/psychology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Motor Activity
10.
ACS Nano ; 14(8): 9755-9770, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32680421

ABSTRACT

Visualizing single organic nanoparticles (NPs) in vivo remains a challenge, which could greatly improve our understanding of the bottlenecks in the field of nanomedicine. To achieve high single-particle fluorescence brightness, we loaded polymer poly(methyl methacrylate)-sulfonate (PMMA-SO3H) NPs with octadecyl rhodamine B together with a bulky hydrophobic counterion (perfluorinated tetraphenylborate) as a fluorophore insulator to prevent aggregation-caused quenching. To create NPs with stealth properties, we used the amphiphilic block copolymers pluronic F-127 and F-68. Fluorescence correlation spectroscopy and Förster resonance energy transfer (FRET) revealed that pluronics remained at the NP surface after dialysis (at one amphiphile per 5.5 nm2) and prevented NPs from nonspecific interactions with serum proteins and surfactants. In primary cultured neurons, pluronics stabilized the NPs, preventing their prompt aggregation and binding to neurons. By increasing dye loading to 20 wt % and optimizing particle size, we obtained 74 nm NPs showing 150-fold higher single-particle brightness with two-photon excitation than commercial Nile Red-loaded FluoSpheres of 39 nm hydrodynamic diameter. The obtained ultrabright pluronic-coated NPs enabled direct single-particle tracking in vessels of mice brains by two-photon intravital microscopy for at least 1 h, whereas noncoated NPs were rapidly eliminated from the circulation. Following brain injury or neuroinflammation, which can open the blood-brain barrier, extravasation of NPs was successfully monitored. Moreover, we demonstrated tracking of individual NPs from meningeal vessels until their uptake by meningeal macrophages. Thus, single NPs can be tracked in animals in real time in vivo in different brain compartments and their dynamics visualized with subcellular resolution.


Subject(s)
Nanoparticles , Poloxamer , Animals , Brain , Fluorescent Dyes , Mice , Particle Size , Polymers
11.
Drug Deliv ; 23(9): 3520-3528, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27278330

ABSTRACT

Currently, traumatic brain injury (TBI) is the leading cause of death or disabilities in young individuals worldwide. The multi-complexity of its pathogenesis as well as impermeability of the blood-brain barrier (BBB) makes the drug choice and delivery very challenging. The brain-derived neurotrophic factor (BDNF) regulates neuronal plasticity, neuronal cell growth, proliferation, cell survival and long-term memory. However, its short half-life and low BBB permeability are the main hurdles to be an effective therapeutic for TBI. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles coated by surfactant can enable the delivery of a variety of molecules across the BBB by receptor-mediated transcytosis. This study examines the ability of PLGA nanoparticles coated with poloxamer 188 (PX) to deliver BDNF into the brain and neuroprotective effects of BNDF in mice with TBI. C57bl/6 mice were subjected to weight-drop closed head injuries under anesthesia. Using enzyme-linked immunosorbent assay, we demonstrated that the intravenous (IV) injection of nanoparticle-bound BDNF coated by PX (NP-BDNF-PX) significantly increased BDNF levels in the brain of sham-operated mice (p < 0.001) and in both ipsi- (p < 0.001) and contralateral (p < 0.001) parts of brain in TBI mice compared to controls. This study also showed using the passive avoidance (PA) test, that IV injection of NP-BDNF-PX 3 h post-injury prolonged the latent time in mice with TBI thereby reversing cognitive deficits caused by brain trauma. Finally, neurological severity score test demonstrated that our compound efficiently reduced the scores at day 7 after the injury indicating the improvement of neurological deficit in animals with TBI. This study shows that PLGA nanoparticles coated with PX effectively delivered BDNF into the brain, and improved neurological and cognitive deficits in TBI mice, thereby providing a neuroprotective effect.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain-Derived Neurotrophic Factor/administration & dosage , Brain/drug effects , Cognition/drug effects , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Animals , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/chemistry , Half-Life , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Poloxamer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Surface-Active Agents/chemistry
12.
Neural Regen Res ; 11(4): 630-5, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27212925

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability in individuals worldwide. Producing a clinically relevant TBI model in small-sized animals remains fairly challenging. For good screening of potential therapeutics, which are effective in the treatment of TBI, animal models of TBI should be established and standardized. In this study, we established mouse models of closed head injury using the Shohami weight-drop method with some modifications concerning cognitive deficiency assessment and provided a detailed description of the severe TBI animal model. We found that 250 g falling weight from 2 cm height produced severe closed head injury in C57BL/6 male mice. Cognitive disorders in mice with severe closed head injury could be detected using passive avoidance test on day 7 after injury. Findings from this study indicate that weight-drop injury animal models are suitable for further screening of brain neuroprotectants and potentially are similar to those seen in human TBI.

13.
Int J Nanomedicine ; 10: 3245-67, 2015.
Article in English | MEDLINE | ID: mdl-25995632

ABSTRACT

Neurodegenerative causes of blindness and deafness possess a major challenge in their clinical management as proper treatment guidelines have not yet been found. Brain-derived neurotrophic factor (BDNF) has been established as a promising therapy against neurodegenerative disorders including hearing and visual loss. Unfortunately, the blood-retinal barrier and blood-cochlear barrier, which have a comparable structure to the blood-brain barrier prevent molecules of larger sizes (such as BDNF) from exiting the circulation and reaching the targeted cells. Anatomical features of the eye and ear allow use of local administration, bypassing histo-hematic barriers. This paper focuses on highlighting a variety of strategies proposed for the local administration of the BDNF, like direct delivery, viral gene therapy, and cell-based therapy, which have been shown to successfully improve development, survival, and function of spiral and retinal ganglion cells. The similarities and controversies for BDNF treatment of posterior eye diseases and inner ear diseases have been analyzed and compared. In this review, we also focus on the possibility of translation of this knowledge into clinical practice. And finally, we suggest that using nanoparticulate drug-delivery systems may substantially contribute to the development of clinically viable techniques for BDNF delivery into the cochlea or posterior eye segment, which, ultimately, can lead to a long-term or permanent rescue of auditory and optic neurons from degeneration.


Subject(s)
Blindness , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor , Deafness , Drug Delivery Systems , Animals , Blindness/drug therapy , Blindness/physiopathology , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Line , Deafness/drug therapy , Deafness/physiopathology , Humans , Mice
14.
Int J Nanomedicine ; 9: 795-811, 2014.
Article in English | MEDLINE | ID: mdl-24550672

ABSTRACT

The protective properties of the blood-brain barrier (BBB) are conferred by the intricate architecture of its endothelium coupled with multiple specific transport systems expressed on the surface of endothelial cells (ECs) in the brain's vasculature. When the stringent control of the BBB is disrupted, such as following EC damage, substances that are safe for peripheral tissues but toxic to neurons have easier access to the central nervous system (CNS). As a consequence, CNS disorders, including degenerative diseases, can occur independently of an individual's age. Although the BBB is crucial in regulating the biochemical environment that is essential for maintaining neuronal integrity, it limits drug delivery to the CNS. This makes it difficult to deliver beneficial drugs across the BBB while preventing the passage of potential neurotoxins. Available options include transport of drugs across the ECs through traversing occludins and claudins in the tight junctions or by attaching drugs to one of the existing transport systems. Either way, access must specifically allow only the passage of a particular drug. In general, the BBB allows small molecules to enter the CNS; however, most drugs with the potential to treat neurological disorders other than infections have large structures. Several mechanisms, such as modifications of the built-in pumping-out system of drugs and utilization of nanocarriers and liposomes, are among the drug-delivery systems that have been tested; however, each has its limitations and constraints. This review comprehensively discusses the functional morphology of the BBB and the challenges that must be overcome by drug-delivery systems and elaborates on the potential targets, mechanisms, and formulations to improve drug delivery to the CNS.


Subject(s)
Blood-Brain Barrier , Drug Delivery Systems , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Animals , Biological Transport, Active , Blood-Brain Barrier/anatomy & histology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Chemistry, Pharmaceutical , Colloids , Humans , Liposomes , Nanomedicine , Nanotechnology
15.
Int J Radiat Biol ; 89(12): 1108-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23786463

ABSTRACT

PURPOSE: The increase in the incidence of the radiation-induced skin injury cases and the absence of standard treatments escalate the interest in finding new and effective drugs for these lesions. We studied the effect of a 40% solution of arginine glutamate on the healing of radiation-induced skin ulcers in guinea pigs. MATERIALS AND METHODS: Radiation skin injury was produced on the thigh of guinea pigs by 60 Gy local X-ray irradiation. Treatment was started 6 weeks after the irradiation when ulcers had been formed. Arginine glutamate was administered by subcutaneous injections around the wound edge. Methyluracil was chosen as the comparison drug. The animals were sacrificed on day 21 after the start of treatment and the irradiated skin tissues were subjected to histological evaluation, cytokines analysis, lipid peroxidation and antioxidant enzymes analysis. RESULTS: We have shown that arginine glutamate significantly (p < 0.05) decreased levels of pro-inflammatory cytokines in the wound, restored the balance between lipid peroxidation formation and antioxidant enzymes activity and promoted cell proliferation as well as collagen synthesis. CONCLUSIONS: These results demonstrate that arginine glutamate successfully improves the healing of radiation-induced skin ulcers. In all probability, the curative effect is associated with the interaction of arginine with nitric oxide synthase II and arginase I, but further investigations are needed to validate this.


Subject(s)
Dipeptides/therapeutic use , Radiation Injuries/drug therapy , Radiation Injuries/metabolism , Skin Ulcer/drug therapy , Animals , Antioxidants/metabolism , Arginase/metabolism , Cytokines/metabolism , Guinea Pigs , Lipid Peroxidation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Skin/radiation effects , Skin Ulcer/etiology , Uracil/analogs & derivatives , Uracil/therapeutic use , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...