1
|
Chen G, Douglas HF, Li Z, Cleveland WJ, Balzer C, Yannopoulos D, Chen IY, Obal D, Riess ML. Cardioprotection by poloxamer 188 is mediated through increased endothelial nitric oxide production. Sci Rep 2025; 15:15170. [PMID: 40307302 PMCID: PMC12043958 DOI: 10.1038/s41598-025-97079-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Ischemia/reperfusion (I/R) injury significantly contributes to the morbidity and mortality associated with cardiac events. Poloxamer 188 (P188), a non-ionic triblock copolymer, has been proposed to mitigate I/R injury by stabilizing cell membranes. However, the underlying mechanisms remain incompletely understood, particularly concerning endothelial cell (EC) function and nitric oxide (NO) production. We employed human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) and ECs to elucidate the effects of P188 on cellular survival, function, and NO secretion under simulated I/R conditions. iPSC-CMs contractility and iPSC-ECs' NO production were assessed following exposure to P188. Further, an isolated heart model using Brown Norway rats subjected to I/R injury was utilized to evaluate the ex-vivo cardioprotective effects of P188, examining cardiac function and NO production, with and without the administration of a NO inhibitor. In iPSC-derived models, P188 significantly preserved CM contractile function and enhanced cell viability after hypoxia/reoxygenation. Remarkably, P188 treatment led to a pronounced increase in NO secretion in iPSC-ECs, a novel finding demonstrating endothelial protective effects beyond membrane stabilization. In the rat isolated heart model, administration of P188 during reperfusion notably improved cardiac function and reduced I/R injury markers. This cardioprotective effect was abrogated by NO inhibition, underscoring the pivotal role of NO. Additionally, a dose-dependent increase in NO production was observed in non-ischemic rat hearts treated with P188, further establishing the critical function of NO in P188 induced cardioprotection. In conclusion, our comprehensive study unveils a novel role of NO in mediating the protective effects of P188 against I/R injury. This mechanism is evident in both cellular models and intact rat hearts, highlighting the potential of P188 as a therapeutic agent against I/R injury. Our findings pave the way for further investigation into P188's therapeutic mechanisms and its potential application in clinical settings to mitigate I/R-related cardiac dysfunction.
Collapse
Affiliation(s)
- Gaoxian Chen
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Hunter F Douglas
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zhu Li
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William J Cleveland
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Claudius Balzer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Demetris Yannopoulos
- Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ian Y Chen
- Departments of Medicine (Cardiovascular Medicine) and Radiology, Stanford University School of Medicine, Stanford, CA, USA
- Medical (Cardiology) and Radiology Services, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Detlef Obal
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Department of Anesthesiology, University of Iowa, Iowa, IA, USA.
| | - Matthias L Riess
- Department of Anesthesiology, TVHS VA Medical Center, Nashville, TN, USA.
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Mondkar PP, Seo HS, Lodge TP, Azarin SM. Diblock Copolymers of Poly(ethylene oxide)- b-poly(propylene oxide) Stabilize a Blood-Brain Barrier Model under Oxidative Stress. Mol Pharm 2024; 21:5646-5660. [PMID: 39400078 DOI: 10.1021/acs.molpharmaceut.4c00608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The blood-brain barrier (BBB) is a highly restrictive barrier at the interface between the brain and the vascular system. Even under BBB dysfunction, it is extremely difficult to deliver therapies across the barrier, limiting the options for treatment of neurological injuries and disorders. To circumvent these challenges, there is interest in developing therapies that directly engage with the damaged BBB to restore its function. Previous studies revealed that poloxamer 188 (P188), a water-soluble triblock copolymer of poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO), partially mitigated BBB dysfunction in vivo. In the context of stabilization of the damaged BBB, the mechanism of action of PEO-PPO block copolymers is unknown, and there has been minimal exploration of polymers beyond P188. In this study, a human-based in vitro BBB model under oxidative stress was used to investigate polymer-BBB interactions since oxidative stress is closely linked with BBB dysfunction in many neurological injuries and disorders. PEO-PPO block copolymers of varied numbers of chemically distinct blocks, PEO block length, and functionality of the end group of the PPO block were assessed for their efficacy in improving key physiological readouts associated with BBB dysfunction. While treatment with P188 did not mitigate damage in the in vitro BBB model, treatment with three diblock copolymers improved barrier integrity under oxidative stress to a similar extent. Of the considered variations in the block copolymer design, the reduction in the number of chemically distinct blocks had the strongest influence on therapeutic function. The demonstrated efficacy of three alternative PEO-PPO diblock copolymers in this work reveals the potential of these polymers as a class of therapeutics that directly treat the damaged BBB, expanding the options for treatment of neurological injuries and disorders.
Collapse
Affiliation(s)
- Pranati P Mondkar
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Hannah S Seo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Timothy P Lodge
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
3
|
Mutore KT, Koduri R, Alatrash N, Nomellini V. THE USE OF POLOXAMER 188 IN BURN INJURY TREATMENT: A SYSTEMATIC LITERATURE REVIEW. Shock 2024; 62:461-469. [PMID: 39178216 DOI: 10.1097/shk.0000000000002439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
ABSTRACT Although there have been numerous advancements in burn wound management, burn injuries are still a major cause of morbidity and mortality in the United States, and novel therapeutics are still needed to improve outcomes. Poloxamer 188 (P188) is a synthetic copolymer with Food and Drug Administration (FDA) approval that has many biological applications. This study aimed to review the literature on P188 in burn injuries and its effects based on burn mechanisms. We employed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines to complete this systematic literature review. We searched the databases of Google Scholar, PubMed, and SCOPUS using the keywords burn, p188, poloxamer 188, and pluronic F68 in combination. Two reviewers independently screened the articles for inclusion. Articles that were not in English, were book chapters or conference proceedings, or did not evaluate P188 in the setting of burn injuries were excluded. We included a total of 33 full-text articles with both in vivo and in vitro preclinical studies. P188 was found to be beneficial in animal and cell studies evaluating electrical and thermal burn injuries. P188 was also found to be useful in burn wound management. Although its utility may be limited in radiation injuries, P188 may be helpful in delaying the initial damage caused by radiation burns. P188 therefore has the potential to be used as a therapy in both burn wound management and in the treatment of systemic injuries sustained through burns. Future studies should aim to assess the efficacy of P188 in clinical models of burn injury.
Collapse
Affiliation(s)
- Kevin T Mutore
- Division of Burn, Trauma, Acute, and Critical Care Surgery, Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | | | | | | |
Collapse
|
4
|
Wu R, Koduri R, Cho M, Alatrash N, Nomellini V. Effects of poloxamer 188 on traumatic brain injury. Brain Behav Immun Health 2024; 38:100762. [PMID: 38590762 PMCID: PMC11000117 DOI: 10.1016/j.bbih.2024.100762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/10/2024] Open
Abstract
Traumatic Brain Injury (TBI) is a major cause of severe disability and death, resulting in significant health care and economic burden. Poloxamer 188, a synthetic tri-block copolymer approved by the FDA, has been studied for its potential effects on traumatic brain injury (TBI). The neuroprotective abilities of P188 have attracted significant attention. This systematic review aims to compile evidence of P188's effect on the treatment of TBI. A comprehensive literature search was conducted using PubMed, SCOPUS, and Google Scholar databases, which yielded 20 articles that satisfied the inclusion criteria. These articles have shown direct protective effects of P188 on brain tissue following TBI, including restitution of the increase cell membrane permeability, attenuation of neuronal necrosis and apoptosis, improvement of mitochondrial viability, reduction in axonal disruption, and restoration of the blood brain barrier. In animals, P188 has been shown to improve sensorimotor functions, as well as spatial learning and memory.
Collapse
Affiliation(s)
- Renqing Wu
- Division of Burn, Trauma, Acute, and Critical Care Surgery, Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Roopa Koduri
- Division of Burn, Trauma, Acute, and Critical Care Surgery, Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Cho
- Department of Bioengineering, UT Arlington, Arlington, TX, USA
| | - Nagham Alatrash
- Division of Burn, Trauma, Acute, and Critical Care Surgery, Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Vanessa Nomellini
- Division of Burn, Trauma, Acute, and Critical Care Surgery, Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Chen G, Douglas HF, Li Z, Cleveland WJ, Balzer C, Yannopolous D, Chen IYL, Obal D, Riess ML. Cardioprotection by Poloxamer 188 is Mediated through Increased Endothelial Nitric Oxide Production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.18.593838. [PMID: 38826479 PMCID: PMC11142105 DOI: 10.1101/2024.05.18.593838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Ischemia/reperfusion (I/R) injury significantly contributes to the morbidity and mortality associated with cardiac events. Poloxamer 188 (P188), a nonionic triblock copolymer, has been proposed to mitigate I/R injury by stabilizing cell membranes. However, the underlying mechanisms remain incompletely understood, particularly concerning endothelial cell function and nitric oxide (NO) production. We employed human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) and endothelial cells (ECs) to elucidate the effects of P188 on cellular survival, function, and NO secretion under simulated I/R conditions. iPSC-CMs contractility and iPSC-ECs' NO production were assessed following exposure to P188. Further, an isolated heart model using Brown Norway rats subjected to I/R injury was utilized to evaluate the ex-vivo cardioprotective effects of P188, examining cardiac function and NO production, with and without the administration of a NO inhibitor. In iPSC-derived models, P188 significantly preserved CM contractile function and enhanced cell viability after hypoxia/reoxygenation. Remarkably, P188 treatment led to a pronounced increase in NO secretion in iPSC-ECs, a novel finding demonstrating endothelial protective effects beyond membrane stabilization. In the rat isolated heart model, administration of P188 during reperfusion notably improved cardiac function and reduced I/R injury markers. This cardioprotective effect was abrogated by NO inhibition, underscoring the pivotal role of NO. Additionally, a dose-dependent increase in NO production was observed in non-ischemic rat hearts treated with P188, further establishing the critical function of NO in P188 induced cardioprotection. In conclusion, our comprehensive study unveils a novel role of NO in mediating the protective effects of P188 against I/R injury. This mechanism is evident in both cellular models and intact rat hearts, highlighting the potential of P188 as a therapeutic agent against I/R injury. Our findings pave the way for further investigation into P188's therapeutic mechanisms and its potential application in clinical settings to mitigate I/R-related cardiac dysfunction.
Collapse
|
6
|
Suarez D, Kjar A, Scott B, Hillam K, Vargis E, Nielson C, Sommer E, Zhang E, Holley A, Traxler A, Hughes M, Wang Y, Firpo MA, Britt D, Park AH. Can Ganciclovir and Quercetin-P188 Ameliorate Cytomegalovirus Induced Hearing Loss? Laryngoscope 2024; 134:1457-1463. [PMID: 37589298 DOI: 10.1002/lary.30975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/12/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVE Determine whether combination therapy with ganciclovir (GCV) and a Quercetin-P188 solution improves hearing outcomes in a murine cytomegalovirus (CMV) model. METHODS BALB/c mice were infected with murine CMV on postnatal day 3 (p3). Quercetin was solubilized in saline using P188 (QP188). Treatment groups received either GCV, QP188, GCV and QP188, or P188 delivery vehicle BID at 12-hour intervals via intraperitoneal injection. All treatment groups were treated for 14 days starting at p3. Uninfected controls were treated with the combined regimen, saline or P188 delivery vehicle. Auditory thresholds were assessed using distortion product otoacoustic emission (DPOAE) and auditory brainstem response (ABR) testing at 4, 6, and 8 weeks of age. Temporal bones from separate CMV-infected groups were harvested at p10, and viral load was determined by quantitative polymerase chain reaction. RESULTS CMV-infected mice receiving combination therapy GCV+QP188 demonstrated statistically significant lower ABR (p < 0.001) and DPOAE thresholds (p < 0.001) compared with mice treated with GCV monotherapy, QP188 monotherapy, and P188 delivery vehicle at 4, 6, and 8 weeks of age. GCV+QP188 combination therapy, GCV monotherapy, and QP188 monotherapy resulted in a nonsignificant reduction in mean viral titers compared to P188 monotherapy (p = 0.08). CONCLUSION Combining GCV with the excipients quercetin and P188 effectively ameliorated CMV-induced sensorineural hearing loss in a murine model. This result may be partially explained by a reduction in viral titers in mouse temporal bones that correlate with in vitro studies demonstrating additive antiviral effect in cell culture. LEVEL OF EVIDENCE NA Laryngoscope, 134:1457-1463, 2024.
Collapse
Affiliation(s)
- Daniel Suarez
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Andrew Kjar
- Department of Biological Engineering, Utah State University, Logan, Utah, U.S.A
| | - Boston Scott
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Katrina Hillam
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Elizabeth Vargis
- Department of Biological Engineering, Utah State University, Logan, Utah, U.S.A
| | - Christopher Nielson
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Elizabeth Sommer
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Emily Zhang
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Anna Holley
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Abigail Traxler
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Maura Hughes
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Yong Wang
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Matthew A Firpo
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - David Britt
- Department of Biological Engineering, Utah State University, Logan, Utah, U.S.A
| | - Albert H Park
- Division of Otolaryngology - Head and Neck Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, Utah, U.S.A
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| |
Collapse
|
7
|
Alfieri L, Montana A, Frisoni P, D'Errico S, Neri M. Application of Aquaporins as Markers in Forensic Pathology: A Systematic Review of the Literature. Int J Mol Sci 2024; 25:2664. [PMID: 38473914 DOI: 10.3390/ijms25052664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The study of aquaporins (AQPs) in various forensic fields has offered a promising horizon in response to the need to have reliable elements for the identification of the manner of death and for the individuation of forensic markers for the timing of lesions and vitality of injury. In the literature, various tissues have been studied; the most investigated are the lungs, brain, kidneys, skin, and blood vessels. A systematic literature review on PubMed following PRISMA 2020 guidelines enabled the identification of 96 articles. In all, 34 of these were enrolled to identify Aquaporin-like (AQP-like) forensic markers. The analysis of the literature demonstrated that the most significant markers among the AQPs are as follows: for the brain, AQP4, which is very important in brain trauma and hypoxic damage; AQP3 in the skin lesions caused by various mechanisms; and AQP5 in the diagnosis of drowning. Other applications are in organ damage due to drug abuse and thrombus dating. The focus of this review is to collect all the data present in the literature about the forensic application of AQPs as forensic markers in the most important fields of application. In the current use, the individuation, validation, and application of markers in forensic investigation are very useful in real forensic applications in cases evaluated in court.
Collapse
Affiliation(s)
- Letizia Alfieri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Angelo Montana
- Department of Biomedical Sciences and Public Health, University Politecnica delle Marche, 60126 Ancona, Italy
| | - Paolo Frisoni
- Unit of Legal Medicine, AUSL Romagna, G.B. Morgagni-L. Pierantoni Hospital, 47100 Forlì, Italy
| | - Stefano D'Errico
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Margherita Neri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
8
|
Chen L, Xiong Y, Chopp M, Pang H, Emanuele M, Zhang ZG, Mahmood A, Zhang Y. Vepoloxamer improves functional recovery in rat after traumatic brain injury: A dose-response and therapeutic window study. Neurochem Int 2024; 173:105659. [PMID: 38142856 PMCID: PMC10872547 DOI: 10.1016/j.neuint.2023.105659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. There are no effective therapies available for TBI patients. Vepoloxamer is an amphiphilic polyethylene-polypropylene-polyethylene tri-block copolymer that seals membranes and restores plasma membrane integrity in damaged cells. We previously demonstrated that treatment of TBI rats with Vepoloxamer improves functional recovery. However, additional studies are needed to potentially translate Vepoloxamer treatment from preclinical studies into clinical applications. We thus conducted a study to investigate dose-response and therapeutic window of Vepoloxamer on functional recovery of adult rats after TBI. To identify the most effective dose of Vepoloxamer, male Wistar adult rats with controlled cortical impact (CCI) injury were randomly treated with 0 (vehicle), 100, 300, or 600 mg/kg of Vepoloxamer, administered intravenously (IV) at 2 h after TBI. We then performed a therapeutic window study in which the rats were treated IV with the most effective single dose of Vepoloxamer at different time points of 2 h, 4 h, 1 day, or 3 days after TBI. A battery of cognitive and neurological tests was performed. Animals were killed 35 days after TBI for histopathological analysis. Dose-response experiments showed that Vepoloxamer at all three tested doses (100, 300, 600 mg/kg) administered 2 h post injury significantly improved cognitive functional recovery, whereas Vepoloxamer at doses of 300 and 600 mg/kg, but not the 100 mg/kg dose, significantly reduced lesion volume compared to saline treatment. However, Vepoloxamer at 300 mg/kg showed significantly improved neurological and cognitive outcomes than treatment with a dose of 600 mg/kg. In addition, our data demonstrated that the dose of 300 mg/kg of Vepoloxamer administered at 2 h, 4 h, 1 day, or 3 days post injury significantly improved neurological function compared with vehicle, whereas Vepoloxamer administered at 2 h or 4 h post injury significantly improved cognitive function compared with the 1-day and 3-day treatments, with the most robust effect administered at 2 h post injury. The present study demonstrated that Vepoloxamer improves functional recovery in a dose-and time-dependent manner, with therapeutic efficacy compared with vehicle evident even when the treatment is initiated 3 days post TBI in the rat.
Collapse
Affiliation(s)
- Liang Chen
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| | - Haiyan Pang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA
| | | | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Yanlu Zhang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, 48202, USA.
| |
Collapse
|
9
|
Fang Z, Cao P, Pan N, Lu H. Pluronic P85 decreases the delivery of phenytoin to the brain in drug-resistant rats with P-glycoprotein overexpressed chronic mesial temporal lobe epilepsy. IBRO Neurosci Rep 2023; 15:100-106. [PMID: 37485299 PMCID: PMC10362368 DOI: 10.1016/j.ibneur.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
P-glycoprotein (Pgp) overexpressed in blood brain barrier (BBB) is hypothesized to lower brain drug concentrations and thus inhibit anticonvulsant effects in drug-resistant epilepsy. Pluronic P85 (P85) was proved to enhance the delivery of drugs into the brain by inhibition of Pgp. To determine whether the surfactant P85 [versus Pgp inhibitor tariquidar (TQD)] enhance phenytoin (PHT) into the brain in drug-resistant rats with chronic mesial temporal lobe epilepsy (MTLE) induced by lithium-pilocarpine, in brain of which Pgp were overexpressed, then direct verification of PHT transport via measurement of PHT concentration in brain using microdialysis. The drug-resistant model rats were randomly divided into three groups, which were treated with PHT, 1%P85 + PHT, or PHT+TQD, respectively. 1%P85 + PHT treatment displayed a lower ratio of the area under the curve (AUC) of the PHT concentration in the brain/plasma even than that of the PHT treatment in model rats (p < 0.05), while PHT+TQD showed the highest ratio of the AUC of all treatments. However, the ratio of the PHT concentration in the liver/plasma was similar in three model groups (p > 0.05). For the ratio of the kidney/plasma, PHT+TQD treatment model group had the highest ratio of the other treatments in model rats. Thus, P85 oppositely decreased PHT concentration in brain in drug-resistant model rats with Pgp overexpressed MTLE while TQD could increase PHT distribution in brain.
Collapse
Affiliation(s)
- Ziyan Fang
- The Affiliated Brain Hospital of Guangzhou Medical University, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
| | - Penghui Cao
- The Affiliated Brain Hospital of Guangzhou Medical University, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
| | - Nannan Pan
- The Affiliated Brain Hospital of Guangzhou Medical University, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
| | - Haoyang Lu
- The Affiliated Brain Hospital of Guangzhou Medical University, 36th Mingxin Road, Guangzhou, Guangdong 510370, PR China
| |
Collapse
|
10
|
Zha D, Wang S, Monaghan-Nichols P, Qian Y, Sampath V, Fu M. Mechanisms of Endothelial Cell Membrane Repair: Progress and Perspectives. Cells 2023; 12:2648. [PMID: 37998383 PMCID: PMC10670313 DOI: 10.3390/cells12222648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Endothelial cells are the crucial inner lining of blood vessels, which are pivotal in vascular homeostasis and integrity. However, these cells are perpetually subjected to a myriad of mechanical, chemical, and biological stresses that can compromise their plasma membranes. A sophisticated repair system involving key molecules, such as calcium, annexins, dysferlin, and MG53, is essential for maintaining endothelial viability. These components orchestrate complex mechanisms, including exocytosis and endocytosis, to repair membrane disruptions. Dysfunctions in this repair machinery, often exacerbated by aging, are linked to endothelial cell death, subsequently contributing to the onset of atherosclerosis and the progression of cardiovascular diseases (CVD) and stroke, major causes of mortality in the United States. Thus, identifying the core machinery for endothelial cell membrane repair is critically important for understanding the pathogenesis of CVD and stroke and developing novel therapeutic strategies for combating CVD and stroke. This review summarizes the recent advances in understanding the mechanisms of endothelial cell membrane repair. The future directions of this research area are also highlighted.
Collapse
Affiliation(s)
- Duoduo Zha
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Shizhen Wang
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA;
| | - Paula Monaghan-Nichols
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Rd, Honggu District, Nanchang 330031, China;
| | - Venkatesh Sampath
- Department of Pediatric, Children’s Mercy Hospital, Children’s Mercy Research Institute, Kansas City, MO 64108, USA;
| | - Mingui Fu
- Department of Biomedical Science, School of Medicine, University of Missouri Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; (D.Z.); (P.M.-N.)
| |
Collapse
|
11
|
Crabtree A, Boehnke N, Bates F, Hackel B. Consequences of poly(ethylene oxide) and poloxamer P188 on transcription in healthy and stressed myoblasts. Proc Natl Acad Sci U S A 2023; 120:e2219885120. [PMID: 37094151 PMCID: PMC10161009 DOI: 10.1073/pnas.2219885120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/26/2023] [Indexed: 04/26/2023] Open
Abstract
Poly(ethylene oxide) (PEO) and poloxamers, a class of poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) (PEO-PPO-PEO) triblock copolymers, have many personal and medical care applications, including the stabilization of stressed cellular membranes. Despite the widespread use, the cellular transcriptional response to these molecules is relatively unknown. C2C12 myoblasts, a model muscle cell, were subjected to short-term Poloxamer 188 (P188) and PEO181 (8,000 g/mol) treatment in culture. RNA was extracted and sequenced to quantify transcriptomic impact. The addition of moderate concentrations (14 µM) of either polymer to unstressed cells caused substantial differential gene expression, including at least twofold modulation of 357 and 588 genes, respectively. In addition, evaluation of the transcriptome response to osmotic stress without polymer treatment revealed dramatic change in RNA expression. Interestingly, the addition of polymer to stressed cells-at concentrations that provide physiological protection-did not yield a significant difference in expression of any gene relative to stress alone. Genome-scale expression analysis was corroborated by single-gene quantitative real-time PCR. Changes in protein expression were measured via western blot, which revealed partial alignment with the RNA results. Collectively, the significant changes to expression of multiple genes and resultant protein translation demonstrates an unexpectedly broad biochemical response to these polymers in healthy myoblasts in vitro. Meanwhile, the lack of substantial transcriptional response to polymer treatment in stressed cells highlights the physical nature of that protective mechanism.
Collapse
Affiliation(s)
- Adelyn A. Crabtree
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
12
|
Yu E, Zhang E, Lv X, Yan L, Lin Z, Siaw-Debrah F, Zhang Y, Yang S, Ruan L, Zhuge Q, Ni H. LDC7559 Exerts Neuroprotective Effects by Inhibiting GSDMD-dependent Pyroptosis of Microglia in Mice with Traumatic Brain Injury. J Neurotrauma 2022; 40:742-757. [PMID: 35920115 DOI: 10.1089/neu.2021.0318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis is considered one of a critical factor in the recovery of neurological function following traumatic brain injury. Brain injury activates a molecular signaling cascade associated with pyroptosis and inflammation, including NLRP3, inflammatory cytokines, caspase-1, gasdermin D (GSDMD), and other pyroptosis-related proteins. In this study, we explored the neuroprotective effects of LDC7559, a GSDMD inhibitor. Briefly, LDC7559, siRNA-GSDMD (si-GSDMD), or equal solvent was administrated to mice with a lipopolysaccharide + nigericin (LPS + Nig) model in vitro or with controlled cortical impact brain injury. The findings revealed that inflammation and pyroptosis levels were decreased by LDC7559 or si-GSDMD treatment both in vitro and in vivo. Immunofluorescence staining, brain water content, hematoxylin and eosin staining, and behavioral investigations suggested that LDC7559 or si-GSDMD inhibited microglial proliferation, ameliorated cerebral edema, reduced brain tissue loss, and promoted brain function recovery. Taken together, LDC7559 may inhibit pyroptosis and reduce inflammation by inhibiting GSDMD, thereby promoting the recovery of neurological function.
Collapse
Affiliation(s)
- Enxing Yu
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,Ningbo City First Hospital, Department of Plastic and Reconstructive Surgery, Ningbo, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery,, Wenzhou, Zhejiang, China;
| | - Erjia Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China;
| | - Xinhuang Lv
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China;
| | - Lin Yan
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Zhongxiao Lin
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Felix Siaw-Debrah
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,Korle Bu Teaching Hospital, Department of Neurosurgery, Korlebu teaching hospital, Accra, Greater Accra, Ghana;
| | - Ying Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Su Yang
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Linhui Ruan
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Qichuan Zhuge
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| | - Haoqi Ni
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Department of Neurosurgery, Wenzhou, Zhejiang, China;
| |
Collapse
|
13
|
Kjar A, Wadsworth I, Vargis E, Britt DW. Poloxamer 188 - quercetin formulations amplify in vitro ganciclovir antiviral activity against cytomegalovirus. Antiviral Res 2022; 204:105362. [PMID: 35709898 DOI: 10.1016/j.antiviral.2022.105362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022]
Abstract
Treatment of human cytomegalovirus (CMV) infection requires long-term administration of nucleoside analog antivirals such as ganciclovir (GCV), a therapy frequently limited by GCV-induced toxicity. Here, combining GCV treatment with two bioactive excipients, poloxamer 188 and quercetin, was investigated in vitro to reduce GCV dosage. Quercetin is a natural flavonoid exhibiting antiviral activity against CMV by a mechanism distinct from GCV, but is poorly soluble, limiting its use as a therapeutic. To overcome this challenge, quercetin was co-formulated with poloxamer 188 (P188, Pluronic ® F68). Quercetin-P188 (QP188) formulations yielded only modest CMV viral inhibition, with a selectivity index of 11.4, contrasted with a GCV selectivity index of 95. More significantly, when coadministered with GCV, QP188 exhibited an additive or synergistic interaction in subtherapeutic ranges of GCV. Fluorescence microscopy revealed QP188 accumulation in fibroblast mitochondria, suggesting that the excipient may modulate mitochondrial processes relevant to CMV infection. GCV antiviral therapy augmented with poloxamer-solubilized quercetin may be a viable approach to maintain CMV inhibition while lowering GCV doses, translating to reduced associated toxicity.
Collapse
Affiliation(s)
- Andrew Kjar
- Biological Engineering Department, Utah State University, Logan Utah, 84325, USA
| | - Ian Wadsworth
- Biological Engineering Department, Utah State University, Logan Utah, 84325, USA
| | - Elizabeth Vargis
- Biological Engineering Department, Utah State University, Logan Utah, 84325, USA.
| | - David W Britt
- Biological Engineering Department, Utah State University, Logan Utah, 84325, USA.
| |
Collapse
|
14
|
Cui N, Dai CY, Mao X, Lv X, Gu Y, Lee ES, Jiang HB, Sun Y. Poloxamer-Based Scaffolds for Tissue Engineering Applications: A Review. Gels 2022; 8:360. [PMID: 35735704 PMCID: PMC9222596 DOI: 10.3390/gels8060360] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/28/2022] [Accepted: 06/01/2022] [Indexed: 12/28/2022] Open
Abstract
Poloxamer is a triblock copolymer with amphiphilicity and reversible thermal responsiveness and has wide application prospects in biomedical applications owing to its multifunctional properties. Poloxamer hydrogels play a crucial role in the field of tissue engineering and have been regarded as injectable scaffolds for loading cells or growth factors (GFs) in the last few years. Hydrogel micelles can maintain the integrity and stability of cells and GFs and form an appropriate vascular network at the application site, thus creating an appropriate microenvironment for cell growth, nerve growth, or bone integration. The injectability and low toxicity of poloxamer hydrogels make them a noninvasive method. In addition, they can also be good candidates for bio-inks, the raw material for three-dimensional (3D) printing. However, the potential of poloxamer hydrogels has not been fully explored owing to the complex biological challenges. In this review, the latest progress and cutting-edge research of poloxamer-based scaffolds in different fields of application such as the bone, vascular, cartilage, skin, nervous system, and organs in tissue engineering and 3D printing are reviewed, and the important roles of poloxamers in tissue engineering scaffolds are discussed in depth.
Collapse
Affiliation(s)
- Naiyu Cui
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Chun-Yu Dai
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Xuran Mao
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Xun Lv
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Yue Gu
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Eui-Seok Lee
- Department of Oral and Maxillofacial Surgery, Graduate School of Clinical Dentistry, Korea University, Seoul 08308, Korea
| | - Heng-Bo Jiang
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| | - Yunhan Sun
- The CONVERSATIONALIST Club, School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an 271016, China; (N.C.); (C.-Y.D.); (X.M.); (X.L.); (Y.G.)
| |
Collapse
|
15
|
Di Meco A, Kemal S, Popovic J, Chandra S, Sadleir KR, Vassar R. Poloxamer-188 Exacerbates Brain Amyloidosis, Presynaptic Dystrophies, and Pathogenic Microglial Activation in 5XFAD Mice. Curr Alzheimer Res 2022; 19:317-329. [DOI: 10.2174/1567205019666220509143823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer’s disease (AD) is initiated by aberrant accumulation of amyloid beta (Aβ) protein in the brain parenchyma. The microenvironment surrounding amyloid plaques is characterized by the swelling of presynaptic terminals (dystrophic neurites) associated with lysosomal dysfunction, microtubule disruption and impaired axonal transport. Aβ-induced plasma membrane damage and calcium influx could be potential mechanisms underlying dystrophic neurite formation.
Objective:
We tested whether promoting membrane integrity by brain administration of a safe FDA approved surfactant molecule poloxamer-188 (P188) could attenuate AD pathology in vivo.
Methods:
Three-month-old 5XFAD male mice were administered several concentrations of P188 in the brain for 42 days with mini-osmotic pumps. After 42 days, mice were euthanized and assessed for amyloid pathology, dystrophic neurites, pathogenic microglia activation, tau phosphorylation and lysosomal / vesicular trafficking markers in the brain.
Results:
P188 was lethal at the highest concentration of 10mM. Lower concentrations of P188 (1.2, 12 and 120μM) were well tolerated. P188 increased brain Aβ burden, potentially through activation of the γ-secretase pathway. Dystrophic neurite pathology was exacerbated in P188 treated mice as indicated by increased LAMP1 accumulation around Aβ deposits. Pathogenic microglial activation was increased by P188. Total tau levels were decreased by P188. Lysosomal enzyme cathepsin D and calcium-dependent vesicular trafficking regulator synaptotagmin-7 (SYT7) were dysregulated upon P188 administration.
Conclusion:
P188 brain delivery exacerbated amyloid pathology, dystrophic neurites and pathogenic microglial activation in 5XFAD mice. These effects correlated with lysosomal dysfunction and dysregulation of plasma membrane vesicular trafficking. P188 is not a promising therapeutic strategy against AD pathogenesis.
Collapse
Affiliation(s)
- Antonio Di Meco
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Shahrnaz Kemal
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Jelena Popovic
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Sidhanth Chandra
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | | | - Robert Vassar
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Mesulam Center for Cognitive Neurology and Alzheimer’s disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
16
|
Chen WN, Shaikh MF, Bhuvanendran S, Date A, Ansari MT, Radhakrishnan AK, Othman I. Poloxamer 188 (P188), A Potential Polymeric Protective Agent for Central Nervous System Disorders: A Systematic Review. Curr Neuropharmacol 2022; 20:799-808. [PMID: 34077349 PMCID: PMC9878954 DOI: 10.2174/1570159x19666210528155801] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 11/22/2022] Open
Abstract
Poloxamer 188 (P188) is an FDA-approved biocompatible block copolymer composed of repeating units of Poly(Ethylene Oxide) (PEO) and poly(propylene oxide) (PPO). Due to its amphiphilic nature and high Hydrophile-Lipophile Balance (HLB) value of 29, P188 is used as a stabilizer/emulsifier in many cosmetics and pharmaceutical preparations. While the applications of P188 as an excipient are widely explored, the data on the pharmacological activity of P188 are scarce. Notably, the neuroprotective potential of P188 has gained a lot of interest. Therefore, this systematic review is aimed at summarizing evidence of neuroprotective potential of P188 in CNS disorders. The PRISMA model was used, and five databases (Google Scholar, Scopus, Wiley Online Library, ScienceDirect, and PubMed) were searched with relevant keywords. The search resulted in 11 articles, which met the inclusion criteria. These articles described the protective effects of P188 on traumatic brain injury or mechanical injury in cells, neurotoxicity, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), and ischemia/ reperfusion injury from stroke. All the articles were original research in experimental or pre-clinical stages using animal models or in vitro systems. The reported activities demonstrated the potential of P188 as a neuroprotective agent in improving CNS conditions such as neurodegeneration.
Collapse
Affiliation(s)
- Win Ning Chen
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia;,Address correspondence to this author at the Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia; Tel: +603 5514 4483; E-mail:
| | - Saatheeyavaane Bhuvanendran
- Brain Research Institute of Monash Sunway (BRIMS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Abhijit Date
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI96720, USA
| | - Mohammad Tahir Ansari
- School of Pharmacy, University of Nottingham Malaysia, Semenyih43500, Selangor, Malaysia
| | - Ammu Kutty Radhakrishnan
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| |
Collapse
|
17
|
Dadgostar E, Rahimi S, Nikmanzar S, Nazemi S, Naderi Taheri M, Alibolandi Z, Aschner M, Mirzaei H, Tamtaji OR. Aquaporin 4 in Traumatic Brain Injury: From Molecular Pathways to Therapeutic Target. Neurochem Res 2022; 47:860-871. [PMID: 35088218 DOI: 10.1007/s11064-021-03512-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) is known as an acute degenerative pathology of the central nervous system, and has been shown to increase brain aquaporin 4 (AQP4) expression. Various molecular mechanisms affect AQP4 expression, including neuronal high mobility group box 1, forkhead box O3a, vascular endothelial growth factor, hypoxia-inducible factor-1 α (HIF-1 α) sirtuin 2, NF-κB, Malat1, nerve growth factor and Angiotensin II receptor type 1. In addition, inhibition of AQP4 with FK-506, MK-801 (indirectly by targeting N-methyl-D-aspartate receptor), inactivation of adenosine A2A receptor, levetiracetam, adjudin, progesterone, estrogen, V1aR inhibitor, hypertonic saline, erythropoietin, poloxamer 188, brilliant blue G, HIF-1alpha inhibitor, normobaric oxygen therapy, astaxanthin, epigallocatechin-3-gallate, sesamin, thaliporphine, magnesium, prebiotic fiber, resveratrol and omega-3, as well as AQP4 gene silencing lead to reduced edema upon TBI. This review summarizes current knowledge and evidence on the relationship between AQP4 and TBI, and the potential mechanisms involved.
Collapse
Affiliation(s)
- Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shiva Rahimi
- School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shahin Nikmanzar
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - Sina Nazemi
- Tracheal Disease Research Center (TDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Naderi Taheri
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Alibolandi
- Anatomical Science Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Omid Reza Tamtaji
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Nugraha DH, Anggadiredja K, Rachmawati H. Mini-Review of Poloxamer as a Biocompatible Polymer for Advanced Drug Delivery. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e21125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
19
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
1,2-Dichloroethane induces apoptosis in the cerebral cortexes of NIH Swiss mice through microRNA-182-5p targeting phospholipase D1 via a mitochondria-dependent pathway. Toxicol Appl Pharmacol 2021; 430:115728. [PMID: 34560092 DOI: 10.1016/j.taap.2021.115728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/25/2021] [Accepted: 09/17/2021] [Indexed: 01/23/2023]
Abstract
1,2-Dichloroethane (1,2-DCE) is a pervasive environmental pollutant found in ambient and residential air, as well as ground and drinking water. Overexposure to it results in cortex edema, in both animals and humans. 1,2-DCE induces apoptosis in the cerebellum, liver and testes. This promotes the hypothesis that 1,2-DCE may induce apoptosis in the cortex as brain edema progresses. To validate our hypothesis, 40 NIH male mice were exposed to 0, 100, 350, 700 mg/m3 1,2-DCE by whole-body dynamic inhalation for 28 consecutive days. MicroRNA (miRNA) and mRNA microarray combined with TdT-mediated dUTP nick-end labeling, flow cytometry, and mitochondrial membrane potential (mtΔΨ) measurement were applied to identify the cortex apoptosis pathways' specific responses to 1,2-DCE, in vitro and in vivo. The results showed that 1,2-DCE caused brain edema and increased apoptosis in the mouse cortexes. We confirmed that 1,2-DCE induced increased apoptosis via mitochondrial pathway, both in vitro and in vivo, as evidenced by increased Caspase-3, cleaved Caspase-3, Cytochrome c and Bax expression, and decreased Bcl-2 expression. Additionally, mtΔΨ decreased after 1,2-DCE treatment in vitro. 1,2-DCE exposure increased miR-182-5p and decreased phospholipase D1 (PLD1) in the cerebral cortex of mice. MiR-182-5p overexpression and PLD1 inhibition reduced mtΔΨ and increased astrocyte apoptosis, yet miR-182-5p inhibition alleviated the 1,2-DCE-induced PLD1 down-regulation and the increased apoptosis. Finally, PLD1 was confirmed to be a target of miR-182-5p by luciferase assay. Taken together, our findings indicate that 1,2-DCE exposure induces apoptosis in the cortex via a mitochondria-dependent pathway. This pathway is regulated by a miR-182-5p⊣PLD1 axie.
Collapse
|
21
|
Lotze FP, Riess ML. Poloxamer 188 Exerts Direct Protective Effects on Mouse Brain Microvascular Endothelial Cells in an In Vitro Traumatic Brain Injury Model. Biomedicines 2021; 9:1043. [PMID: 34440247 PMCID: PMC8393826 DOI: 10.3390/biomedicines9081043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/04/2022] Open
Abstract
Traumatic Brain Injury (TBI), the main contributor to morbidity and mortality worldwide, can disrupt the cell membrane integrity of the vascular endothelial system, endangering blood-brain barrier function and threatening cellular subsistence. Protection of the vascular endothelial system might enhance clinical outcomes after TBI. Poloxamer 188 (P188) has been shown to improve neuronal function after ischemia/reperfusion (I/R) injury as well as after TBI. We aimed to establish an in vitro compression-type TBI model, comparing mild-to-moderate and severe injury, to observe the direct effects of P188 on Mouse Brain Microvascular Endothelial Cells (MBEC). Confluent MBEC were exposed to normoxic or hypoxic conditions for either 5 or 15 h (hours). 1 h compression was added, and P188 was administered during 2 h reoxygenation. A direct effect of P188 on MBEC was tested by assessing cell number/viability, cytotoxicity/membrane damage, metabolic activity, and total nitric oxide production (tNOp). While P188 enhanced cell number/viability, metabolic activity, and tNOp, an increase in cytotoxicity/membrane damage after mild-to-moderate injury was prevented. In severely injured MBEC, P188 improved metabolic activity only. P188, present during reoxygenation, influenced MBEC function directly in simulated I/R and compression-type TBI.
Collapse
Affiliation(s)
- Felicia P. Lotze
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Anesthesiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Matthias L. Riess
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
22
|
Tang SE, Liao WI, Pao HP, Hsu CW, Wu SY, Huang KL, Chu SJ. Poloxamer 188 Attenuates Ischemia-Reperfusion-Induced Lung Injury by Maintaining Cell Membrane Integrity and Inhibiting Multiple Signaling Pathways. Front Pharmacol 2021; 12:650573. [PMID: 34335242 PMCID: PMC8319770 DOI: 10.3389/fphar.2021.650573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/05/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Poloxamer 188 (P188) possesses anti-inflammatory properties and can help to maintain plasma membrane function. P188 has been reported to exert beneficial effects in the treatment of various disorders. However, the effects of P188 in ischemia/reperfusion (IR)-induced acute lung injury have not been examined. Methods: We investigated the ability of P188 to attenuate IR-induced acute lung injury in rats and hypoxia/reoxygenation (HR) injury in murine epithelial cells. Isolated perfused rat lungs were exposed to 40 min ischemia followed by 60 min reperfusion to induce IR injury. Results: IR led to lung edema, increased pulmonary arterial pressure, promoted lung tissue inflammation and oxidative stress, and upregulated the levels of TNF-α, IL-6 and CINC-1, and increased Lactic dehydrogenase (LDH) activity in bronchoalveolar lavage fluid. IR also downregulated the levels of inhibitor of κB (IκB-α), upregulated nuclear factor (NF)-κB (NF-κB), and promoted apoptosis in lung tissues. P188 significantly suppressed all these effects. In vitro, P188 also exerted a similar effect in murine lung epithelial cells exposed to HR. Furthermore, P188 reduced the number of propidium iodide-positive cells, maintained cell membrane integrity, and enhanced cell membrane repair following HR. Conclusion: We conclude that P188 protects against lung IR injury by suppressing multiple signaling pathways and maintaining cell membrane integrity.
Collapse
Affiliation(s)
- Shih-En Tang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Wang Hsu
- Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
23
|
Choudhary RC, Shoaib M, Sohnen S, Rolston DM, Jafari D, Miyara SJ, Hayashida K, Molmenti EP, Kim J, Becker LB. Pharmacological Approach for Neuroprotection After Cardiac Arrest-A Narrative Review of Current Therapies and Future Neuroprotective Cocktail. Front Med (Lausanne) 2021; 8:636651. [PMID: 34084772 PMCID: PMC8167895 DOI: 10.3389/fmed.2021.636651] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiac arrest (CA) results in global ischemia-reperfusion injury damaging tissues in the whole body. The landscape of therapeutic interventions in resuscitation medicine has evolved from focusing solely on achieving return of circulation to now exploring options to mitigate brain injury and preserve brain function after CA. CA pathology includes mitochondrial damage and endoplasmic reticulum stress response, increased generation of reactive oxygen species, neuroinflammation, and neuronal excitotoxic death. Current non-pharmacologic therapies, such as therapeutic hypothermia and extracorporeal cardiopulmonary resuscitation, have shown benefits in protecting against ischemic brain injury and improving neurological outcomes post-CA, yet their application is difficult to institute ubiquitously. The current preclinical pharmacopeia to address CA and the resulting brain injury utilizes drugs that often target singular pathways and have been difficult to translate from the bench to the clinic. Furthermore, the limited combination therapies that have been attempted have shown mixed effects in conferring neuroprotection and improving survival post-CA. The global scale of CA damage and its resultant brain injury necessitates the future of CA interventions to simultaneously target multiple pathways and alleviate the hemodynamic, mitochondrial, metabolic, oxidative, and inflammatory processes in the brain. This narrative review seeks to highlight the current field of post-CA neuroprotective pharmaceutical therapies, both singular and combination, and discuss the use of an extensive multi-drug cocktail therapy as a novel approach to treat CA-mediated dysregulation of multiple pathways, enhancing survival, and neuroprotection.
Collapse
Affiliation(s)
- Rishabh C Choudhary
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Muhammad Shoaib
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Samantha Sohnen
- Department of Anesthesiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Daniel M Rolston
- Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Daniel Jafari
- Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY, United States
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States
| | | | - Junhwan Kim
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Lance B Becker
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Emergency Medicine, Northshore University Hospital, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
24
|
Silencing long non-coding RNA zinc finger antisense 1 restricts secondary cerebral edema and neuron injuries after traumatic brain injury. Neurosci Lett 2021; 756:135958. [PMID: 34000346 DOI: 10.1016/j.neulet.2021.135958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To investigate the interaction of long non-coding RNA zinc finger antisense 1 (lncRNA ZFAS1) in secondary cerebral edema (CE) and neuron injuries after traumatic brain injury (TBI) in a mouse model. METHODS TBI mouse models was established by free-fall strike. Adeno-associated virus-short hairpin-ZFAS1 was administrated into mice via intracerebral injection to downregulate lncRNA ZFAS1. LncRNA ZFAS1 in mouse brain was examined. Neurological severity score (NSS), cerebral water content (CWC) and lesion volume were measured. The number of TUNEL-positive cells in brain tissue was accessed. Bax and cleaved caspase-3 in brain tissues were measured by western blot analysis, and pro-inflammatory factor levels were detected. RESULTS LncRNA ZFAS1 expression was upregulated in mouse brain tissues 3 days after TBI modelling. After the knockdown of lncRNA ZFAS1, NSS, CWC and lesion volume were decreased, apoptotic gene levels were decreased and pro-inflammatory cytokine levels were reduced, suggesting that lncRNA ZFAS1 knockdown could alleviate TBI-induced brain injuries in mice. CONCLUSION This study demonstrated that silencing lncRNA ZFAS1 inhibited TBI by quenching apoptosis, reducing inflammatory response and improving the recovery of neurological function in TBI mice. LncRNA ZFAS1 might function as a possible curative management in secondary CE and neuron injury in TBI mice.
Collapse
|
25
|
Meyer LJ, Riess ML. Evaluation of In Vitro Neuronal Protection by Postconditioning with Poloxamer 188 Following Simulated Traumatic Brain Injury. Life (Basel) 2021; 11:316. [PMID: 33917288 PMCID: PMC8067401 DOI: 10.3390/life11040316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) leads to morbidity and mortality worldwide. Reperfusion after ischemia adds detrimental injury to cells. Ischemia/reperfusion (I/R) injures cells in a variety of ways including cell membrane disruption. Hence, methods to improve endogenous membrane resealing capacity are crucial. Poloxamer (P) 188, an amphiphilic triblock copolymer, was found to be effective against I/R and mechanical injury in various experimental settings. The aim of this study was to establish an in vitro mouse neuronal TBI model and, further, to investigate if postconditioning with P188 directly interacts with neurons after compression and simulated I/R injury, when administered at the start of reoxygenation. Cellular function was assessed by cell number/viability, mitochondrial viability, membrane damage by lactated dehydrogenase (LDH) release and FM1-43 incorporation as well as apoptosis-activation by Caspase 3. Five hours hypoxia ± compression with 2 h reoxygenation proved to be a suitable model for TBI. Compared to normoxic cells not exposed to compression, cell number and mitochondrial viability decreased, whereas membrane injury by LDH release/FM1-43 dye incorporation and Caspase 3 activity increased in cells exposed to hypoxic conditions with compression followed by reoxygenation. P188 did not protect neurons from simulated I/R and/or compression injury. Future research is indicated.
Collapse
Affiliation(s)
- Luise J. Meyer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Anesthesiology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Matthias L. Riess
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA; Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
26
|
Lorente L, Martín MM, Pérez-Cejas A, González-Rivero AF, Ramos-Gómez L, Solé-Violán J, Cáceres JJ, Ferrer-Moure C, Jiménez A. Low blood caspase-8 levels in survivor patients of traumatic brain injury. Neurol Sci 2021; 42:5065-5070. [PMID: 33759054 DOI: 10.1007/s10072-021-05205-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/18/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE High concentrations of caspase-8 (main initiator caspase of the extrinsic pathway of apoptosis) have been found in brain tissue of patients with traumatic brain injury (TBI) and in the blood of patients with different diseases. However, blood caspase-8 concentrations in TBI patients have not been reported. Therefore, our aim was to analyze whether blood caspase-8 concentrations are associated with mortality in TBI patients. METHOD Patients with isolated and severe TBI were included. TBI was considered isolated if it showed an Injury Severity Score (ISS) <10 points on non-cranial aspects. TBI was considered severe if it showed a Glasgow Coma Scale (GCS) <9 points. This prospective observational study was conducted in 5 Intensive Care Units. Serum caspase-8 concentrations were measured on day 1 of TBI. RESULTS Surviving patients (n=59) had lower age (p=0.004), higher GCS (p=0.001), lower APACHE-II score (p<0.001), lower high-risk-of-death computed tomography (CT) findings (p=0.02), lower intracranial pressure (ICP) (p=0.01), and lower serum caspase-8 concentrations (p<0.001) than non-surviving patients (n=24). An association was found between serum caspase-8 levels and mortality after controlling for CT findings, GCS, and age (OR=1.037; 95% CI=1.013-1.062; p=0.002), and after controlling for CT findings, APACHE-II, and ICP (OR=1.042; 95% CI=1.013-1.071; p=0.004) in multiple logistic regression. CONCLUSIONS To our knowledge, this is the first series describing blood caspase-8 concentrations in patients with TBI. The association of high blood caspase-8 concentrations with mortality was the main new finding of the study. However, further investigations are needed to validate the preliminary results of our study.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, Ofra s/n La Laguna, 38320, Santa Cruz de Tenerife, Spain.
| | - María M Martín
- Intensive Care Unit, Hospital Universitario Nuestra Señora de Candelaria, Crta del Rosario s/n, 38010, Santa Cruz de Tenerife, Spain
| | - Antonia Pérez-Cejas
- Laboratory Department, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Agustín F González-Rivero
- Laboratory Department, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Luis Ramos-Gómez
- Intensive Care Unit, Hospital General de La Palma, Buenavista de Arriba s/n, 38713, Breña Alta, La Palma, Spain
| | - Jordi Solé-Violán
- Intensive Care Unit, Hospital Universitario Dr. Negrín, CIBERES Barranco de la Ballena s/n, 35010, Las Palmas de Gran Canaria, Spain
| | - Juan J Cáceres
- Intensive Care Unit, Hospital Insular, Plaza Dr. Pasteur s/n, 35016, Las Palmas de Gran Canaria, Spain
| | - Carmen Ferrer-Moure
- Laboratory Department, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Alejandro Jiménez
- Research Unit, Hospital Universitario de Canarias, Ofra s/n La Laguna, 38320, Santa Cruz de Tenerife, Spain
| |
Collapse
|
27
|
Salzman MM, Bartos JA, Yannopoulos D, Riess ML. Poloxamer 188 Protects Isolated Adult Mouse Cardiomyocytes from Reoxygenation Injury. Pharmacol Res Perspect 2020; 8:e00639. [PMID: 33073927 PMCID: PMC7570448 DOI: 10.1002/prp2.639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
Reperfusion injury is a complex pathological event involving processes that can lead to further disruption of the cell membrane and function following an ischemic event. Return of blood flow allows for the needed reperfusion; however, for a period of time before remaining viable cells stabilize, reperfusion results in additional cellular injury. In cardiomyocytes, loss of membrane integrity allows abnormal influx of extracellular calcium, leading to hyper-contracture and cell death. Methods to improve the membrane integrity of cardiomyocytes overwhelmed by pathological disruptions, such as reperfusion injury, are needed to prevent cell death, because of the myocardium's limited ability to regenerate. Research has shown administration of the copolymer P(oloxamer) 188 before ischemia/reperfusion can protect cardiomyocytes through membrane stabilization. This study sought to determine whether the administration of P188 at the beginning of the clinically more relevant time of reperfusion after ischemia will attenuate any additional damage to cardiomyocytes by stabilizing membrane integrity to allow the cells to maintain function. Using an in-vitro cardiomyocyte model subjected to hypoxia/reoxygenation to simulate ischemia/reperfusion injury, we show that reoxygenation significantly potentiates the injury caused by hypoxia itself. P188, with its unique combination of hydrophobic and hydrophilic chemical properties, and only delivered at the beginning of reoxygenation, dose-dependently protected cardiomyocytes from injury due to reoxygenation by repairing cell membranes, decreasing calcium influx, and maintaining cellular morphology. Our study also shows the hydrophobic portion of P188 is necessary for the stabilization of cell membrane integrity in providing protection to cardiomyocytes against reoxygenation injury.
Collapse
Affiliation(s)
- Michele M. Salzman
- Department of AnesthesiologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Present address:
Department of Pediatrics ‐ NeonatologyVanderbilt University Medical CenterNashvilleTNUnited States
| | - Jason A. Bartos
- Department of Medicine – Cardiovascular DivisionUniversity of MinnesotaMinneapolisMNUSA
| | - Demetris Yannopoulos
- Department of Medicine – Cardiovascular DivisionUniversity of MinnesotaMinneapolisMNUSA
| | - Matthias L. Riess
- Department of AnesthesiologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Department of AnesthesiologyTVHS VA Medical CenterNashvilleTNUSA
| |
Collapse
|
28
|
Naqvi S, Kumar P, Flora SJS. Comparative efficacy of Nano and Bulk Monoisoamyl DMSA against arsenic-induced neurotoxicity in rats. Biomed Pharmacother 2020; 132:110871. [PMID: 33069968 DOI: 10.1016/j.biopha.2020.110871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022] Open
Abstract
Chelation therapy is considered as a safe and effective strategy to combat metal poisoning. Arsenic is known to cause neurological dysfunctions such as impaired memory, encephalopathy, and peripheral neuropathy as it easily crosses the blood-brain barrier. Oxidative stress is one of the mechanisms suggested for arsenic-induced neurotoxicity. We prepared Solid Lipid nanoparticles loaded with Monoisoamyl 2, 3-dimercaptosuccinic acid (Nano-MiADMSA), and compared their efficacy with bulk MiADMSA for treating arsenic-induced neurological and other biochemical effects. Solid lipid nanoparticles entrapping MiADMSA were synthesized and particle characterization was carried out by transmission electron microscopy (TEM) and dynamic light scattering (DLS). An in vivo study was planned to investigate the therapeutic efficacy of MiADMSA-encapsulated solid lipid nanoparticles (Nano-MiADMSA; 50 mg/kg orally for 5 days) and compared it with bulk MiADMSA against sodium meta-arsenite exposed rats (25 ppm in drinking water, for 12 weeks) in male rats. The results suggested the size of Nano-MiADMSA was between 100-120 nm ranges. We noted enhanced chelating properties of Nano-MiADMSA compared with bulk MiADMSA as evident by the reversal of oxidative stress variables like blood δ-aminolevulinic acid dehydratase (δ-ALAD), Reactive Oxygen Species (ROS), Catalase activity, Superoxide Dismutase (SOD), Thiobarbituric Acid Reactive Substances (TBARS), Reduced Glutathione (GSH) and Oxidized Glutathione (GSSG), Glutathione Peroxidase (GPx), Glutathione-S-transferase (GST) and efficient removal of arsenic from the blood and tissues. Recoveries in neurobehavioral parameters further confirmed nano-MiADMSA to be more effective than bulk MiADMSA. We conclude that treatment with Nano-MiADMSA is a better therapeutic strategy than bulk MiADMSA in reducing the effects of arsenic-induced oxidative stress and associated neurobehavioral changes.
Collapse
Affiliation(s)
- Saba Naqvi
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - Prince Kumar
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, CRPF Base Camp, P.O. Mati, Sarojini Nagar, Lucknow, UP, 226002, India.
| |
Collapse
|
29
|
Zarrintaj P, Ramsey JD, Samadi A, Atoufi Z, Yazdi MK, Ganjali MR, Amirabad LM, Zangene E, Farokhi M, Formela K, Saeb MR, Mozafari M, Thomas S. Poloxamer: A versatile tri-block copolymer for biomedical applications. Acta Biomater 2020; 110:37-67. [PMID: 32417265 DOI: 10.1016/j.actbio.2020.04.028] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 11/16/2022]
Abstract
Poloxamers, also called Pluronic, belong to a unique class of synthetic tri-block copolymers containing central hydrophobic chains of poly(propylene oxide) sandwiched between two hydrophilic chains of poly(ethylene oxide). Some chemical characteristics of poloxamers such as temperature-dependent self-assembly and thermo-reversible behavior along with biocompatibility and physiochemical properties make poloxamer-based biomaterials promising candidates for biomedical application such as tissue engineering and drug delivery. The microstructure, bioactivity, and mechanical properties of poloxamers can be tailored to mimic the behavior of various types of tissues. Moreover, their amphiphilic nature and the potential to self-assemble into the micelles make them promising drug carriers with the ability to improve the drug availability to make cancer cells more vulnerable to drugs. Poloxamers are also used for the modification of hydrophobic tissue-engineered constructs. This article collects the recent advances in design and application of poloxamer-based biomaterials in tissue engineering, drug/gene delivery, theranostic devices, and bioinks for 3D printing. STATEMENT OF SIGNIFICANCE: Poloxamers, also called Pluronic, belong to a unique class of synthetic tri-block copolymers containing central hydrophobic chains of poly(propylene oxide) sandwiched between two hydrophilic chains of poly(ethylene oxide). The microstructure, bioactivity, and mechanical properties of poloxamers can be tailored to mimic the behavior of various types of tissues. Moreover, their amphiphilic nature and the potential to self-assemble into the micelles make them promising drug carriers with the ability to improve the drug availability to make cancer cells more vulnerable to drugs. However, no reports have systematically reviewed the critical role of poloxamer for biomedical applications. Research on poloxamers is growing today opening new scenarios that expand the potential of these biomaterials from "traditional" treatments to a new era of tissue engineering. To the best of our knowledge, this is the first review article in which such issue is systematically reviewed and critically discussed in the light of the existing literature.
Collapse
Affiliation(s)
- Payam Zarrintaj
- Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, United States
| | - Joshua D Ramsey
- Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, United States
| | - Ali Samadi
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia, Iran
| | - Zhaleh Atoufi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohsen Khodadadi Yazdi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Reza Ganjali
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran; Biosensor Research Center, Endocrinology & Metabolism Molecular-Cellular Sciences, University of Tehran, Tehran, Iran
| | | | - Ehsan Zangene
- Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Krzysztof Formela
- Department of Polymer Technology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Mohammad Reza Saeb
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran.
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sabu Thomas
- School of Chemical Sciences, M G University, Kottayam 686560, Kerala, India
| |
Collapse
|
30
|
Gao C, Wang H, Wang T, Luo C, Wang Z, Zhang M, Chen X, Tao L. Platelet regulates neuroinflammation and restores blood-brain barrier integrity in a mouse model of traumatic brain injury. J Neurochem 2020; 154:190-204. [PMID: 32048302 DOI: 10.1111/jnc.14983] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 01/14/2020] [Accepted: 02/09/2020] [Indexed: 12/19/2022]
Abstract
Neuroinflammation accompanied by microglial activation triggers multiple cell death after traumatic brain injury (TBI). The secondary injury caused by inflammation may persist for a long time. Recently, platelet C-type lectin-like 2 receptor (CLEC-2) has been shown to regulate inflammation in certain diseases. However, its possible effects on TBI remain poorly understood. Here, we aimed to investigate the role of platelet CLEC-2 in the pathological process of neuroinflammation after TBI. In this study, mice were subjected to sham or controlled cortical impact injury, and arbitrarily received recombinant platelet CLEC-2. In parallel, BV2 cells were treated with lipopolysaccharide (LPS) to mimic microglial activation after TBI. Primary endothelial cells were also subjected to LPS in order to replicate the inflammatory damage caused by TBI. We used western blot analysis, reverse transcription polymerase chain reaction (RT-PCR), and immunostaining to evaluate the role of platelet CLEC-2 in TBI. In conditional knock out platelet CLEC-2 mice, trauma worsened the integrity of the blood-brain barrier and amplified the release of inflammatory cytokines. In wild type mice subjected to controlled cortical impact injury, recombinant platelet CLEC-2 administration altered the secretion of inflammatory cytokines, reduced brain edema, and improved neurological function. In vitro, the polarization phenotype of microglia induced by LPS was transformed by recombinant platelet CLEC-2, and this conversion depended on the mammalian target of rapamycin (mTOR) pathway. Endothelial cell injury by LPS was ameliorated when microglia expressed mostly M2 phenotype markers. In conclusion, platelet CLEC-2 regulates trauma-induced neuroinflammation and restores blood-brain barrier integrity.
Collapse
Affiliation(s)
- Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Haochen Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Tao Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Zufeng Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Mingyang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Xiping Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
31
|
The Function and Mechanisms of Autophagy in Traumatic Brain Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:635-648. [PMID: 32671781 DOI: 10.1007/978-981-15-4272-5_46] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Traumatic brain injury (TBI) is one of the most common causes of long-term disability and death worldwide. Autophagy is activated and autophagic flux is impaired following TBI. But the controversial roles and underlying mechanisms of autophagy after TBI are not clear. This chapter will update the current state of knowledge in the process of autophagy, the roles of autophagy in TBI as well as some upstream moleculars and pharmacological regulators of autophagy involved in TBI. We also discuss autophagy mechanism-based preclinical pharmacological intervention. These observations make autophagy an attractive therapeutic target for developing new therapeutic strategies to achieve better outcomes for patients suffering from TBI.
Collapse
|
32
|
Mayer D, Armstrong D, Schultz G, Percival S, Malone M, Romanelli M, Keast D, Jeffery S. Cell salvage in acute and chronic wounds: a potential treatment strategy. Experimental data and early clinical results. J Wound Care 2019; 27:594-605. [PMID: 30204575 DOI: 10.12968/jowc.2018.27.9.594] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On 9 May 2018, the authors took part in a closed panel discussion on the impact of cell salvage in acute and chronic wounds. The goal was to deliberate the possible use of plurogel micelle matrix (PMM) as a new treatment strategy for wound healing and the authors openly shared their experiences, thoughts, experimental data and early clinical results. The outcome of the panel discussion has been abridged in this paper. The cell membrane consists of a lipid bilayer, which provides a diffusion barrier separating the inside of a cell from its environment. Cell membrane injury can result in acute cellular necrosis when defects are too large and cannot be resealed. There is a potential hazard to the body when these dying cells release endogenous alarm signals referred to as 'damage (or danger) associated molecular patterns' (DAMPs), which trigger the innate immune system and modulate inflammation. Cell salvage by membrane resealing is a promising target to ensure the survival of the individual cell and prevention of further tissue degeneration by inflammatory processes. Non-ionic surfactants such as poloxamers, poloxamines and PMM have the potential to resuscitate cells by inserting themselves into damaged membranes and stabilising the unstable portions of the lipid bilayers. The amphiphilic properties of these molecules are amenable to insertion into cell wall defects and so can play a crucial, reparative role. This new approach to cell rescue or salvage has gained increasing interest as several clinical conditions have been linked to cell membrane injury via oxidative stress-mediated lipid peroxidation or thermal disruption. The repair of the cell membrane is an important step in salvaging cells from necrosis to prevent further tissue degeneration by inflammatory processes. This is applicable to acute burns and chronic wounds such as diabetic foot ulcers (DFUs), chronic venous leg ulcers (VLUs), and pressure ulcers (PUs). Experimental data shows that PMM is biocompatible and able to insert itself into damaged membranes, salvaging their barrier function and aiding cell survival. Moreover, the six case studies presented in this paper reveal the potential of this treatment strategy.
Collapse
Affiliation(s)
| | | | | | | | - Matt Malone
- South West Sydney Limb Preservation and Wound Research, South Western Syndey Local Health District, Ingham Institute of Applied Medical Research, Syndey, Australia and Infectious Disease and Microbiology, School of Medicine, Western Sydney University, Sydney, Australia
| | | | | | - Steven Jeffery
- The Queen Elizabeth Hospital, Birmingham, UK and Birmingham City University
| |
Collapse
|
33
|
Chen B, Tjahja J, Malla S, Liebman C, Cho M. Astrocyte Viability and Functionality in Spatially Confined Microcavitation Zone. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4889-4899. [PMID: 30638362 DOI: 10.1021/acsami.8b21410] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Blast-induced traumatic brain injury (bTBI) can result in cell/tissue damage and lead to clinical and neuropsychiatric symptoms. Shock waves from a blast propagate through the brain and initiate cascades of mechanical and physiological events that can adversely affect the brain function. Although studies using animal models and brain slices have shown macroscale changes in the brain tissue in response to blast, systematic elucidation of coupling mechanisms is currently lacking. One mechanism that has been postulated and demonstrated repeatedly is the blast-induced generation and subsequent collapse of micron-size bubbles (i.e., microcavitation). Using a custom-designed exposure system, we have previously reported that upon collapsing of microbubbles, astrocytes exhibited changes in the cell viability, cellular biomechanics, production of reactive oxygen species, and activation of apoptotic signaling pathways. In this paper, we have applied microfabrication techniques and seeded astrocytes in a spatially controlled manner to determine the extent of cell damage from the site of the collapse of microbubbles. Such a novel experimental design is proven to facilitate our effort to examine the altered cell viability and functionality by monitoring the transient calcium spiking activity in real-time. We now report that the effect of microcavitation depends on the distance from which cells are seeded, and the cell functionality assessed by calcium dynamics is significantly diminished in the cells located within ∼800 μm of the collapsing microbubbles. Both calcium influx across the cell membrane via N-type calcium channels and intracellular calcium store are altered in response to microcavitation. Finally, the FDA-approved poloxamer 188 (P188) was used to reconstitute the compromised cell membrane and restore the cell's reparative capability. This finding may lead to a feasible treatment for partially mitigating the tissue damage associated with bTBI.
Collapse
Affiliation(s)
- Bo Chen
- Department of Bioengineering , University of Texas at Arlington , Arlington , Texas 76019 , United States
| | - Jessica Tjahja
- Department of Bioengineering , University of Texas at Arlington , Arlington , Texas 76019 , United States
| | - Sameep Malla
- Department of Bioengineering , University of Texas at Arlington , Arlington , Texas 76019 , United States
| | - Caleb Liebman
- Department of Bioengineering , University of Texas at Arlington , Arlington , Texas 76019 , United States
| | - Michael Cho
- Department of Bioengineering , University of Texas at Arlington , Arlington , Texas 76019 , United States
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Survival with favorable neurological function after cardiac arrest remains low. The purpose of this review is to identify recent advances that focus on neuroprotection during cardiopulmonary resuscitation (CPR). RECENT FINDINGS Multiple strategies have been shown to enhance neuroprotection during CPR. Brain perfusion during CPR is increased with therapies such as active compression decompression CPR and intrathoracic pressure regulation that improve cardiac preload and decrease intracranial pressure. Head Up CPR has been shown to decrease intracranial pressure thereby increasing cerebral perfusion pressure and cerebral blood flow. Sodium nitroprusside enhanced CPR increases cerebral perfusion, facilitates heat exchange, and improves neurologic survival in swine after cardiac arrest. Postconditioning has been administered during CPR in laboratory settings. Poloxamer 188, a membrane stabilizer, and ischemic postconditioning have been shown to improve cardiac and neural function after cardiac arrest in animal models. Postconditioning with inhaled gases protects the myocardium, with more evidence mounting for the potential for neural protection. SUMMARY Multiple promising neuroprotective therapies are being developed in animal models of cardiac arrest, and are in early stages of human trials. These therapies have the potential to be bundled together to improve rates of favorable neurological survival after cardiac arrest.
Collapse
|
35
|
Guo RM, Li QL, Zhong LR, Guo Y, Jiao J, Chen SQ, Wang J, Zhang Y. Brain MRI findings in acute hepatic encephalopathy in liver transplant recipients. Acta Neurol Belg 2018; 118:251-258. [PMID: 29275444 DOI: 10.1007/s13760-017-0875-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Acute hepatic encephalopathy has significant morbidity and mortality in liver transplant recipients unless it is promptly treated. We evaluated the brain magnetic resonance (MR) imaging findings associated with acute hepatic encephalopathy in transplant recipients. We retrospectively reviewed the clinical and imaging data and outcomes of twenty-five liver transplant patients (16 male; mean age, 49.3 years) with clinically diagnosed acute hepatic encephalopathy and forty liver transplant patients (20 males; mean age, 45.5 years) without neurological symptoms suggestive of hepatic encephalopathy at our institution. Bilateral symmetric hyperintensities of the insular cortex and cingulate gyrus were observed in twenty-one patients (84.00%), bilateral symmetric extensive increased cortical signal intensity (involving two or more regions) was observed in 72.00% of the patients, leptomeningeal enhancement in 73.68%, and visualization of prominent venules in 52.00%. The most common symptom at diagnosis was rigidity (n = 14), and the plasma ammonia levels ranged from 68.63 to 192.16 μmol/L. After active treatment, 17 patients gradually recovered, four patients suffered from mild or moderate neurologic deficits, and four patients with widespread brain edema died. The specific brain MR imaging features were bilateral symmetric increased cortical signal intensity, especially in the insular cortex and cingulate gyrus, leptomeningeal enhancement, visualization of the prominent venules, and widespread brain edema. These features may indicate poor prognosis and should alert radiologists to the possibility of acute hepatic encephalopathy in liver transplant recipients and encourage clinicians to prepare appropriate treatment in advance.
Collapse
Affiliation(s)
- Ruo-Mi Guo
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Nuclear Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Qing-Ling Li
- Department of VIP Medical Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Li-Ru Zhong
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yu Guo
- Department of VIP Medical Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Ju Jiao
- Department of Nuclear Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Shao-Qiong Chen
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
36
|
Lee CAA, Seo HS, Armien AG, Bates FS, Tolar J, Azarin SM. Modeling and rescue of defective blood-brain barrier function of induced brain microvascular endothelial cells from childhood cerebral adrenoleukodystrophy patients. Fluids Barriers CNS 2018; 15:9. [PMID: 29615068 PMCID: PMC5883398 DOI: 10.1186/s12987-018-0094-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/06/2018] [Indexed: 01/12/2023] Open
Abstract
Background X-linked adrenoleukodystrophy (X-ALD) is caused by mutations in the ABCD1 gene. 40% of X-ALD patients will convert to the deadly childhood cerebral form (ccALD) characterized by increased permeability of the brain endothelium that constitutes the blood–brain barrier (BBB). Mutation information and molecular markers investigated to date are not predictive of conversion. Prior reports have focused on toxic metabolic byproducts and reactive oxygen species as instigators of cerebral inflammation and subsequent immune cell invasion leading to BBB breakdown. This study focuses on the BBB itself and evaluates differences in brain endothelium integrity using cells from ccALD patients and wild-type (WT) controls. Methods The blood–brain barrier of ccALD patients and WT controls was modeled using directed differentiation of induced pluripotent stem cells (iPSCs) into induced brain microvascular endothelial cells (iBMECs). Immunocytochemistry and PCR confirmed characteristic expression of brain microvascular endothelial cell (BMEC) markers. Barrier properties of iBMECs were measured via trans-endothelial electrical resistance (TEER), sodium fluorescein permeability, and frayed junction analysis. Electron microscopy and RNA-seq were used to further characterize disease-specific differences. Oil-Red-O staining was used to quantify differences in lipid accumulation. To evaluate whether treatment with block copolymers of poly(ethylene oxide) and poly(propylene oxide) (PEO–PPO) could mitigate defective properties, ccALD-iBMECs were treated with PEO–PPO block copolymers and their barrier properties and lipid accumulation levels were quantified. Results iBMECs from patients with ccALD had significantly decreased TEER (2592 ± 110 Ω cm2) compared to WT controls (5001 ± 172 Ω cm2). They also accumulated lipid droplets to a greater extent than WT-iBMECs. Upon treatment with a PEO–PPO diblock copolymer during the differentiation process, an increase in TEER and a reduction in lipid accumulation were observed for the polymer treated ccALD-iBMECs compared to untreated controls. Conclusions The finding that BBB integrity is decreased in ccALD and can be rescued with block copolymers opens the door for the discovery of BBB-specific molecular markers that can indicate the onset of ccALD and has therapeutic implications for preventing the conversion to ccALD. Electronic supplementary material The online version of this article (10.1186/s12987-018-0094-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Catherine A A Lee
- Department of Genetics and Cell Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hannah S Seo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anibal G Armien
- Ultrastructural Pathology Unit, Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, 55108, USA
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jakub Tolar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
37
|
Li D, Ji JX, Xu YT, Ni HB, Rui Q, Liu HX, Jiang F, Gao R, Chen G. Inhibition of Lats1/p-YAP1 pathway mitigates neuronal apoptosis and neurological deficits in a rat model of traumatic brain injury. CNS Neurosci Ther 2018; 24:906-916. [PMID: 29488331 DOI: 10.1111/cns.12833] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/20/2022] Open
Abstract
AIMS To investigate the roles of Lats1/p-YAP1 pathway in TBI-induced neuronal apoptosis and neurological deficits in rats. RESULTS We found that Lats1 and YAP1 were expressed in cerebral cortex neurons of Sprague-Dawley rats, and the phosphorylation levels of Lats1 and YAP1 in injured regions were significantly increased after TBI. Furthermore, inhibition of Lats1 not only decreased the level of p-YAP1, but also attenuated neuronal apoptosis and neurological impairment. CONCLUSIONS Our work demonstrates that inhibition of Lats1/p-YAP1 pathway mitigates neuronal apoptosis and neurological deficits in a rat model of TBI.
Collapse
Affiliation(s)
- Di Li
- Department of Neurosurgery and Translational Medicine Center, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Jia-Xuan Ji
- Department of Neurosurgery, Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Suzhou, China
| | - Yi-Tian Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Hai-Bo Ni
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Qin Rui
- Clinical laboratory, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Hui-Xiang Liu
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Feng Jiang
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Zhang Y, Chopp M, Emanuele M, Zhang L, Zhang ZG, Lu M, Zhang T, Mahmood A, Xiong Y. Treatment of Traumatic Brain Injury with Vepoloxamer (Purified Poloxamer 188). J Neurotrauma 2018; 35:661-670. [PMID: 29121826 DOI: 10.1089/neu.2017.5284] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vepoloxamer is an amphipathic polymer that has shown potent hemorrheologic, cytoprotective, and anti-inflammatory effects in both pre-clinical and clinical studies. This study was designed to investigate the therapeutic effects of vepoloxamer on sensorimotor and cognitive functional recovery in rats after traumatic brain injury (TBI) induced by controlled cortical impact. Young adult male Wistar rats were randomly divided into the following groups: 1) sham; 2) saline; or 3) vepoloxamer. Vepoloxamer (300 mg/kg) or saline was administered over 60 min via intravenous infusion into tail veins starting at 2 h post-injury. Sensorimotor function and spatial learning were assessed using a modified neurological severity score and foot fault test, and Morris water maze test, respectively. The animals were sacrificed 35 days after injury and their brains were processed for measurement of lesion volume and neuroinflammation. Compared with the saline treatment, vepoloxamer initiated 2 h post-injury significantly improved sensorimotor functional recovery (Days 1-35; p < 0.0001) and spatial learning (Days 32-35; p < 0.0001), reduced cortical lesion volume by 20%, and reduced activation of microglia/macrophages and astrogliosis in many brain regions including injured cortex, corpus callosum, and hippocampus, as well as normalized the bleeding time and reduced brain hemorrhage and microthrombosis formation. In summary, vepoloxamer treatment initiated 2 h post-injury provides neuroprotection and anti-inflammation in rats after TBI and improves functional outcome, indicating that vepoloxamer treatment may have potential value for treatment of TBI. Further investigation of the optimal dose and therapeutic window of vepoloxamer treatment for TBI and the mechanisms underlying beneficial effects are warranted.
Collapse
Affiliation(s)
- Yanlu Zhang
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Michael Chopp
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan.,3 Department of Physics, Oakland University , Rochester, Michigan
| | | | - Li Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Zheng Gang Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Mei Lu
- 5 Department of Biostatistics and Research Epidemiology, Henry Ford Hospital , Detroit, Michigan
| | - Talan Zhang
- 5 Department of Biostatistics and Research Epidemiology, Henry Ford Hospital , Detroit, Michigan
| | - Asim Mahmood
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Ye Xiong
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
39
|
Borsini A, Cattaneo A, Malpighi C, Thuret S, Harrison NA, MRC ImmunoPsychiatry Consortium, Zunszain PA, Pariante CM. Interferon-Alpha Reduces Human Hippocampal Neurogenesis and Increases Apoptosis via Activation of Distinct STAT1-Dependent Mechanisms. Int J Neuropsychopharmacol 2017; 21:187-200. [PMID: 29040650 PMCID: PMC5793815 DOI: 10.1093/ijnp/pyx083] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In humans, interferon-α treatment for chronic viral hepatitis is a well-recognized clinical model for inflammation-induced depression, but the molecular mechanisms underlying these effects are not clear. Following peripheral administration in rodents, interferon-α induces signal transducer and activator of transcription-1 (STAT1) within the hippocampus and disrupts hippocampal neurogenesis. METHODS We used the human hippocampal progenitor cell line HPC0A07/03C to evaluate the effects of 2 concentrations of interferon-α, similar to those observed in human serum during its therapeutic use (500 pg/mL and 5000 pg/mL), on neurogenesis and apoptosis. RESULTS Both concentrations of interferon-α decreased hippocampal neurogenesis, with the high concentration also increasing apoptosis. Moreover, interferon-α increased the expression of interferon-stimulated gene 15 (ISG15), ubiquitin-specific peptidase 18 (USP18), and interleukin-6 (IL-6) via activation of STAT1. Like interferon-α, co-treatment with a combination of ISG15, USP18, and IL-6 was able to reduce neurogenesis and enhance apoptosis via further downstream activation of STAT1. Further experiments showed that ISG15 and USP18 mediated the interferon-α-induced reduction in neurogenesis (potentially through upregulation of the ISGylation-related proteins UBA7, UBE2L6, and HERC5), while IL-6 mediated the interferon-α-induced increase in apoptosis (potentially through downregulation of aquaporin 4). Using transcriptomic analyses, we showed that interferon-α regulated pathways involved in oxidative stress and immune response (e.g., Nuclear Factor (erythroid-derived 2)-like 2 [Nrf2] and interferon regulatory factor [IRF] signaling pathway), neuronal formation (e.g., CAMP response element-binding protein [CREB] signaling), and cell death regulation (e.g., tumor protein(p)53 signaling). CONCLUSIONS We identify novel molecular mechanisms mediating the effects of interferon-α on the human hippocampus potentially involved in inflammation-induced neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Alessandra Borsini
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, United Kingdom,Correspondence: Alessandra Borsini, PhD, Stress, Psychiatry and Immunology Lab and Perinatal Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King’s College London, Cutcombe Road, London, SE5 9RT ()
| | - Annamaria Cattaneo
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| | - Chiara Malpighi
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| | - Sandrine Thuret
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, United Kingdom
| | - Neil A Harrison
- University of Sussex, Department of Neuroscience, Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Patricia A Zunszain
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom
| | - Carmine M Pariante
- Section of Stress, Psychiatry and Immunology and Perinatal Psychiatry, King’s College London, London, United Kingdom,Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, London, United Kingdom,IRCCS Fatebenefratelli Institute, Biological Psychiatry Laboratory, Brescia, Italy
| |
Collapse
|
40
|
Zhou J, Liu T, Cui H, Fan R, Zhang C, Peng W, Yang A, Zhu L, Wang Y, Tang T. Xuefu zhuyu decoction improves cognitive impairment in experimental traumatic brain injury via synaptic regulation. Oncotarget 2017; 8:72069-72081. [PMID: 29069769 PMCID: PMC5641112 DOI: 10.18632/oncotarget.18895] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/12/2017] [Indexed: 11/25/2022] Open
Abstract
An overarching consequence of traumatic brain injury (TBI) is the cognitive impairment. It may hinder individual performance of daily tasks and determine people's subjective well-being. The damage to synaptic plasticity, one of the key mechanisms of cognitive dysfunction, becomes the potential therapeutic strategy of TBI. In this study, we aimed to investigate whether Xuefu Zhuyu Decoction (XFZYD), a traditional Chinese medicine, provided a synaptic regulation to improve cognitive disorder following TBI. Morris water maze and modified neurological severity scores were performed to assess the neurological and cognitive abilities. The PubChem Compound IDs of the major compounds of XFZYD were submitted into BATMAN-TCM, an online bioinformatics analysis tool, to predict the druggable targets related to synaptic function. Furthermore, we validated the prediction through immunohistochemical, RT-PCR and western blot analyses. We found that XFZYD enhanced neuroprotection, simultaneously improved learning and memory performances in controlled cortical impact rats. Bioinformatics analysis revealed that the improvements of XFZYD implied the Long-term potentiation relative proteins including NMDAR1, CaMKII and GAP-43. The further confirmation of molecular biological studies confirmed that XFZYD upregulated the mRNA and protein levels of NMDAR1, CaMKII and GAP-43. Pharmacological synaptic regulation of XFZYD could provide a novel therapeutic strategy for cognitive impairment following TBI.
Collapse
Affiliation(s)
- Jing Zhou
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Liu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
- Department of Gerontology, Traditional Chinese Medicine Hospital Affiliate to Xinjiang Medical University, 830000 Urumqi, China
| | - Hanjin Cui
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Rong Fan
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Chunhu Zhang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Weijun Peng
- Department of Traditional Chinese Medicine, 2nd Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Ali Yang
- Department of Neurology, Henan Province People’ Hospital, 450003 Zhengzhou, China
| | - Lin Zhu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| |
Collapse
|
41
|
Zhu W, Chi N, Zou P, Chen H, Tang G, Zhao W. Effect of docosahexaenoic acid on traumatic brain injury in rats. Exp Ther Med 2017; 14:4411-4416. [PMID: 29075341 DOI: 10.3892/etm.2017.5054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 06/20/2017] [Indexed: 12/23/2022] Open
Abstract
The present study aimed to investigate the protective effects of docosahexaenoic acid (DHA) on traumatic brain injury (TBI) in rats. A model of TBI was induced by lateral fluid percussion injury in adult rats and rats were randomly divided into the TBI-model group, TBI-low DHA group and TBI-high DHA group, while other healthy rats were assigned to the sham-operated group. Motor recovery was tested with beam-walking trials at 2, 7 and 15 days post-TBI. Cognitive recovery was tested with Morris water maze trials at 15 days post-TBI. The expression levels of caspase-3, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were measured by western blotting. DHA protected against motor deficits induced by TBI in beam walking tests. All TBI-model groups had longer escape latency and swimming distances than the sham groups. Compared with the TBI-low DHA group, the TBI-high DHA group demonstrated shorter escape latency and swimming distances. DHA inhibited the expression of caspase-3 and the inhibition effect was more obvious at a high dosage. Furthermore, DHA dose-dependently rescued neurons by upregulating the Bcl-2:Bax ratio. DHA supplementation was a viable strategy to mitigate injury from TBI.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Nan Chi
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Peng Zou
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Hongguang Chen
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Guotai Tang
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Wei Zhao
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
42
|
Kim M, Haman KJ, Houang EM, Zhang W, Yannopoulos D, Metzger JM, Bates FS, Hackel BJ. PEO-PPO Diblock Copolymers Protect Myoblasts from Hypo-Osmotic Stress In Vitro Dependent on Copolymer Size, Composition, and Architecture. Biomacromolecules 2017; 18:2090-2101. [PMID: 28535058 DOI: 10.1021/acs.biomac.7b00419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Poloxamer 188, a triblock copolymer of poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO), protects cellular membranes from various stresses. Though numerous block copolymer variants exist, evaluation of alternative architecture, composition, and size has been minimal. Herein, cultured murine myoblasts are exposed to the stresses of hypotonic shock and isotonic recovery, and membrane integrity was evaluated by quantifying release of lactate dehydrogenase. Comparative evaluation of a systematic set of PEO-PPO diblock and PEO-PPO-PEO triblock copolymers demonstrates that the diblock architecture can be protective in vitro. Short PPO blocks hinder protection with >9 PPO units needed for protection at 150 μM and >16 units needed at 14 μM. Addition of a tert-butyl end group enhances protection at reduced concentration. When the end group and PPO length are fixed, increasing the PEO length improves protection. This systematic evaluation establishes a new in vitro screening tool for evaluating membrane-sealing amphiphiles and provides mechanistic insight to guide future copolymer design for membrane stabilization in vivo.
Collapse
Affiliation(s)
- Mihee Kim
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Karen J Haman
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Evelyne M Houang
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Wenjia Zhang
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Demetris Yannopoulos
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Joseph M Metzger
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, ‡Department of Integrative Biology and Physiology, and §Department of Medicine, Cardiovascular Division, University of Minnesota , Minneapolis, Minnesota 55455, United States
| |
Collapse
|
43
|
Poellmann MJ, Lee RC. Repair and Regeneration of the Wounded Cell Membrane. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0031-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Bao H, Yang X, Zhuang Y, Huang Y, Wang T, Zhang M, Dai D, Wang S, Xiao H, Huang G, Kuai J, Tao L. The effects of poloxamer 188 on the autophagy induced by traumatic brain injury. Neurosci Lett 2016; 634:7-12. [DOI: 10.1016/j.neulet.2016.09.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/18/2016] [Accepted: 09/28/2016] [Indexed: 01/22/2023]
|
45
|
Du G, Zhao Z, Chen Y, Li Z, Tian Y, Liu Z, Liu B, Song J. Quercetin attenuates neuronal autophagy and apoptosis in rat traumatic brain injury model via activation of PI3K/Akt signaling pathway. Neurol Res 2016; 38:1012-1019. [PMID: 27690831 DOI: 10.1080/01616412.2016.1240393] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Neuronal autophagy and apoptosis play an irreplaceable role in brain injury pathogenesis and may represent a hopeful target for treatment. Previous studies have demonstrated that administration of quercetin-attenuated brain damage in a variety of brain injury models including traumatic brain injury (TBI). However, whether PI3K/Akt signaling pathway mediates the neuroprotection of quercetin following TBI is not well clarified. We sought to propose a hypothesis that quercetin could attenuate neuronal autophagy and apoptosis via enhancing PI3K/Akt signaling. METHODS All rats were randomly arranged into four groups as follows: sham group (n = 25), TBI group (n = 25), TBI + quercetin group (n = 25), TBI + quercetin + LY294002 group (n = 25). Quercetin (Sigma, USA, dissolved in 0.9% saline solution) was administered intraperitoneally at a dose of 50 mg/kg at 30 min, 12 h, and 24 h after TBI. The neurological impairment and spatial cognitive function was assessed by the neurologic severity score and Morris water maze, respectively. Immunohistochemistry staining and western blotting was used to evaluate the expression of LC3, p-Akt, caspase-3, Bcl-2, and Bax. RESULTS Quercetin treatment significantly attenuated TBI-induced neurological impairment (1-3 days, p < 0.05) and improved cognitive function (5-8 days, p < 0.05). Double immunolabeling demonstrated that quercetin significantly reduced the LC3-positive cells co-labeled with NeuN, whereas significantly enhanced p-Akt-positive cells co-labeled with NeuN. Furthermore, quercetin treatment reduced the expression of LC3、caspase-3 and Bax levels induced following TBI (p < 0.05), and increased the expression of p-Akt and Bcl-2 at 48 h (p < 0.05). CONCLUSION In conclusion, our observations indicate that post-injury treatment with quercetin could inhibit neuronal autophagy and apoptosis in the hippocampus in a rat model of TBI. The neuroprotective effects of quercetin may be related to modulation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Guoliang Du
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Zongmao Zhao
- b Department of Neurosurgery , Second Hospital of Hebei Medical University , Shijiazhuang , People's Republic of China
| | - Yonghan Chen
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Zonghao Li
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Yaohui Tian
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Zhifeng Liu
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Bin Liu
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| | - Jianqiang Song
- a Department of Neurosurgery , Central Hospital of Cangzhou , Cangzhou , People's Republic of China
| |
Collapse
|
46
|
Bao HJ, Qiu HY, Kuai JX, Song CJ, Wang SX, Wang CQ, Peng HB, Han WC, Wu YP. Apelin-13 as a novel target for intervention in secondary injury after traumatic brain injury. Neural Regen Res 2016; 11:1128-33. [PMID: 27630697 PMCID: PMC4994456 DOI: 10.4103/1673-5374.187049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The adipocytokine, apelin-13, is an abundantly expressed peptide in the nervous system. Apelin-13 protects the brain against ischemia/reperfusion injury and attenuates traumatic brain injury by suppressing autophagy. However, secondary apelin-13 effects on traumatic brain injury-induced neural cell death and blood-brain barrier integrity are still not clear. Here, we found that apelin-13 significantly decreases cerebral water content, mitigates blood-brain barrier destruction, reduces aquaporin-4 expression, diminishes caspase-3 and Bax expression in the cerebral cortex and hippocampus, and reduces apoptosis. These results show that apelin-13 attenuates secondary injury after traumatic brain injury and exerts a neuroprotective effect.
Collapse
Affiliation(s)
- Hai-jun Bao
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Hai-yang Qiu
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Jin-xia Kuai
- Department of Forensic Medicine, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Cheng-jie Song
- Department of Physiology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Shao-xian Wang
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Chao-qun Wang
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Hua-bin Peng
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Wen-can Han
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Yong-ping Wu
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu Province, China,Correspondence to: Yong-ping Wu, .
| |
Collapse
|
47
|
Bao H, Yang X, Huang Y, Qiu H, Huang G, Xiao H, Kuai J. The neuroprotective effect of apelin-13 in a mouse model of intracerebral hemorrhage. Neurosci Lett 2016; 628:219-24. [PMID: 27343409 DOI: 10.1016/j.neulet.2016.06.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022]
Abstract
Adipocytokine apelin-13 is a peptide which could reportedly protect the brain against ischemic reperfusion injury and traumatic brain injury (TBI). Whether apelin-13 has any roles to play in intracerebral hemorrhage (ICH) has not been clarified. We aimed to investigate the roles of apelin-13 in ICH and effects on ICH-induced apoptosis. Firstly, CD-1 mice were subjected to infusion of Type IV collagenase (to induce ICH) or saline (for shams) into the left striatum. ICH animals received intracerebroventricular administration of vehicle, apelin-13 (50μg dissolved in 5μl saline) immediately after ICH. The motor function and the cerebral water content (CWC) as well as blood brain barrier (BBB) disruption were measured, coupled with determination of ICH-induced neural cell death by Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL). The apoptosis-associated proteins caspase-3 and Bcl-2 as well as the brain edema-associated proteins aquaporin-4 (AQP4) and MMP-9 were all assessed with western blotting. The results showed that apelin-13 decreased CWC and reduced Evans blue leakage into injured hemispheres, with the motor function significantly improved. Additionally, apelin-13 also acutely decreased the number of ICH-induced TUNEL-positive (TUNEL(+)) cells at 48h after ICH. The expressions of AQP4, MMP-9, caspse-3 and Bcl-2 were all downregulated by apelin-13 at 24h and 48h after ICH. All these results revealed that apelin-13 attenuated brain edema and reduced cellular death by suppressing apoptosis after ICH in mice.
Collapse
Affiliation(s)
- Haijun Bao
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Xiaofang Yang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - YuXiu Huang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Haiyang Qiu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Genping Huang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Hua Xiao
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Jinxia Kuai
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
48
|
Zander NE, Piehler T, Banton R, Benjamin R. Effects of repetitive low-pressure explosive blast on primary neurons and mixed cultures. J Neurosci Res 2016; 94:827-36. [PMID: 27317559 DOI: 10.1002/jnr.23786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/28/2016] [Accepted: 05/23/2016] [Indexed: 02/05/2023]
Abstract
Repetitive mild traumatic brain injury represents a considerable health concern, particularly for athletes and military personnel. For blast-induced brain injury, threshold shock-impulse levels required to induce such injuries and cumulative effects with single and/or multiple exposures are not well characterized. Currently, there is no established in vitro experimental model with blast pressure waves generated by live explosives. This study presents results of primary neurons and mixed cultures subjected to our unique in vitro indoor experimental platform that uses real military explosive charges to probe the effects of primary explosive blast at the cellular level. The effects of the blast on membrane permeability, generation of reactive oxygen species (ROS), uptake of sodium ions, intracellular calcium, and release of glutamate were probed 2 and 24 hr postblast. Significant changes in membrane permeability and sodium uptake among the sham, single-blast-injured, and triple-blast-injured samples were observed. A significant increase in ROS and glutamate release was observed for the triple-blast-injured samples compared with the sham. Changes in intracellular calcium were not significant. These results suggest that blast exposure disrupts the integrity of the plasma membrane, leading to the upset of ion homeostasis, formation of ROS, and glutamate release. Published 2016. †This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Nicole E Zander
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Thuvan Piehler
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Rohan Banton
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Richard Benjamin
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| |
Collapse
|
49
|
Lurie KG, Nemergut EC, Yannopoulos D, Sweeney M. The Physiology of Cardiopulmonary Resuscitation. Anesth Analg 2016; 122:767-783. [DOI: 10.1213/ane.0000000000000926] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
50
|
Zhang L, Ding K, Wang H, Wu Y, Xu J. Traumatic Brain Injury-Induced Neuronal Apoptosis is Reduced Through Modulation of PI3K and Autophagy Pathways in Mouse by FTY720. Cell Mol Neurobiol 2016; 36:131-42. [PMID: 26099903 PMCID: PMC11482378 DOI: 10.1007/s10571-015-0227-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/12/2015] [Indexed: 01/13/2023]
Abstract
FTY720 is a synthetic compound produced by modification of metabolite from Isaria sinclairii. It is a novel type of immunosuppressive agent inhibiting lymphocyte egress from secondary lymphoid tissues, thereby causing peripheral lymphopenia. Growing evidences have suggested that apoptosis and autophagy were involved in the secondary brain injury after traumatic brain injury (TBI) although FTY720 exerted neuroprotective effects in a variety of neurological diseases except TBI. The present study was aimed to investigate the role of FTY720 in a mouse model of TBI. In experiment 1, ICR mice were divided into four groups: sham group, TBI group, TBI + vehicle group, and TBI + FTY720 group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 administration after TBI improved neurobehavioral function, alleviated brain edema, accompanied by modulation of apoptotic indicators such as Bcl-2, Bcl-xL, Bax, and cytochrome c. In experiment 2, ICR mice were also divided into four groups: sham group, TBI + vehicle group, TBI + FTY720 group, and TBI + FTY720 + inhibitors group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 increased the expression of phospho-protein kinase B (AKT) and some autophagy markers such as LC3 and Beclin 1. In addition, the apoptosis inhibition effect of FTY720 was partly abrogated by the phosphatidylinositide 3-kinases (PI3K)/AKT pathway inhibitor LY294002 and autophagy inhibitor 3-methyladenine. Collectively, our data provide the first evidence that FTY720 exerted neuroprotective effects after TBI, at least in part, through the activation of PI3K/AKT pathway and autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Yong Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Jianguo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| |
Collapse
|