Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurooncol ; 165(1): 209-218, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37889443

ABSTRACT

PURPOSE: New treatments are needed to improve the overall survival of patients with glioblastoma Metformin is known for anti-tumorigenic effects in cancers, including breast and pancreas cancers. In this study, we assessed the association between metformin use and overall survival in glioblastoma patients. METHODS: We retrospectively studied 241 patients who underwent surgery at diagnosis of glioblastoma between 2014 and 2018. Metformin was used for pre-existing type 2 diabetes mellitus or in the prevention or management of glucocorticoid induced hyperglycemia. Kaplan-Meier curves and log-rank p test were used for univariate analysis. Cox-proportional hazards model was used to generate adjusted hazard ratios for multivariate analysis. RESULTS: Metformin use was associated with longer survival in patients with tumors that had a methylated O6-methylguanine DNA methyltransferase gene (MGMT) promoter (484 days 95% CI: 56-911 vs. 394 days 95% CI: 249-538, Log-Rank test: 6.5, p = 0.01). Cox regression analysis shows that metformin associates with lower risk of death at 2 years in patients with glioblastoma containing a methylated MGMT promoter (aHR = 0.497, 95% CI 0.26-0.93, p = 0.028). CONCLUSION: Our findings suggest a survival benefit with metformin use in patients with glioblastomas having methylation of the MGMT promoter.


Subject(s)
Brain Neoplasms , Diabetes Mellitus, Type 2 , Glioblastoma , Metformin , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Methyltransferases/genetics , Retrospective Studies , Metformin/therapeutic use , Diabetes Mellitus, Type 2/genetics , DNA Methylation , DNA Modification Methylases/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Silencing , DNA Repair Enzymes/genetics , Prognosis , Tumor Suppressor Proteins/genetics
2.
J Neuroinflammation ; 19(1): 10, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991629

ABSTRACT

BACKGROUND: Astrocytes are the most numerous glial cell type with important roles in maintaining homeostasis and responding to diseases in the brain. Astrocyte function is subject to modulation by microRNAs (miRs), which are short nucleotide strands that regulate protein expression in a post-transcriptional manner. Understanding the miR expression profile of astrocytes in disease settings provides insight into the cellular stresses present in the microenvironment and may uncover pathways of therapeutic interest. METHODS: Laser-capture microdissection was used to isolate human astrocytes surrounding stroke lesions and those from neurological control tissue. Astrocytic miR expression profiles were examined using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Primary human fetal astrocytes were cultured under in vitro stress conditions and transfection of a miR mimic was used to better understand how altered levels of miR-210 affect astrocyte function. The astrocytic response to stress was studied using qPCR, enzyme-linked immunosorbent assays (ELISAs), measurement of released lactate, and Seahorse. RESULTS: Here, we measured miR expression levels in astrocytes around human ischemic stroke lesions and observed differential expression of miR-210 in chronic stroke astrocytes compared to astrocytes from neurological control tissue. We also identified increased expression of miR-210 in mouse white matter tissue around middle cerebral artery occlusion (MCAO) brain lesions. We aimed to understand the role of miR-210 in primary human fetal astrocytes by developing an in vitro assay of hypoxic, metabolic, and inflammatory stresses. A combination of hypoxic and inflammatory stresses was observed to upregulate miR-210 expression. Transfection with miR-210-mimic (210M) increased glycolysis, enhanced lactate export, and promoted an anti-inflammatory transcriptional and translational signature in astrocytes. Additionally, 210M transfection resulted in decreased expression of complement 3 (C3) and semaphorin 5b (Sema5b). CONCLUSIONS: We conclude that miR-210 expression in human astrocytes is modulated in response to ischemic stroke disease and under in vitro stress conditions, supporting a role for miR-210 in the astrocytic response to disease conditions. Further, the anti-inflammatory and pro-glycolytic impact of miR-210 on astrocytes makes it a potential candidate for further research as a neuroprotective agent.


Subject(s)
Astrocytes/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Stroke/metabolism , Animals , HeLa Cells , Humans , Inflammation/genetics , Laser Capture Microdissection , Mice , MicroRNAs/genetics , Stroke/genetics
3.
Am J Pathol ; 188(12): 2902-2911, 2018 12.
Article in English | MEDLINE | ID: mdl-30248342

ABSTRACT

Patient-derived xenografts retain the genotype of the parent tumors more readily than tumor cells maintained in culture. The two previously reported clival chordoma xenografts were derived from recurrent tumors after radiation. To study the genetics of clival chordoma in the absence of prior radiation exposure we established a patient-derived xenograft at primary resection of a clival chordoma. Epicranial grafting of clival chordoma collected during surgery was performed. Tumor growth was established in a nonobese diabetic/severe combined immunodeficiency mouse and tumors have been passaged serially for seven generations. Physaliferous cell architecture was shown in the regenerated tumors, which stained positive for Brachyury, cytokeratin, and S100 protein. The tumors showed bone invasion. Single-nucleotide polymorphism analysis of the tumor xenograft was compared with the parental tumor. Copy number gain of the T gene (brachyury) and heterozygous loss of cyclin dependent kinase inhibitor 2A (CDKN2A) was observed. Heterozygous loss of the tumor-suppressor fragile histidine triad (FHIT) gene also was observed, although protein expression was preserved. Accumulation of copy number losses and gains as well as increased growth rate was observed over three generations. The patient-derived xenograft reproduces the phenotype of clival chordoma. This model can be used in the future to study chordoma biology and to assess novel treatments.


Subject(s)
Biomarkers, Tumor/genetics , Chordoma/genetics , Genomic Instability , Polymorphism, Single Nucleotide , Skull Base Neoplasms/genetics , Aged , Animals , Apoptosis , Cell Proliferation , Chordoma/pathology , Gene Expression Profiling , Genome, Human , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Skull Base Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Am J Physiol Cell Physiol ; 306(12): C1191-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24760980

ABSTRACT

We have previously shown that ischemic preconditioning (IPC) protection against necrosis in whole hearts and in both fresh and cultured cardiomyocytes, as well as the improved regulatory volume decrease to hypoosmotic swelling in cardiomyocytes, is abrogated through Cl(-) channel blockade, pointing to a role for enhanced cell volume regulation in IPC. To further define this cardioprotective mechanism, cultured rabbit ventricular cardiomyocytes were preconditioned either by 10-min simulated ischemia (SI) followed by 10-min simulated reperfusion (SR), by 10-min exposure/10-min washout of remote IPC (rIPC) plasma dialysate (from rabbits subjected to repetitive limb ischemia), or by adenoviral transfection with the constitutively active PKC-ε gene. These interventions were done before cardiomyocytes were subjected to either 60- or 75-min SI/60-min SR to assess cell necrosis (by trypan blue staining), 30-min SI to assess ischemic cell swelling, or 30-min hypoosmotic (200 mosM) stress to assess cell volume regulation. Necrosis after SI/SR and both SI- and hypoosmotic stress-induced swelling was reduced in preconditioned cardiomyocytes compared with control cardiomyocytes (neither preconditioned nor transfected). These effects on necrosis and cell swelling were blocked by either Cl(-) channel blockade or dominant negative knockdown of inwardly rectifying K(+) channels with adenoviruses, suggesting that Cl(-) and K(+) movements across the sarcolemma are critical for cell volume regulation and, thereby, cell survival under hypoxic/ischemic conditions. Our results define enhanced cell volume regulation as a key common mechanism of cardioprotection by preconditioning in cardiomyocytes.


Subject(s)
Cell Size , Ischemic Preconditioning, Myocardial , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Animals , Chloride Channels/metabolism , Ischemia , Myocardial Infarction/prevention & control , Necrosis/physiopathology , Potassium Channels/metabolism , Rabbits , Reperfusion , Sarcolemma/metabolism
5.
J Mol Cell Cardiol ; 60: 142-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23608604

ABSTRACT

Multiple initiatives are underway to harness the clinical benefits of remote ischemic preconditioning (rIPC) based on applying non-invasive, brief, intermittent limb ischemia/reperfusion using an external occluder. However, little is known about how rIPC induces protection in cardiomyocytes, particularly through G-protein coupled receptors. In these studies, we determined the role of opioid and adenosine receptors and their functional interactions in rIPC cardioprotection. In freshly isolated cardiomyocytes subjected to 45-min simulated ischemia followed by 60-min simulated reperfusion, we examined the ability of plasma dialysate (derived from blood obtained from rabbits remotely preconditioned by application of brief cycles of hind limb ischemia/reperfusion, rIPC dialysate) to protect cells against necrosis. rIPC dialysate and selective activation of either δ-opioid receptors or κ-opioid receptors significantly reduced the % of dead cells after simulated ischemia and simulated reperfusion. Inhibition of adenosine A1 receptors, but not adenosine A3 receptors, blocked the protection by rIPC dialysate, δ-opioid receptor and κ-opioid receptor activation. In HEK293 cells expressing either hemagglutinin A-tagged δ-opioid receptors or hemagglutinin A-tagged κ-opioid receptors, selective immunoprecipitation of adenosine A1 receptors pulled down both δ-opioid and κ-opioid receptors. This molecular association of adenosine A1 receptors with δ-opioid and κ-opioid receptors was confirmed by reverse pull-down assays. These findings strongly suggest that rIPC cardioprotection requires the activation of δ-opioid and κ-opioid receptors and relies on these receptors functionally interacting with adenosine A1 receptors.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocytes, Cardiac/metabolism , Receptor, Adenosine A1/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Animals , HEK293 Cells , Humans , Myocytes, Cardiac/pathology , Rabbits
6.
Nanomedicine ; 8(7): 1133-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22349099

ABSTRACT

The blood brain barrier (BBB) is a major impediment to the delivery of therapeutics into the central nervous system (CNS). Gold nanoparticles (AuNPs) have been successfully employed in multiple potential therapeutic and diagnostic applications outside the CNS. However, AuNPs have very limited biodistribution within the CNS following intravenous administration. Magnetic resonance imaging guided focused ultrasound (MRgFUS) is a novel technique that can transiently increase BBB permeability allowing delivery of therapeutics into the CNS. MRgFUS has not been previously employed for delivery of AuNPs into the CNS. This work represents the first demonstration of focal enhanced delivery of AuNPs into the CNS using MRgFUS in a rat model both safely and effectively. Histologic visualization and analytical quantification of AuNPs within the brain parenchyma suggest BBB transgression. These results suggest a role for MRgFUS in the delivery of AuNPs with therapeutic potential into the CNS for targeting neurological diseases. FROM THE CLINICAL EDITOR: Gold nanoparticles have been successfully utilized in experimental diagnostic and therapeutic applications; however, the blood-brain barrier (BBB) is not permeable to these particles. In this paper, the authors demonstrated that MRI guided focused ultrasound is capable to transiently open the BBB thereby enabling CNS access.


Subject(s)
Brain/metabolism , Drug Delivery Systems/methods , Gold/metabolism , Magnetic Resonance Imaging/methods , Nanoparticles/analysis , Sound , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/radiation effects , Brain/radiation effects , Gold/chemistry , Rats , Rats, Wistar
7.
Neurosurg Focus ; 32(1): E3, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22208896

ABSTRACT

Recent advances in molecular neurooncology provide unique opportunities for targeted molecular-based therapies. However, the blood-brain barrier (BBB) remains a major limitation to the delivery of tumor-specific therapies directed against aberrant signaling pathways in brain tumors. Given the dismal prognosis of patients with malignant brain tumors, novel strategies that overcome the intrinsic limitations of the BBB are therefore highly desirable. Focused ultrasound BBB disruption is emerging as a novel strategy for enhanced delivery of therapeutic agents into the brain via focal, reversible, and safe BBB disruption. This review examines the potential role and implications of focused ultrasound in molecular neurooncology.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Brain Neoplasms/therapy , Drug Delivery Systems , Ultrasonics/methods , Animals , Biological Transport , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Magnetic Resonance Imaging , Rats , Ultrasonography
8.
Transl Oncol ; 14(6): 101051, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33761369

ABSTRACT

Actin is a key structural protein that makes up the cytoskeleton of cells, and plays a role in functions such as division, migration, and vesicle trafficking. It comprises six different cell-type specific isoforms: ACTA1, ACTA2, ACTB, ACTC1, ACTG1, and ACTG2. Abnormal actin isoform expression has been reported in many cancers, which led us to hypothesize that it may serve as an early biomarker of cancer. We show an overview of the different actin isoforms and highlight mechanisms by which they may contribute to tumorigenicity. Furthermore, we suggest how the aberrant expression of actin subunits can confer cells with greater proliferation ability, increased migratory capability, and chemoresistance through incorporation into the normal cellular F-actin network and altered actin binding protein interaction. Studying this fundamental change that takes place within cancer cells can further our understanding of neoplastic transformation in multiple tissue types, which can ultimately aid in the early-detection, diagnosis and treatment of cancer.

9.
Biochem Pharmacol ; 194: 114801, 2021 12.
Article in English | MEDLINE | ID: mdl-34678222

ABSTRACT

The regulation of the apoptotic pathway is one of the most studied mechanisms regarding cancer cell resistance. Many mutations have been linked to drug resistance. The DNA fragmentation factor 40 (DFF40) has been gaining interest regarding cancer cell response to chemotherapy and patient outcomes. Glioblastomas and uterine leiomyosarcomas have been shown to have a downregulation of DFF40 expression, conferring a poor patient prognosis. In concordance with these observations, in this study, we showed that DFF40 gene is also downregulated in breast, endocervical, ovarian, lung, pancreas and glioblastomas. DFF40 is the endonuclease responsible of DNA fragmentation during apoptosis. In this study, we sought to determine if a DFF40 deficiency in Jurkat T cells could impact the sensitivity to conventional chemotherapy drugs. CRISPR-cas9 generated DFF40 knockout (DFF40 KO) stable Jurkat cells and wild-type (DFF40 WT) cells were treated with different antimetabolites and topoisomerase II (TOP2) inhibitors, and cell viability was subsequently assessed. DFF40 deficient cells show chemoresistance to antimetabolites (e.g. methotrexate, 6-mercaptopurine and cytarabine) and surprisingly, they are more sensitive to TOP2 inhibitors (e.g. etoposide and teniposide). DFF40 deficient cells exposed to cytarabine present lower phosphatidylserine translocation levels to the outer cell membrane layer. Etoposide exposure in DFF40 deficient cells induces higher mortality levels and downregulation of Bcl-xL cells compared to DFF40 expressing T cells. The abolition of DFF40 expression in Jurkat cells significantly impairs histone H2AX phosphorylation following etoposide and cytarabine treatments. Our findings suggest that DFF40 is a novel key target in cancer cell resistance that potentially regulates genomic stability.


Subject(s)
Apoptosis/physiology , Deoxyribonucleases/deficiency , Drug Resistance, Neoplasm/physiology , Neoplasms/metabolism , Poly-ADP-Ribose Binding Proteins/deficiency , Signal Transduction/physiology , T-Lymphocytes/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Deoxyribonucleases/genetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Gene Knockout Techniques , HeLa Cells , Humans , Jurkat Cells , Neoplasms/drug therapy , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Signal Transduction/drug effects , T-Lymphocytes/drug effects
10.
Neuro Oncol ; 23(9): 1470-1480, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33433612

ABSTRACT

BACKGROUND: Sixty percent of surgically resected brain metastases (BrM) recur within 1 year. These recurrences have long been thought to result from the dispersion of cancer cells during surgery. We tested the alternative hypothesis that invasion of cancer cells into the adjacent brain plays a significant role in local recurrence and shortened overall survival. METHODS: We determined the invasion pattern of 164 surgically resected BrM and correlated with local recurrence and overall survival. We performed single-cell RNA sequencing (scRNAseq) of >15,000 cells from BrM and adjacent brain tissue. Validation of targets was performed with a novel cohort of BrM patient-derived xenografts (PDX) and patient tissues. RESULTS: We demonstrate that invasion of metastatic cancer cells into the adjacent brain is associated with local recurrence and shortened overall survival. scRNAseq of paired tumor and adjacent brain samples confirmed the existence of invasive cancer cells in the tumor-adjacent brain. Analysis of these cells identified cold-inducible RNA-binding protein (CIRBP) overexpression in invasive cancer cells compared to cancer cells located within the metastases. Applying PDX models that recapitulate the invasion pattern observed in patients, we show that CIRBP is overexpressed in highly invasive BrM and is required for efficient invasive growth in the brain. CONCLUSIONS: These data demonstrate peritumoral invasion as a driver of treatment failure in BrM that is functionally mediated by CIRBP. These findings improve our understanding of the biology underlying postoperative treatment failure and lay the groundwork for rational clinical trial development based upon invasion pattern in surgically resected BrM.


Subject(s)
Brain Neoplasms , Radiosurgery , Brain , Brain Neoplasms/genetics , Brain Neoplasms/surgery , Humans , Neoplasm Recurrence, Local/genetics , RNA-Binding Proteins/genetics
11.
Clin Sci (Lond) ; 117(5): 191-200, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19175358

ABSTRACT

rIPC (remote ischaemic preconditioning) is a phenomenon whereby short periods of ischaemia and reperfusion of a tissue or organ (e.g. mesentery, kidney) can protect a distant tissue or organ (e.g. heart) against subsequent, potentially lethal, ischaemia. We, and others, have shown that transient limb ischaemia can provide potent myocardial protection experimentally and clinically during cardiac surgery. Nonetheless, our understanding of the signal transduction from remote stimulus to local effect remains incomplete. The aim of the present study was to define the humoral nature of rIPC effector(s) from limb ischaemia and to study their local effects in isolated heart and cardiomyocyte models. Using a Langendorff preparation, we show that infarct size after coronary artery ligation and reperfusion was substantially reduced by rIPC in vivo, this stimulus up-regulating the MAPKs (mitogen-activating protein kinases) p42/p44, and inducing PKCepsilon (protein kinase Cepsilon) subcellular redistribution. Pre-treatment with the plasma and dialysate of plasma (obtained using 15 kDa cut-off dialysis membrane) from donor rabbits subjected to rIPC similarly protected against infarction. The effectiveness of the rIPC dialysate was abrogated by passage through a C18 hydrophobic column, but eluate from this column provided the same level of protection. The dialysate of rIPC plasma from rabbits and humans was also tested in an isolated fresh cardiomyocyte model of simulated ischaemia and reperfusion. Necrosis in cardiomyocytes treated with rIPC dialysate was substantially reduced compared with control, and was similar to cells pre-treated by 'classical' preconditioning. This effect, by rabbit rIPC dialysate, was blocked by pre-treatment with the opiate receptor blocker naloxone. In conclusion, in vivo transient limb ischaemia releases a low-molecular-mass (<15 kDa) hydrophobic circulating factor(s) which induce(s) a potent protection against myocardial ischaemia/reperfusion injury in Langendorff-perfused hearts and isolated cardiomyocytes in the same species. This cardioprotection is transferable across species, independent of local neurogenic activity, and requires opioid receptor activation.


Subject(s)
Blood , Hindlimb/blood supply , Ischemia , Ischemic Preconditioning, Myocardial/methods , Myocardial Reperfusion Injury/prevention & control , Animals , Dialysis Solutions/pharmacology , Humans , Ligation , Muscle, Skeletal/blood supply , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion , Myocardium , Myocytes, Cardiac/pathology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Rabbits , Random Allocation , Receptors, Opioid/drug effects
12.
World Neurosurg ; 117: e588-e594, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29933086

ABSTRACT

BACKGROUND: Laser interstitial thermal therapy (LITT) enables ablation of lesions using thermal energy with minimal damage to surrounding regions. Bevacizumab has been used as an adjuvant therapy in recurrent glioblastoma (GBM). At present, bevacizumab is administered at least 4-6 weeks after surgical intervention; however, given the minimally invasive nature of LITT, we suggest that bevacizumab can be safely administered at a shorter interval after LITT. METHODS: Three patients who showed evidence of recurrent GBM on magnetic resonance imaging (MRI), underwent LITT, and were subsequently administered bevacizumab within 4 weeks were identified retrospectively. Postoperative MRI was performed 24 hours after treatment and before follow-up. All 3 patients were placed on dexamethasone taper postoperatively. The ablated tumors were analyzed radiographically, and bevacizumab symptoms were monitored carefully. RESULTS: The patients ranged in age from 39 to 69 years. The median duration of follow-up was 26 weeks (range, 13-51 weeks). All 3 patients expired due to disease progression. The median progression-free survival (PFS) was 17 weeks (range, 12-22 weeks), and the median overall survival (OS) was 32 weeks (range, 12-51 weeks). There were no postoperative complications or complications due to hemorrhage, infection, or thrombosis (complication rate, 0%; 95% confidence interval 0-56%). CONCLUSIONS: This case series suggests that bevacizumab administration is safe within 1 month after LITT, thus showing promise in treating recurrent GBM. Larger studies are warranted to assess the efficacy of combined bevacizumab and LITT.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Laser Therapy , Adult , Aged , Antineoplastic Agents, Immunological/adverse effects , Bevacizumab/adverse effects , Brain Neoplasms/diagnostic imaging , Combined Modality Therapy , Fatal Outcome , Female , Glioblastoma/diagnostic imaging , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/therapy
13.
Cureus ; 10(6): e2894, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30175000

ABSTRACT

Laser thermal ablation is a novel minimally invasive neurosurgical technique that has proven to be beneficial in the treatment of a select group of neurosurgical conditions such as primary brain neoplasms, brain metastases, radiation necrosis, and epileptogenic lesions such as cortical dysplasia and mesial temporal sclerosis. The applicability of laser thermal ablation and its utility in the treatment of extra-axial (EA) brain neoplasms, mainly meningioma, is another novel use of this technique. Our article discusses the use and benefits of this technique in this particular clinical scenario. We describe our experience in a group of symptomatic patients from our institution with EA masses, mainly recurrent meningiomas, that failed previous more conventional treatment therapies such as surgery and radiotherapy. Our paper emphasizes patient selection, indications for the procedure, and post-treatment imaging characteristics of the ablated lesions.

14.
Int J Med Robot ; 14(4): e1914, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29708640

ABSTRACT

INTRODUCTION: Since the introduction of wearable head-up displays, there has been much interest in the surgical community adapting this technology into routine surgical practice. METHODS: We used the keywords augmented reality OR wearable device OR head-up display AND surgery using PubMed, EBSCO, IEEE and SCOPUS databases. After exclusions, 74 published articles that evaluated the utility of wearable head-up displays in surgical settings were included in our review. RESULTS: Across all studies, the most common use of head-up displays was in cases of live streaming from surgical microscopes, navigation, monitoring of vital signs, and display of preoperative images. The most commonly used head-up display was Google Glass. Head-up displays enhanced surgeons' operating experience; common disadvantages include limited battery life, display size and discomfort. CONCLUSIONS: Due to ergonomic issues with dual-screen devices, augmented reality devices with the capacity to overlay images onto the surgical field will be key features of next-generation surgical head-up displays.


Subject(s)
Surgery, Computer-Assisted/methods , Wearable Electronic Devices , Computer Simulation , Equipment Design , Humans , Surgeons , Surgery, Computer-Assisted/instrumentation , Surgery, Computer-Assisted/trends , Wearable Electronic Devices/trends
15.
Cardiovasc Res ; 70(2): 286-96, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16413515

ABSTRACT

Isolated cardiomyocytes, obtained by enzymatic digestion of whole hearts, have multiple advantages, most related to their accessibility to microscopic visualization, beyond the obvious elimination of other cell types that exist in the heart. Conversely, they cannot reproduce the mechanical disruption of reperfusion hypercontracture or the vascular phenomena of leukocyte plugging and compression from interstitial edema and contracture that can lead to the no-reflow phenomenon. Nevertheless, ischemic preconditioning has been consistently demonstrated to be a potent protective mechanism in freshly isolated and cultured cardiomyocytes across multiple species, indicating that much of the innate protection of ischemic preconditioning resides in cardiomyocytes. Centrifuging freshly isolated cardiomyocytes into a pellet with only a thin layer of supernatant covered by oil has proven to be an excellent model of simulated ischemia. In culture, cardiomyocytes may be exposed to severe hypoxia only or to various protocols for simulated ischemia in which an acid/lactate-rich, hyperkalemic extracellular environment with substrate deprivation (lacking glucose) is typically added. Reperfusion is simulated by well-oxygenated media of normal ionic composition. Cardiomyocyte injury has been usually evaluated by cell membrane permeability to dyes, often under hypo-osmotic conditions (osmotic fragility) or enzyme release. A survey of the use of cardiomyocyte models to study preconditioning is presented with the emphasis on examples of the innovative measurements, increasingly involving molecular techniques, that point to an increasing future role for these models in preconditioning research and, more generally, in the mechanistic study of myocardial ischemia/reperfusion.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Adult , Animals , Cells, Cultured , Heart/embryology , Humans , Mitochondria, Heart/pathology , Research Design
16.
Int J Med Robot ; 13(4)2017 Dec.
Article in English | MEDLINE | ID: mdl-28524449

ABSTRACT

BACKGROUND: Wearable technology is growing in popularity as a result of its ability to interface with normal human movement and function. METHODS: Using proprietary hardware and software, neuronavigation images were captured and transferred wirelessly via a password-encrypted network to the head-up display. The operating surgeon wore a loupe-mounted wearable head-up display during image-guided parieto-occipital ventriculoperitoneal shunt placement in two patients. RESULTS: The shunt placement was completed successfully without complications. The tip of the catheter ended well within the ventricles away from the ventricular wall. The wearable device allowed for continuous monitoring of neuronavigation images in the right upper corner of the surgeon's visual field without the need for the surgeon to turn his head to view the monitors. CONCLUSIONS: The adaptable nature of this proposed system permits the display of video data to the operating surgeon without diverting attention away from the operative task. This technology has the potential to enhance image-guided procedures.


Subject(s)
Catheterization/methods , Neuronavigation/instrumentation , Wearable Electronic Devices , Brain/diagnostic imaging , Catheters , Cohort Studies , Computers , Equipment Design , Eyeglasses , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Internet , Movement , Neuronavigation/methods , Phantoms, Imaging , Prospective Studies , Software , Tomography, X-Ray Computed , User-Computer Interface , Video Recording
17.
Circ Res ; 95(3): 325-32, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15231687

ABSTRACT

Volume regulatory Cl- channels are key regulators of ischemic preconditioning (IPC). Because Cl- efflux must be balanced by an efflux of cations to maintain cell membrane electroneutrality during volume regulation, we hypothesize that I(K1) channels may play a role in IPC. We subjected cultured cardiomyocytes to 60-minute simulated ischemia (SI) followed by 60-minute of simulated reperfusion (SR) and assessed percent cell death using trypan blue staining. Ischemic preconditioning (10-minute SI/10-minute SR) significantly (P<0.0001) reduced the percent cell death in nontransfected cardiomyocytes [IPC(CM) 18.0+/-2.1% versus control (C(CM)) 48.3+/-1.0%]. IPC protection was not altered by overexpression of the reporter gene (enhanced green fluorescent protein, EGFP). However, overexpression of dominant-negative Kir2.1 or Kir2.2 genes using adenoviruses (AdEGFPKir2.1DN or AdEGFPKir2.2DN) encoding the reporter gene EGFP prevented IPC protection [both IPC(CM)+AdEGFPKir2.1DN 45.8+/-2.3% (mean+/-SEM) and IPC(CM)+AdEGFPKir2.2DN 47.9+/-1.4% versus IPC(CM); P<0.0001] in cultured cardiomyocytes (n=8 hearts). Transfection of cardiomyocytes with AdEGFPKir2.1DN or AdEGFPKir2.2DN did not affect cell death in control (nonpreconditioned) cardiomyocytes (both C(CM)+ AdEGFPKir2.1DN 45.8+/-0.7% and C(CM)+AdEGFPKir2.2DN 46.2+/-1.3% versus C(CM); not statistically significant). Similar effects were observed in both cultured (n=5 hearts) and freshly isolated (n=4 hearts) ventricular cardiomyocytes after I(K1) blockade with 20 micromol/L BaCl2 plus 1 micromol/L nifedipine (to prevent Ba2+ uptake). Nifedipine alone neither protected against ischemic injury nor blocked IPC protection. Our findings establish that I(K1) channels play an important role in IPC protection.


Subject(s)
Ischemic Preconditioning , Myocytes, Cardiac/drug effects , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Adenoviridae/genetics , Animals , Barium/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Cell Survival , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chlorides/metabolism , Genetic Vectors/pharmacology , Ion Channel Gating/drug effects , Ion Transport/drug effects , Myocardial Reperfusion , Myocytes, Cardiac/metabolism , Nifedipine/pharmacology , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Rabbits , Recombinant Fusion Proteins/physiology , Sarcolemma/drug effects , Sarcolemma/metabolism , Transfection
18.
World Neurosurg ; 94: 418-425, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27402436

ABSTRACT

OBJECTIVE: In recent years laser interstitial thermal therapy (LITT) has become the ablative neurosurgical procedure of choice. Multiple methods for registration and laser fiber verification have been described, with each method requiring multiple steps and significant time expenditure. We evaluated the use of a commercially available mobile computed tomography (CT) scanner for stereotactic registration during LITT for brain tumors in an attempt to simplify the procedure and improve intraoperative awareness of laser position. METHODS: This is a retrospective chart review comparing LITT of brain tumors in 23 patients undergoing a standard protocol requiring skull pins and transport of the patient to a CT suite to obtain a reference scan compared with 14 patients in whom the Medtronic O-arm was used intraoperatively for navigation registration and confirmation of laser position. RESULTS: Total ablation of the target was achieved in all patients with no surgical complications. Total surgery time was shorter for the O-arm group than for the standard protocol group, once experience was gained with bringing the O-arm in and out of the surgical field. Return from the magnetic resonance imaging suite to the operating room for repositioning of the laser was required for 1 patient in the standard protocol group, but for no patients in the O-arm group. Once experience was gained with using the O-arm, estimated surgical costs were lower for this group. CONCLUSIONS: Use of a mobile intraoperative CT scanner for navigation registration and confirmation of laser position during LITT may play a role in streamlining the procedure and improving patient safety and comfort.


Subject(s)
Brain Neoplasms/therapy , Hyperthermia, Induced/instrumentation , Laser Therapy/instrumentation , Neuronavigation/instrumentation , Surgery, Computer-Assisted/instrumentation , Tomography, X-Ray Computed/instrumentation , Brain Neoplasms/diagnostic imaging , Equipment Design , Equipment Failure Analysis , Female , Humans , Male , Middle Aged , Retrospective Studies , Subtraction Technique/instrumentation , Treatment Outcome
19.
Cardiovasc Res ; 55(3): 660-71, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12160963

ABSTRACT

OBJECTIVES: We have recently proposed that chloride (Cl(-)) channels contribute to ischemic preconditioning (IPC) in the myocardium. To further evaluate this hypothesis, we investigated the role of Cl(-) channels in pharmacological preconditioning. METHODS: Isolated rabbit cardiomyocytes and isolated buffer-perfused rabbit hearts were initially preconditioned with a 10 min exposure to either an adenosine receptor agonist [2-chloro-N(6)-cyclopentyladenosine (CCPA, 200 nM) and/or N(6)-2-(4-aminophenyl)ethyladenosine (APNEA, 1 microM)] or the PKC activator phorbol 12-myristate 13-acetate (PMA, 1 microM) followed by a 10 or 20 min washout or not preconditioned (control). Cardiomyocytes or whole hearts were then subjected to prolonged ischemic period (45 min simulated ischemia or 40 min of regional myocardial ischemia, respectively) followed by 60 min reperfusion (resuspension in oxygenated medium or release of the transient coronary occlusion, respectively). RESULTS: Indanyloxyacetic acid 94, a selective Cl(-) channel inhibitor that produced substantial inhibition of the regulatory volume decrease (RVD) when given at 10 microM concentration in cultured cardiomyocytes, was administered before ischemia to block RVD through Cl(-) channel inhibition. CCPA, APNEA and PMA significantly (P<0.01) reduced the % of dead cardiomyocytes (by trypan blue staining) after 45 min SI/60 min SR, as compared to controls, while IAA-94 abolished this protection but did not affect PKCepsilon translocation by IPC. We confirmed that IAA-94 blocked IPC-, APNEA- and PMA-induced protection against infarction in the isolated heart model. CONCLUSIONS: These findings support our contention that Cl(-) channels are downstream effectors of IPC.


Subject(s)
Adenosine/analogs & derivatives , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Receptors, Purinergic P1/metabolism , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Cells, Cultured , Chloride Channels/drug effects , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , Glycolates/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Perfusion , Protein Kinase C/metabolism , Rabbits , Tetradecanoylphorbol Acetate/therapeutic use
20.
Oncotarget ; 6(25): 21718-29, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26061748

ABSTRACT

Advances in the molecular biology of medulloblastoma revealed four genetically and clinically distinct subgroups. Group 3 medulloblastomas are characterized by frequent amplifications of the oncogene MYC, a high incidence of metastasis, and poor prognosis despite aggressive therapy. We investigated several potential small molecule inhibitors to target Group 3 medulloblastomas based on gene expression data using an in silico drug screen. The Connectivity Map (C-MAP) analysis identified piperlongumine as the top candidate drug for non-WNT medulloblastomas and the cyclin-dependent kinase (CDK) inhibitor alsterpaullone as the compound predicted to have specific antitumor activity against Group 3 medulloblastomas. To validate our findings we used these inhibitors against established Group 3 medulloblastoma cell lines. The C-MAP predicted drugs reduced cell proliferation in vitro and increased survival in Group 3 medulloblastoma xenografts. Alsterpaullone had the highest efficacy in Group 3 medulloblastoma cells. Genomic profiling of Group 3 medulloblastoma cells treated with alsterpaullone confirmed inhibition of cell cycle-related genes, and down-regulation of MYC. Our results demonstrate the preclinical efficacy of using a targeted therapy approach for Group 3 medulloblastomas. Specifically, we provide rationale for advancing alsterpaullone as a targeted therapy in Group 3 medulloblastoma.


Subject(s)
Antineoplastic Agents/chemistry , Benzazepines/chemistry , Drug Screening Assays, Antitumor , Indoles/chemistry , Medulloblastoma/drug therapy , Acetophenones/chemistry , Animals , Benzopyrans/chemistry , Brain Neoplasms/drug therapy , Cell Line , Cell Proliferation , Cyclin-Dependent Kinases/antagonists & inhibitors , Dioxolanes/chemistry , Flunarizine/chemistry , Gene Expression Profiling , Genomics , Humans , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL