Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Exp Neurol ; 361: 114320, 2023 03.
Article in English | MEDLINE | ID: mdl-36627040

ABSTRACT

Endocannabinoids [2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine (AEA)], endogenously produced arachidonate-based lipids, are anti-inflammatory physiological ligands for two known cannabinoid receptors, CB1 and CB2, yet the molecular and cellular mechanisms underlying their effects after brain injury are poorly defined. In the present study, we hypothesize that traumatic brain injury (TBI)-induced loss of endocannabinoids exaggerates neurovascular injury, compromises brain-cerebrospinal fluid (CSF) barriers (BCB) and causes behavioral dysfunction. Preliminary analysis in human CSF and plasma indicates changes in endocannabinoid levels. This encouraged us to investigate the levels of endocannabinoid-metabolizing enzymes in a mouse model of controlled cortical impact (CCI). Reductions in endocannabinoid (2-AG and AEA) levels in plasma were supported by higher expression of their respective metabolizing enzymes, monoacylglycerol lipase (MAGL), fatty acid amide hydrolase (FAAH), and cyclooxygenase 2 (Cox-2) in the post-TBI mouse brain. Following increased metabolism of endocannabinoids post-TBI, we observed increased expression of CB2, non-cannabinoid receptor Transient receptor potential vanilloid-1 (TRPV1), aquaporin 4 (AQP4), ionized calcium binding adaptor molecule 1 (IBA1), glial fibrillary acidic protein (GFAP), and acute reduction in cerebral blood flow (CBF). The BCB and pericontusional cortex showed altered endocannabinoid expressions and reduction in ventricular volume. Finally, loss of motor functions and induced anxiety behaviors were observed in these TBI mice. Taken together, our findings suggest endocannabinoids and their metabolizing enzymes play an important role in the brain and BCB integrity and highlight the need for more extensive studies on these mechanisms.


Subject(s)
Antineoplastic Agents , Brain Injuries, Traumatic , Brain Injuries , Mice , Humans , Animals , Endocannabinoids/metabolism , Brain/metabolism , Brain Injuries, Traumatic/complications , Receptor, Cannabinoid, CB1/metabolism
2.
Front Neurosci ; 16: 791035, 2022.
Article in English | MEDLINE | ID: mdl-35645722

ABSTRACT

Spontaneous Intracerebral hemorrhage (ICH) is a devastating injury that accounts for 10-15% of all strokes. The rupture of cerebral blood vessels damaged by hypertension or cerebral amyloid angiopathy creates a space-occupying hematoma that contributes toward neurological deterioration and high patient morbidity and mortality. Numerous protocols have explored a role for surgical decompression of ICH via craniotomy, stereotactic guided endoscopy, and minimally invasive catheter/tube evacuation. Studies including, but not limited to, STICH, STICH-II, MISTIE, MISTIE-II, MISTIE-III, ENRICH, and ICES have all shown that, in certain limited patient populations, evacuation can be done safely and mortality can be decreased, but functional outcomes remain statistically no different compared to medical management alone. Only 10-15% of patients with ICH are surgical candidates based on clot location, medical comorbidities, and limitations regarding early surgical intervention. To date, no clearly effective treatment options are available to improve ICH outcomes, leaving medical and supportive management as the standard of care. We recently identified that remote ischemic conditioning (RIC), the non-invasive, repetitive inflation-deflation of a blood pressure cuff on a limb, non-invasively enhanced hematoma resolution and improved neurological outcomes via anti-inflammatory macrophage polarization in pre-clinical ICH models. Herein, we propose a pilot, placebo-controlled, open-label, randomized trial to test the hypothesis that RIC accelerates hematoma resorption and improves outcomes in ICH patients. Twenty ICH patients will be randomized to receive either mock conditioning or unilateral arm RIC (4 cycles × 5 min inflation/5 min deflation per cycle) beginning within 48 h of stroke onset and continuing twice daily for one week. All patients will receive standard medical care according to latest guidelines. The primary outcome will be the safety evaluation of unilateral RIC in ICH patients. Secondary outcomes will include hematoma volume/clot resorption rate and functional outcomes, as assessed by the modified Rankin Scale (mRS) at 1- and 3-months post-ICH. Additionally, blood will be collected for exploratory genomic analysis. This study will establish the feasibility and safety of RIC in acute ICH patients, providing a foundation for a larger, multi-center clinical trial.

3.
World Neurosurg ; 158: e1017-e1021, 2022 02.
Article in English | MEDLINE | ID: mdl-34906752

ABSTRACT

OBJECTIVE: Decompressive craniectomy (DC) is an established optional treatment for malignant hemispheric infarction (MHI). We analyzed relevant clinical factors and computed tomography (CT) measurements in patients with DC for MHI to identify predictors of functional outcome 3-6 months after stroke. METHODS: This study was performed at 2 comprehensive stroke centers. The inclusion criteria required DC for MHI, no additional intraoperative procedures (strokectomy or cerebral ventricular drain placement), and documented functional status 3-6 months after the stroke. We classified functional outcome as acceptable if the modified Rankin Scale score was <5, or as unacceptable if it was 5 or 6 (bedbound and totally dependent on others or death). Multiple logistic regression analyzed relevant clinical factors and multiple perioperative CT measurements to identify predictors of acceptable functional outcome. RESULTS: Of 87 identified consecutive patients, 66 met the inclusion criteria. Acceptable functional outcome occurred in 35 of 66 (53%) patients. Likelihood of acceptable functional outcome decreased significantly with increasing age (OR 0.92, 95% CI 0.82-0.97, P = 0.004) and with increasing post-DC midline brain shift (OR 0.78, 95% CI 0.64-0.96, P = 0.016), and decreased non-significantly with left-sided stroke (OR 0.30, 95% CI 0.08-1.10, P = 0.069) and with increasing craniectomy barrier thickness (OR 0.92, 95% CI 0.85-1.01, P = 0.076). CONCLUSIONS: Patient age and the post-DC midline shift may be useful in prognosticating functional outcome after DC for MHI. Stroke side and craniectomy barrier thickness merit further ideally prospective outcome prediction testing.


Subject(s)
Decompressive Craniectomy , Stroke , Cerebral Infarction/diagnostic imaging , Cerebral Infarction/surgery , Decompressive Craniectomy/methods , Humans , Prospective Studies , Stroke/surgery , Tomography, X-Ray Computed , Treatment Outcome
4.
Neurochem Int ; 150: 105192, 2021 11.
Article in English | MEDLINE | ID: mdl-34560175

ABSTRACT

Mitochondria are dynamic organelles responsible for cellular energy production. Besides, regulating energy homeostasis, mitochondria are responsible for calcium homeostasis, signal transmission, and the fate of cellular survival in case of injury and pathologies. Accumulating reports have suggested multiple roles of mitochondria in neuropathologies, neurodegeneration, and immune activation under physiological and pathological conditions. Mitochondrial dysfunction, which occurs at the initial phase of brain injury, involves oxidative stress, inflammation, deficits in mitochondrial bioenergetics, biogenesis, transport, and autophagy. Thus, development of targeted therapeutics to protect mitochondria may improve functional outcomes following traumatic brain injury (TBI) and intracerebral hemorrhages (ICH). In this review, we summarize mitochondrial dysfunction related to TBI and ICH, including the mechanisms involved, and discuss therapeutic approaches with special emphasis on past and current clinical trials.


Subject(s)
Brain Injuries, Traumatic/metabolism , Cerebral Hemorrhage/metabolism , Mitochondria/metabolism , Mitophagy/physiology , Animals , Autophagy/drug effects , Autophagy/physiology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Homeostasis/drug effects , Homeostasis/physiology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Mitochondria/drug effects , Mitochondria/pathology , Mitophagy/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Oxidative Stress/physiology
5.
J Stroke Cerebrovasc Dis ; 30(7): 105830, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33945955

ABSTRACT

OBJECTIVE: Decompressive craniectomy (DC) improves functional outcomes in selected patients with malignant hemispheric infarction (MHI), but variability in the surgical technique and occasional complications may be limiting the effectiveness of this procedure. Our aim was to evaluate predefined perioperative CT measurements for association with post-DC midline brain shift in patients with MHI. METHODS: At two medical centers we identified 87 consecutive patients with MHI and DC between January 2007 and December 2019. We used our previously tested methods to measure the craniectomy surface area, extent of transcalvarial brain herniation, thickness of tissues overlying the craniectomy, diameter of the cerebral ventricle atrium contralateral to the stroke, extension of infarction beyond the craniectomy edges, and the pre and post-DC midline brain shifts. To avoid potential confounding from medical treatments and additional surgical procedures, we excluded patients with the first CT delayed >30 hours post-DC, resection of infarcted brain, or insertion of an external ventricular drain during DC. The primary outcome in multiple linear regression analysis was the postoperative midline brain shift. RESULTS: We analyzed 72 qualified patients. The average midline brain shift decreased from 8.7 mm pre-DC to 5.4 post-DC. The only factors significantly associated with post-DC midline brain shift at the p<0.01 level were preoperative midline shift (coefficient 0.32, standard error 0.10, p=0.002) and extent of transcalvarial brain herniation (coefficient -0.20, standard error 0.05, p <0.001). CONCLUSIONS: In patients with MHI and DC, smaller post-DC midline shift is associated with smaller pre-DC midline brain shift and greater transcalvarial brain herniation. This knowledge may prove helpful in assessing DC candidacy and surgical success. Additional studies to enhance the surgical success of DC are warranted.


Subject(s)
Brain Edema/surgery , Cerebral Infarction/surgery , Decompressive Craniectomy , Hernia/prevention & control , Adult , Brain Edema/diagnostic imaging , Brain Edema/physiopathology , Cerebral Infarction/diagnostic imaging , Cerebral Infarction/physiopathology , Clinical Decision-Making , Decompressive Craniectomy/adverse effects , Female , Georgia , Hernia/diagnostic imaging , Hernia/etiology , Humans , Male , Middle Aged , Recovery of Function , Registries , Retrospective Studies , Risk Assessment , Risk Factors , Tomography, X-Ray Computed , Treatment Outcome , Virginia
6.
JCI Insight ; 6(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33427206

ABSTRACT

The CNS is regarded as an immunoprivileged organ, evading routine immune surveillance; however, the coordinated development of immune responses profoundly influences outcomes after brain injury. Innate lymphoid cells (ILCs) are cytokine-producing cells that are critical for the initiation, modulation, and resolution of inflammation, but the functional relevance and mechanistic regulation of ILCs are unexplored after acute brain injury. We demonstrate increased proliferation of all ILC subtypes within the meninges for up to 1 year after experimental traumatic brain injury (TBI) while ILCs were present within resected dura and elevated within cerebrospinal fluid (CSF) of moderate-to-severe TBI patients. In line with energetic derangements after TBI, inhibition of the metabolic regulator, AMPK, increased meningeal ILC expansion, whereas AMPK activation suppressed proinflammatory ILC1/ILC3 and increased the frequency of IL-10-expressing ILC2 after TBI. Moreover, intracisternal administration of IL-33 activated AMPK, expanded ILC2, and suppressed ILC1 and ILC3 within the meninges of WT and Rag1-/- mice, but not Rag1-/- IL2rg-/- mice. Taken together, we identify AMPK as a brake on the expansion of proinflammatory, CNS-resident ILCs after brain injury. These findings establish a mechanistic framework whereby immunometabolic modulation of ILCs may direct the specificity, timing, and magnitude of cerebral immunity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain Injuries, Traumatic/enzymology , Brain Injuries, Traumatic/immunology , Immunity, Innate , Lymphocytes/immunology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Adolescent , Adult , Aged , Animals , Brain Injuries, Traumatic/cerebrospinal fluid , Disease Models, Animal , Female , Humans , Lymphocytes/classification , Lymphocytes/pathology , Male , Meninges/immunology , Meninges/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Young Adult
7.
Biomedicines ; 8(10)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003373

ABSTRACT

Studying the complex molecular mechanisms involved in traumatic brain injury (TBI) is crucial for developing new therapies for TBI. Current treatments for TBI are primarily focused on patient stabilization and symptom mitigation. However, the field lacks defined therapies to prevent cell death, oxidative stress, and inflammatory cascades which lead to chronic pathology. Little can be done to treat the mechanical damage that occurs during the primary insult of a TBI; however, secondary injury mechanisms, such as inflammation, blood-brain barrier (BBB) breakdown, edema formation, excitotoxicity, oxidative stress, and cell death, can be targeted by therapeutic interventions. Elucidating the many mechanisms underlying secondary injury and studying targets of neuroprotective therapeutic agents is critical for developing new treatments. Therefore, we present a review on the molecular events following TBI from inflammation to programmed cell death and discuss current research and the latest therapeutic strategies to help understand TBI-mediated secondary injury.

8.
World Neurosurg ; 144: e693-e700, 2020 12.
Article in English | MEDLINE | ID: mdl-32942058

ABSTRACT

BACKGROUND: Pseudomeningocele is an uncommon but widely recognized complication of spinal surgery that can be challenging to correct. When conservative measures fail, patients frequently require reoperation to attempt primary closure of the durotomy, yet attempts at true watertight closures of the dura or fascia sometimes fall short. We describe a technique of lumbosacral pseudomeningocele repair involving a 2-layer pants-over-vest closure of the pseudomeningocele coupled with mobilization of bilateral paraspinal musculature to create a Z-plasty, or a Z-shaped flap. We have demonstrated a high success rate with our small series. METHODS: The technique used meticulous manipulation of the pseudomeningocele to make a 2-layer pants-over-vest closure. This closure coupled with wide mobilization and importation of paraspinous muscle into the wound effectively obliterated dead space with simultaneous tamponade of the dural tear. The lateral row perforators were left intact, providing excellent vascularity with adequate mobility to the patient. RESULTS: This technique was incorporated into the care of 10 patients between 2004 and July 2019. All wounds were closed in a single stage after careful flap section based on the wound's needs. We demonstrated successful pseudomeningocele resolution in all 10 patients with no observed clinical recurrence of symptomatic pseudomeningocele after at least 6 months of follow-up. CONCLUSIONS: This technique provides a straightforward option for the spine surgeon to manage these challenging spinal wounds with minimal, if any, need for further laminectomy as well as a high fistula control rate with minimal morbidity.


Subject(s)
Meningocele/surgery , Neurosurgical Procedures/methods , Paraspinal Muscles/surgery , Postoperative Complications/surgery , Adolescent , Adult , Aged, 80 and over , Child , Female , Humans , Lumbosacral Region/pathology , Lumbosacral Region/surgery , Male , Meningocele/etiology , Middle Aged , Reoperation/methods , Surgical Flaps , Treatment Outcome , Young Adult
9.
Sci Adv ; 6(22): eaax8847, 2020 05.
Article in English | MEDLINE | ID: mdl-32523980

ABSTRACT

Traumatic brain injury (TBI) is a major cause of mortality and morbidity. Preventative measures reduce injury incidence and/or severity, yet one-third of hospitalized patients with TBI die from secondary pathological processes that develop during supervised care. Neutrophils, which orchestrate innate immune responses, worsen TBI outcomes via undefined mechanisms. We hypothesized that formation of neutrophil extracellular traps (NETs), a purported mechanism of microbial trapping, exacerbates acute neurological injury after TBI. NET formation coincided with cerebral hypoperfusion and tissue hypoxia after experimental TBI, while elevated circulating NETs correlated with reduced serum deoxyribonuclease-1 (DNase-I) activity in patients with TBI. Functionally, Toll-like receptor 4 (TLR4) and the downstream kinase peptidylarginine deiminase 4 (PAD4) mediated NET formation and cerebrovascular dysfunction after TBI. Last, recombinant human DNase-I degraded NETs and improved neurological function. Thus, therapeutically targeting NETs may provide a mechanistically innovative approach to improve TBI outcomes without the associated risks of global neutrophil depletion.


Subject(s)
Brain Injuries, Traumatic , Extracellular Traps , Brain Injuries, Traumatic/complications , Deoxyribonuclease I/metabolism , Extracellular Traps/metabolism , Humans , Immunity, Innate , Neutrophils/metabolism
10.
World Neurosurg ; 139: 314-317, 2020 07.
Article in English | MEDLINE | ID: mdl-32344136

ABSTRACT

BACKGROUND: Glossopharyngeal neuralgia/neuropathy is rare, and less than 3% of cases involve cardiac arrhythmias of syncope due to activated vagal reflex pathways. Most of these cases are successfully treated with medical management with or without pacemaker placement. We present the first reported case of glossopharyngeal neuralgia/neuropathy with cardiac symptoms refractory to medical management including pacemaker placement but successfully treated with Gamma Knife Radiosurgery. CASE DESCRIPTION: A 70-year-old Caucasian man with recurrent squamous cell carcinoma of the tongue base treated with multiple surgeries, lymph node excision, chemotherapy, and external beam radiation, developed episodes of severe right ear and throat pain. These episodes would be followed by syncopal episodes associated with hypotension and bradycardia. Aggressive medical management including pacemaker could not stabilize the patient's hemodynamic instability. After extensive workup, the patient was diagnosed with glossopharyngeal neuralgia/neuropathy with associated carotid sinus instability. The patient was not a strong surgical candidate, thus the patient underwent Gamma Knife Radiosurgery. The target was set as the glossopharyngeal meatus. Within days of treatment, the patient had no further clinically significant syncope or hemodynamic instability for the remaining 6 months of his life. CONCLUSIONS: To our knowledge, we present the first case of glossopharyngeal neuralgia/neuropathy with medically refractory cardiac dysfunction successfully treated with Gamma Knife Radiosurgery. We advocate that Gamma Knife be considered for similar subsets of patients.


Subject(s)
Glossopharyngeal Nerve Diseases/therapy , Radiosurgery/methods , Syncope/therapy , Aged , Combined Modality Therapy/adverse effects , Glossopharyngeal Nerve Diseases/etiology , Hemodynamics , Humans , Male , Squamous Cell Carcinoma of Head and Neck/therapy , Syncope/etiology , Tongue Neoplasms/therapy
11.
World Neurosurg ; 122: e713-e722, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30394359

ABSTRACT

BACKGROUND: The management of brain arteriovenous malformations (AVMs) remains a controversial topic. Given the relatively low incidence, high heterogeneity, and high morbidity and mortality of these lesions, consensus on treatment strategies is an issue of concern to organized neurosurgery. The present retrospective analysis examined and quantified the outcomes of patients with an initial presentation of intracranial hemorrhage from a Spetzler-Martin grade III or IV AVM, later ruled out as surgical candidates. METHODS: A total of 16 patients (5 females; 11 males) had presented with symptomatic hemorrhage confirmed by non-contrast-enhanced computed tomography and were deemed to not be surgical candidates owing to AVM location and/or architecture. The patients underwent combined endovascular embolization and gamma knife stereotactic radiosurgery (SRS). The modified Rankin scale was used to measure the clinical outcomes, comparing the scores at presentation, gamma knife treatment, and the last known follow-up examination. A radiographic evaluation was used to determine the level of AVM nidus involution after the procedure. RESULTS: The present study identified 16 patients with ruptured high-grade AVMs of high surgical risk. All the patients had undergone immediate embolization with delayed SRS for treatment of the hemorrhage and nidus of the AVM. A statistically significant proportion of patients showed marked improvement in the modified Rankin scale scores. No subsequent repeat hemorrhage or any associated complications after embolization occurred in any patient. CONCLUSION: These findings warrant consideration of endovascular embolization with adjuvant SRS as a powerful treatment option for cases with high surgical morbidity due to AVM characteristics.


Subject(s)
Arteriovenous Fistula/therapy , Cerebral Hemorrhage/therapy , Embolization, Therapeutic/methods , Intracranial Arteriovenous Malformations/therapy , Radiosurgery/methods , Adolescent , Adult , Aged , Arteriovenous Fistula/diagnostic imaging , Cerebral Hemorrhage/diagnostic imaging , Child , Cohort Studies , Female , Follow-Up Studies , Humans , Intracranial Arteriovenous Malformations/diagnostic imaging , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
12.
World Neurosurg ; 122: 593-598, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30465962

ABSTRACT

BACKGROUND: Extracranial metastasis, mainly a feature of World Health Organization (WHO) grade III meningiomas, is only rarely reported in grade II meningiomas. CASE DESCRIPTION: We report a case of a 48-year-old man who was initially diagnosed in 2010 with an occipital convexity meningioma based on computed tomography scan/magnetic resonance imaging (MRI) and treated with surgical therapy and gamma knife. The first operation achieved a macroscopically complete resection. The tumor was histologically classified as an atypical meningioma. The patient had a recurrence in 2014 on the left tentorial leaflet as noted on postcontrast MRI. The patient was asymptomatic, without focal neurologic deficits. In 2016, the patient reported new-onset pain in the neck and left upper extremity. MRI indicated complete replacement of the C7 vertebral marrow, with a soft tissue component extending posteriorly into the epidural space that appeared to be flattening the thecal sac but without evidence of abnormal cord signal. Histopathology of resection confirmed atypical meningioma. CONCLUSIONS: This case represents a rare instance of intraosseous spine as the first site of metastasis of WHO grade II atypical meningioma and is the first reported case of extracranial metastasis of a meningioma to the C7 vertebral body.


Subject(s)
Brain Neoplasms/diagnostic imaging , Cervical Vertebrae/diagnostic imaging , Meningeal Neoplasms/diagnostic imaging , Meningioma/diagnostic imaging , Spinal Neoplasms/diagnostic imaging , Spinal Neoplasms/secondary , Brain Neoplasms/surgery , Cervical Vertebrae/surgery , Humans , Male , Meningeal Neoplasms/surgery , Meningioma/surgery , Middle Aged , Spinal Neoplasms/surgery
13.
J Exp Med ; 215(10): 2636-2654, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30190288

ABSTRACT

Spontaneous intracerebral hemorrhage (ICH) produces the highest acute mortality and worst outcomes of all stroke subtypes. Hematoma volume is an independent determinant of ICH patient outcomes, making clot resolution a primary goal of clinical management. Herein, remote-limb ischemic post-conditioning (RIC), the repetitive inflation-deflation of a blood pressure cuff on a limb, accelerated hematoma resolution and improved neurological outcomes after ICH in mice. Parabiosis studies revealed RIC accelerated clot resolution via a humoral-mediated mechanism. Whereas RIC increased anti-inflammatory macrophage activation, myeloid cell depletion eliminated the beneficial effects of RIC after ICH. Myeloid-specific inactivation of the metabolic regulator, AMPKα1, attenuated RIC-induced anti-inflammatory macrophage polarization and delayed hematoma resolution, providing a molecular link between RIC and immune activation. Finally, chimera studies implicated myeloid CD36 expression in RIC-mediated neurological recovery after ICH. Thus, RIC, a clinically well-tolerated therapy, noninvasively modulates innate immune responses to improve ICH outcomes. Moreover, immunometabolic changes may provide pharmacodynamic blood biomarkers to clinically monitor the therapeutic efficacy of RIC.


Subject(s)
AMP-Activated Protein Kinases/immunology , Hematoma/immunology , Ischemic Postconditioning , Macrophage Activation , Macrophages/immunology , Stroke/immunology , AMP-Activated Protein Kinases/genetics , Animals , Hematoma/pathology , Hematoma/therapy , Macrophages/pathology , Mice , Mice, Knockout , Stroke/pathology , Stroke/therapy
14.
South Med J ; 110(9): 586-593, 2017 09.
Article in English | MEDLINE | ID: mdl-28863223

ABSTRACT

Cancer metastasis is a key event in tumor progression associated not only with mortality but also significant morbidity. Metastatic disease can promote end-organ dysfunction and even failure through mass effect compression of various vital organs including the spinal cord. In such cases, prompt medical attention is needed to restore neurological function, relieve pain, and prevent permanent damage. The three therapeutic approaches to managing metastatic spinal cord compression include corticosteroids, surgery, and radiation therapy. Although each may improve patients' symptoms, their combination has yielded the best outcome. In cancer patients with clinical suspicion of spinal cord compression, dexamethasone should be initiated followed by surgical decompression, when possible, and radiation. The latter becomes the preferred treatment in patients with inoperable disease.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Decompression, Surgical , Dexamethasone/therapeutic use , Radiotherapy , Spinal Cord Compression/therapy , Spinal Neoplasms/secondary , Combined Modality Therapy , Humans , Radiotherapy/adverse effects , Radiotherapy/methods , Spinal Cord Compression/etiology , Spinal Neoplasms/complications
16.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2614-2626, 2017 10.
Article in English | MEDLINE | ID: mdl-28533056

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of mortality and long-term morbidity worldwide. Despite decades of pre-clinical investigation, therapeutic strategies focused on acute neuroprotection failed to improve TBI outcomes. This lack of translational success has necessitated a reassessment of the optimal targets for intervention, including a heightened focus on secondary injury mechanisms. Chronic immune activation correlates with progressive neurodegeneration for decades after TBI; however, significant challenges remain in functionally and mechanistically defining immune activation after TBI. In this review, we explore the burgeoning evidence implicating the acute release of damage associated molecular patterns (DAMPs), such as adenosine 5'-triphosphate (ATP), high mobility group box protein 1 (HMGB1), S100 proteins, and hyaluronic acid in the initiation of progressive neurological injury, including white matter loss after TBI. The role that pattern recognition receptors, including toll-like receptor and purinergic receptors, play in progressive neurological injury after TBI is detailed. Finally, we provide support for the notion that resident and infiltrating macrophages are critical cellular targets linking acute DAMP release with adaptive immune responses and chronic injury after TBI. The therapeutic potential of targeting DAMPs and barriers to clinical translational, in the context of TBI patient management, are discussed.


Subject(s)
Brain Injuries, Traumatic/metabolism , White Matter/metabolism , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Hyaluronic Acid/immunology , Hyaluronic Acid/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , S100 Proteins/immunology , S100 Proteins/metabolism , White Matter/immunology , White Matter/pathology
17.
J Immunol ; 198(9): 3615-3626, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28341672

ABSTRACT

Traumatic brain injury (TBI) is a major public health issue, producing significant patient mortality and poor long-term outcomes. Increasing evidence suggests an important, yet poorly defined, role for the immune system in the development of secondary neurologic injury over the days and weeks following a TBI. In this study, we tested the hypothesis that peripheral macrophage infiltration initiates long-lasting adaptive immune responses after TBI. Using a murine controlled cortical impact model, we used adoptive transfer, transgenic, and bone marrow chimera approaches to show increased infiltration and proinflammatory (classically activated [M1]) polarization of macrophages for up to 3 wk post-TBI. Monocytes purified from the injured brain stimulated the proliferation of naive T lymphocytes, enhanced the polarization of T effector cells (TH1/TH17), and decreased the production of regulatory T cells in an MLR. Similarly, elevated T effector cell polarization within blood and brain tissue was attenuated by myeloid cell depletion after TBI. Functionally, C3H/HeJ (TLR4 mutant) mice reversed M1 macrophage and TH1/TH17 polarization after TBI compared with C3H/OuJ (wild-type) mice. Moreover, brain monocytes isolated from C3H/HeJ mice were less potent stimulators of T lymphocyte proliferation and TH1/TH17 polarization compared with C3H/OuJ monocytes. Taken together, our data implicate TLR4-dependent, M1 macrophage trafficking/polarization into the CNS as a key mechanistic link between acute TBI and long-term, adaptive immune responses.


Subject(s)
Brain Injuries, Traumatic/immunology , Macrophages/physiology , Th1 Cells/immunology , Th17 Cells/immunology , Toll-Like Receptor 4/genetics , Adaptive Immunity , Adoptive Transfer , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Phenotype
18.
Radiother Oncol ; 117(3): 483-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26277434

ABSTRACT

This study presents a MLC-based, 3D grid-therapy technique with characteristics of both 3D-conformal-radiotherapy and grid-therapy. It generates a brachytherapy-like dose distribution, with D50% of 20, 9.8, 5.4 and 2.9-Gy, for the spheres, target, 1 cm-outershell and 2 cm-outershell, respectively. It may provide a strategy to deliver ablative doses to large tumors safely.


Subject(s)
Dose Fractionation, Radiation , Radiotherapy, Conformal/methods , Humans , Radiotherapy Planning, Computer-Assisted/methods
19.
BMC Cancer ; 15: 118, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25851054

ABSTRACT

BACKGROUND: Malignant glioma is one of the most devastating tumors in adults with poor patient prognosis. Notably, glioma often exhibits resistance to conventional chemotherapeutic approaches, complicating patient treatments. However, the molecular mediators involved in tumor chemoresistance remain poorly defined, creating a barrier to the successful management of glioma. In the present study, we hypothesized that the antioxidant transcription factor, Nrf2 (nuclear factor erythroid-derived 2 like 2), attenuates glioma cytotoxicity to Carmustine (BCNU), a widely used chemotherapeutic agent known to modulate cellular oxidative balance. METHODS: To test the hypothesis, we employed human malignant glioma cell line, U87MG and overexpression of Nrf2 in glioma cells was achieved using both pharmacological and genetic approaches. RESULTS: Notably, induction of Nrf2 was associated with increased expression of heme oxygenase-1 (HO-1), a stress inducible enzyme involved in anti-oxidant defense. In addition, over expression of Nrf2 in U87MG cells significantly attenuated the cytotoxicity of Carmustine as evidenced by both cellular viability assay and flow cytometry analysis. Consistent with this, antioxidants such as glutathione and N-acetyl cysteine significantly reduced Carmustine mediated glioma cytotoxicity. CONCLUSIONS: Taken together, these data strongly implicate an unexplored role of Nrf2 in glioma resistance to Carmustine and raise the possible use of Nrf2 inhibitors as adjunct to Carmustine for the treatment of malignant glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression , Glioma/genetics , NF-E2-Related Factor 2/genetics , Antineoplastic Agents, Alkylating/toxicity , Antioxidants/pharmacology , Carmustine/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Glioma/metabolism , Humans , Hydroquinones/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Up-Regulation/drug effects
20.
Glia ; 62(1): 26-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166800

ABSTRACT

Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Cerebral edema, a life-threatening medical complication, contributes to elevated intracranial pressure (ICP) and a poor clinical prognosis after TBI. Unfortunately, treatment options to reduce post-traumatic edema remain suboptimal, due in part, to a dearth of viable therapeutic targets. Herein, we tested the hypothesis that cerebral innate immune responses contribute to edema development after TBI. Our results demonstrate that high-mobility group box protein 1 (HMGB1) was released from necrotic neurons via a NR2B-mediated mechanism. HMGB1 was clinically associated with elevated ICP in patients and functionally promoted cerebral edema after TBI in mice. The detrimental effects of HMGB1 were mediated, at least in part, via activation of microglial toll-like receptor 4 (TLR4) and the subsequent expression of the astrocytic water channel, aquaporin-4 (AQP4). Genetic or pharmacological (VGX-1027) TLR4 inhibition attenuated the neuroinflammatory response and limited post-traumatic edema with a delayed, clinically implementable therapeutic window. Human and rodent tissue culture studies further defined the cellular mechanisms demonstrating neuronal HMGB1 initiates the microglial release of interleukin-6 (IL-6) in a TLR4 dependent mechanism. In turn, microglial IL-6 increased the astrocytic expression of AQP4. Taken together, these data implicate microglia as key mediators of post-traumatic brain edema and suggest HMGB1-TLR4 signaling promotes neurovascular dysfunction after TBI.


Subject(s)
Brain Edema/etiology , Brain Injuries/complications , HMGB1 Protein/metabolism , Microglia/metabolism , Neurons/metabolism , Toll-Like Receptor 4/metabolism , Acetates/pharmacology , Animals , Brain Edema/pathology , Brain Injuries/cerebrospinal fluid , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Humans , Immunologic Factors/pharmacology , Male , Mice , Mice, Inbred C3H , Microglia/drug effects , Neurons/drug effects , Oxazoles/pharmacology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...