Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
2.
Sci Rep ; 13(1): 19264, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37935811

RESUMEN

Raising reactive oxygen species (ROS) levels in cancer cells to cause macromolecular damage and cell death is a promising anticancer treatment strategy. Observations that electromagnetic fields (EMF) elevate intracellular ROS and cause cancer cell death, have led us to develop a new portable wearable EMF device that generates spinning oscillating magnetic fields (sOMF) to selectively kill cancer cells while sparing normal cells in vitro and to shrink GBM tumors in vivo through a novel mechanism. Here, we characterized the precise configurations and timings of sOMF stimulation that produce cytotoxicity due to a critical rise in superoxide in two types of human glioma cells. We also found that the antioxidant Trolox reverses the cytotoxic effect of sOMF on glioma cells indicating that ROS play a causal role in producing the effect. Our findings clarify the link between the physics of magnetic stimulation and its mechanism of anticancer action, facilitating the development of a potential new safe noninvasive device-based treatment for GBM and other gliomas.


Asunto(s)
Glioma , Estrés Oxidativo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Glioma/terapia , Glioma/patología , Superóxidos , Campos Electromagnéticos
3.
J Neurosurg Case Lessons ; 6(16)2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37870750

RESUMEN

BACKGROUND: Erdheim-Chester disease (ECD) is a rare non-Langerhans cell histiocytosis characterized histologically by foamy histiocytes and Touton giant cells in a background of fibrosis. Bone pain with long bone osteosclerosis is highly specific for ECD. Central nervous system involvement is rare, although dural, hypothalamic, cerebellar, brainstem, and sellar region involvement has been described. OBSERVATIONS: A 59-year-old man with a history of ureteral obstruction, medically managed petit mal seizures, and a left temporal lesion followed with serial magnetic resonance imaging (MRI) presented with worsening seizure control. Repeat MRI identified bilateral amygdala region lesions. Gradual growth of the left temporal lesion over 1 year with increasing seizure frequency prompted resection. A non-Langerhans cell histiocytosis with a BRAF V600E mutation was identified on pathology. Imaging findings demonstrated retroperitoneal fibrosis and long bone osteosclerosis with increased fluorodeoxyglucose uptake that, together with the neuropathologic findings, were diagnostic of ECD. LESSONS: This case of biopsy-proven ECD is unique in that the singular symptom was seizures well controlled with medical management in the presence of similarly located bilateral anterior mesial temporal lobe lesions. Although ECD is rare intracranially, its variable imaging presentation, including the potential to mimic seizure-associated medial temporal lobe tumors, emphasizes the need for a wide differential diagnosis.

4.
Int J Mol Sci ; 24(4)2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36834607

RESUMEN

Based on the postulate that glioblastoma (GBM) tumors generate anti-inflammatory prostaglandins and bile salts to gain immune privilege, we analyzed 712 tumors in-silico from three GBM transcriptome databases for prostaglandin and bile synthesis/signaling enzyme-transcript markers. A pan-database correlation analysis was performed to identify cell-specific signal generation and downstream effects. The tumors were stratified by their ability to generate prostaglandins, their competency in bile salt synthesis, and the presence of bile acid receptors nuclear receptor subfamily 1, group H, member 4 (NR1H4) and G protein-coupled bile acid receptor 1 (GPBAR1). The survival analysis indicates that tumors capable of prostaglandin and/or bile salt synthesis are linked to poor outcomes. Tumor prostaglandin D2 and F2 syntheses are derived from infiltrating microglia, whereas prostaglandin E2 synthesis is derived from neutrophils. GBMs drive the microglial synthesis of PGD2/F2 by releasing/activating complement system component C3a. GBM expression of sperm-associated heat-shock proteins appears to stimulate neutrophilic PGE2 synthesis. The tumors that generate bile and express high levels of bile receptor NR1H4 have a fetal liver phenotype and a RORC-Treg infiltration signature. The bile-generating tumors that express high levels of GPBAR1 are infiltrated with immunosuppressive microglia/macrophage/myeloid-derived suppressor cells. These findings provide insight into how GBMs generate immune privilege and may explain the failure of checkpoint inhibitor therapy and provide novel targets for treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Masculino , Humanos , Prostaglandinas , Glioblastoma/metabolismo , Ácidos y Sales Biliares , Privilegio Inmunológico , Semen/metabolismo , Dinoprostona , Prostaglandinas Sintéticas , Neoplasias Encefálicas/metabolismo , Receptores Acoplados a Proteínas G/genética
6.
Electromagn Biol Med ; 41(4): 419-428, 2022 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-36154345

RESUMEN

We recently reported shrinkage of untreatable recurrent glioblastoma (GBM) in an end-stage patient using noninvasive brain stimulation with a spinning oscillating magnetic field (sOMF)-generating device called the Oncomagnetic device. Our in vitro experiments demonstrated selective cancer cell death while sparing normal cells by sOMF-induced increase in intracellular reactive oxygen species (ROS) levels due to magnetic perturbation of mitochondrial electron transport. Here, we describe the results of an in vivo study assessing the toxicity of chronic sOMF stimulation in mice using a newly constructed apparatus comprised of the sOMF-generating active components of the Oncomagnetic device. We chronically stimulated 10 normal 60-day old female C57BL/6 mice in their housing cages for 2 h 3 times a day, as in the patient treatment protocol, over 4 months. We also studied the effects of 2-h acute sOMF stimulation. Our observations and those of blinded independent veterinary staff observers, indicated no significant adverse effects of chronic or acute sOMF stimulation on the health, behavior, electrocardiographic and electroencephalographic activities, hematologic profile, and brain and other tissue and organ morphology of treated mice compared to age-matched untreated control mice. These findings suggest that short- and long-term therapies with the Oncomagnetic device are safe and well tolerated.


Asunto(s)
Glioblastoma , Campos Magnéticos , Animales , Ratones , Femenino , Especies Reactivas de Oxígeno , Ratones Endogámicos C57BL , Encéfalo
7.
Surg Neurol Int ; 13: 348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36128155

RESUMEN

Background: Chordomas are rare neuraxial tumors arising from remnants of primitive notochord. They are generally slow-growing malignant neoplasms. Only four adult cases of multicentric chordomas have been reported, all with aggressive and rapid growth. Here, we present an unusual case of indolent multicentric chordomas involving cervical and thoracic spine, sacrum, and calvarium. Case Description: A 60-year-old male was found to have multiple lesions throughout his neuroaxis incidentally on workup for colitis. A needle biopsy documented the diagnosis of chordoma. This has been followed for more than 4 years with no progression. Conclusion: We present the first reported case of indolent multicentric chordomas. Due to the extreme rarity of indolent multicentric chordomas, close follow-up is needed and recommended.

8.
Nat Commun ; 13(1): 767, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35140215

RESUMEN

A major rate-limiting step in developing more effective immunotherapies for GBM is our inadequate understanding of the cellular complexity and the molecular heterogeneity of immune infiltrates in gliomas. Here, we report an integrated analysis of 201,986 human glioma, immune, and other stromal cells at the single cell level. In doing so, we discover extensive spatial and molecular heterogeneity in immune infiltrates. We identify molecular signatures for nine distinct myeloid cell subtypes, of which five are independent prognostic indicators of glioma patient survival. Furthermore, we identify S100A4 as a regulator of immune suppressive T and myeloid cells in GBM and demonstrate that deleting S100a4 in non-cancer cells is sufficient to reprogram the immune landscape and significantly improve survival. This study provides insights into spatial, molecular, and functional heterogeneity of glioma and glioma-associated immune cells and demonstrates the utility of this dataset for discovering therapeutic targets for this poorly immunogenic cancer.


Asunto(s)
Inmunoterapia , Proteína de Unión al Calcio S100A4/aislamiento & purificación , Análisis de la Célula Individual/métodos , Animales , Neoplasias Encefálicas/inmunología , Femenino , Glioma/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides , Pronóstico , Proteína de Unión al Calcio S100A4/genética , Microambiente Tumoral/inmunología
9.
Neuro Oncol ; 24(4): 556-568, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34515312

RESUMEN

BACKGROUND: We postulate that meningiomas undergo distinct metabolic reprogramming in tumorigenesis and unraveling their metabolic phenotypes provide new therapeutic insights. Glutamine catabolism is key to the growth and proliferation of tumors. Here, we investigated the metabolomics of freshly resected meningiomas and glutamine metabolism in patient-derived meningioma cells. METHODS: 1H NMR spectroscopy of tumor tissues from meningioma patients was used to differentiate the metabolite profiles of grade-I and grade-II meningiomas. Glutamine metabolism was examined using 13C/15N glutamine tracer, in 5 patient-derived meningioma cells. RESULTS: Alanine, lactate, glutamate, glutamine, and glycine were predominantly elevated only in grade-II meningiomas by 74%, 76%, 35%, 75%, and 33%, respectively, with alanine and glutamine levels being statistically significant (P ≤ .02). 13C/15N glutamine tracer experiments revealed that both grade-I and -II meningiomas actively metabolize glutamine to generate various key carbon intermediates including alanine and proline that are necessary for the tumor growth. Also, it is shown that glutaminase (GLS1) inhibitor, CB-839 is highly effective in downregulating glutamine metabolism and decreasing proliferation in meningioma cells. CONCLUSION: Alanine and glutamine/glutamate are mainly elevated in grade-II meningiomas. Grade-I meningiomas possess relatively higher glutamine metabolism providing carbon/nitrogen for the biosynthesis of key nonessential amino acids. GLS1 inhibitor (CB-839) is very effective in downregulating glutamine metabolic pathways in grade-I meningiomas leading to decreased cellular proliferation.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Aminoácidos , Niño , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo
10.
Cancer Res Commun ; 2(6): 402-416, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36688010

RESUMEN

The emergence of treatment resistance significantly reduces the clinical utility of many effective targeted therapies. Although both genetic and epigenetic mechanisms of drug resistance have been reported, whether these mechanisms are stochastically selected in individual tumors or governed by a predictable underlying principle is unknown. Here, we report that the dependence of cancer stem cells (CSCs), not bulk tumor cells, on the targeted pathway determines the molecular mechanism of resistance in individual tumors. Using both spontaneous and transplantable mouse models of sonic hedgehog (SHH) medulloblastoma (MB) treated with an SHH/Smoothened inhibitor, sonidegib/LDE225, we show that genetic-based resistance occurs only in tumors that contain SHH-dependent CSCs (SD-CSCs). In contrast, SHH MBs containing SHH-dependent bulk tumor cells but SHH-independent CSCs (SI-CSCs) acquire resistance through epigenetic reprogramming. Mechanistically, elevated proteasome activity in SMOi-resistant SI-CSC MBs alters the tumor cell maturation trajectory through enhanced degradation of specific epigenetic regulators, including histone acetylation machinery components, resulting in global reductions in H3K9Ac, H3K14Ac, H3K56Ac, H4K5Ac, and H4K8Ac marks and gene expression changes. These results provide new insights into how selective pressure on distinct tumor cell populations contributes to different mechanisms of resistance to targeted therapies. This insight provides a new conceptual framework to understand responses and resistance to SMOis and other targeted therapies.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Animales , Ratones , Transducción de Señal , Proteínas Hedgehog/genética , Meduloblastoma/genética , Neoplasias Cerebelosas/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo
11.
Front Oncol ; 11: 768758, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858847

RESUMEN

Electromagnetic fields (EMF) raise intracellular levels of reactive oxygen species (ROS) that can be toxic to cancer cells. Because weak magnetic fields influence spin state pairing in redox-active radical electron pairs, we hypothesize that they disrupt electron flow in the mitochondrial electron transport chain (ETC). We tested this hypothesis by studying the effects of oscillating magnetic fields (sOMF) produced by a new noninvasive device involving permanent magnets spinning with specific frequency and timing patterns. We studied the effects of sOMF on ETC by measuring the consumption of oxygen (O2) by isolated rat liver mitochondria, normal human astrocytes, and several patient derived brain tumor cells, and O2 generation/consumption by plant cells with an O2 electrode. We also investigated glucose metabolism in tumor cells using 1H and 13C nuclear magnetic resonance and assessed mitochondrial alterations leading to cell death by using fluorescence microscopy with MitoTracker™ and a fluorescent probe for Caspase 3 activation. We show that sOMF of appropriate field strength, frequency, and on/off profiles completely arrest electron transport in isolated, respiring, rat liver mitochondria and patient derived glioblastoma (GBM), meningioma and diffuse intrinsic pontine glioma (DIPG) cells and can induce loss of mitochondrial integrity. These changes correlate with a decrease in mitochondrial carbon flux in cancer cells and with cancer cell death even in the non-dividing phase of the cell cycle. Our findings suggest that rotating magnetic fields could be therapeutically efficacious in brain cancers such as GBM and DIPG through selective disruption of the electron flow in immobile ETC complexes.

12.
Int J Mol Sci ; 22(20)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34681642

RESUMEN

Regulatory T-cells (Tregs) are immunosuppressive T-cells, which arrest immune responses to 'Self' tissues. Some immunosuppressive Tregs that recognize seminal epitopes suppress immune responses to the proteins in semen, in both men and women. We postulated that GBMs express reproductive-associated proteins to manipulate reproductive Tregs and to gain immune privilege. We analyzed four GBM transcriptome databases representing ≈900 tumors for hypoxia-responsive Tregs, steroidogenic pathways, and sperm/testicular and placenta-specific genes, stratifying tumors by expression. In silico analysis suggested that the presence of reproductive-associated Tregs in GBM tumors was associated with worse patient outcomes. These tumors have an androgenic signature, express male-specific antigens, and attract reproductive-associated Related Orphan Receptor C (RORC)-Treg immunosuppressive cells. GBM patient sera were interrogated for the presence of anti-sperm/testicular antibodies, along with age-matched controls, utilizing monkey testicle sections. GBM patient serum contained anti-sperm/testicular antibodies at levels > six-fold that of controls. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are associated with estrogenic tumors which appear to mimic placental tissue. We demonstrate that RORC-Tregs drive poor patient outcome, and Treg infiltration correlates strongly with androgen levels. Androgens support GBM expression of sperm/testicular proteins allowing Tregs from the patient's reproductive system to infiltrate the tumor. In contrast, estrogen appears responsible for MDSC/TAM immunosuppression.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Linfocitos Infiltrantes de Tumor/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Andrógenos/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Bases de Datos Factuales , Estrógenos/metabolismo , Femenino , Glioblastoma/inmunología , Glioblastoma/mortalidad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Microglía/inmunología , Microglía/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología
13.
J Cancer Res Clin Oncol ; 147(12): 3577-3589, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34477946

RESUMEN

PURPOSE: The mechanisms underlying anticancer effects of electromagnetic fields are poorly understood. An alternating electric field-generating therapeutic device called Optune™ device has been approved for the treatment of glioblastoma (GBM). We have developed a new device that generates oscillating magnetic fields (OMF) by rapid rotation of strong permanent magnets in specially designed patterns of frequency and timing and have used it to treat an end-stage recurrent GBM patient under an expanded access/compassionate use treatment protocol. Here, we ask whether OMF causes selective cytotoxic effects in GBM and whether it is through generation of reactive oxygen species (ROS). METHODS: We stimulated patient derived GBM cells, lung cancer cells, normal human cortical neurons, astrocytes, and bronchial epithelial cells using OMF generators (oncoscillators) of our Oncomagnetic Device and compared the results to those obtained under unstimulated or sham-stimulated control conditions. Quantitative fluorescence microscopy was used to assess cell morphology, viability, and ROS production mechanisms. RESULTS: We find that OMF induces highly selective cell death of patient derived GBM cells associated with activation of caspase 3, while leaving normal tissue cells undamaged. The cytotoxic effect of OMF is also seen in pulmonary cancer cells. The underlying mechanism is a marked increase in ROS in the mitochondria, possibly in part through perturbation of the electron flow in the respiratory chain. CONCLUSION: Rotating magnetic fields produced by a new noninvasive device selectively kill cultured human glioblastoma and non-small cell lung cancer cells by raising intracellular reactive oxygen species, but not normal human tissue cells.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Magnetoterapia/métodos , Muerte Celular , Humanos , Células Tumorales Cultivadas
14.
Front Oncol ; 11: 708017, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367992

RESUMEN

Alternating electric field therapy has been approved for glioblastoma (GBM). We have preclinical evidence for anticancer effects in GBM cell cultures and mouse xenografts with an oscillating magnetic field (OMF) generating device. Here we report OMF treatment of end-stage recurrent glioblastoma in a 53-year-old man who had undergone radical surgical excision and chemoradiotherapy, and experimental gene therapy for a left frontal tumor. He experienced tumor recurrence and progressive enlargement with leptomeningeal involvement. OMF for 5 weeks was well tolerated, with 31% reduction of contrast-enhanced tumor volume and reduction in abnormal T2-weighted Fluid-Attenuated Inversion Recovery volume. Tumor shrinkage appeared to correlate with treatment dose. These findings suggest a powerful new noninvasive therapy for glioblastoma.

15.
Cancers (Basel) ; 13(8)2021 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-33920278

RESUMEN

BACKGROUND: Glioblastoma (GBM) can use metabolic fuels other than glucose (Glc). The ability of GBM to use galactose (Gal) as a fuel via the Leloir pathway is investigated. METHODS: Gene transcript data were accessed to determine the association between expression of genes of the Leloir pathway and patient outcomes. Growth studies were performed on five primary patient-derived GBM cultures using Glc-free media supplemented with Gal. The role of Glut3/Glut14 in sugar import was investigated using antibody inhibition of hexose transport. A specific inhibitor of GALK1 (Cpd36) was used to inhibit Gal catabolism. Gal metabolism was examined using proton, carbon and phosphorous NMR spectroscopy, with 13C-labeled Glc and Gal as tracers. RESULTS: Data analysis from published databases revealed that elevated levels of mRNA transcripts of SLC2A3 (Glut3), SLC2A14 (Glut14) and key Leloir pathway enzymes correlate with poor patient outcomes. GBM cultures proliferated when grown solely on Gal in Glc-free media and switching Glc-grown GBM cells into Gal-enriched/Glc-free media produced elevated levels of Glut3 and/or Glut14 enzymes. The 13C NMR-based metabolic flux analysis demonstrated a fully functional Leloir pathway and elevated pentose phosphate pathway activity for efficient Gal metabolism in GBM cells. CONCLUSION: Expression of Glut3 and/or Glut14 together with the enzymes of the Leloir pathway allows GBM to transport and metabolize Gal at physiological glucose concentrations, providing GBM cells with an alternate energy source. The presence of this pathway in GBM and its selective targeting may provide new treatment strategies.

16.
Mol Cancer Ther ; 19(12): 2445-2453, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33033175

RESUMEN

We have previously reported the in vitro and in vivo efficacy of N,N-bis(2-chloroethyl)-2-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)propenamide (MP-MUS), a prodrug that targeted the mitochondria of glioblastoma (GBM). The mitochondrial enzyme, monoamine oxidase B (MAOB), is highly expressed in GBM and oxidizes an uncharged methyl-tetrahydropyridine (MP-) moiety into the mitochondrially targeted cationic form, methyl-pyridinium (P+-). Coupling this MAOB-sensitive group to a nitrogen mustard produced a prodrug that damaged GBM mitochondria and killed GBM cells. Unfortunately, the intrinsic reactivity of the nitrogen mustard group and low solubility of MP-MUS precluded clinical development. In our second-generation prodrug, MP-Pt(IV), we coupled the MP group to an unreactive cisplatin precursor. The enzymatic conversion of MP-Pt(IV) to P+-Pt(IV) was tested using recombinant human MAOA and rhMAOB. The generation of cisplatin from Pt(IV) by ascorbate was studied optically and using mass spectroscopy. Efficacy toward primary GBM cells and tumors was studied in vitro and in an intracranial patient-derived xenograft mice GBM model. Our studies demonstrate that MP-Pt(IV) is selectively activated by MAOB. MP-Pt(IV) is highly toxic toward GBM cells in vitro MP-Pt(IV) toxicity against GBM is potentiated by elevating mitochondrial ascorbate and can be arrested by MAOB inhibition. In in vitro studies, sublethal MP-Pt(IV) doses elevated mitochondrial MAOB levels in surviving GBM cells. MP-Pt(IV) is a potent chemotherapeutic in intracranial patient-derived xenograft mouse models of primary GBM and potentiates both temozolomide and temozolomide-chemoradiation therapies. MP-Pt(IV) was well tolerated and is highly effective against GBM in both in vitro and in vivo models.


Asunto(s)
Antineoplásicos/farmacología , Glioblastoma/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Profármacos , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioblastoma/tratamiento farmacológico , Humanos , Ratones , Inhibidores de la Monoaminooxidasa/uso terapéutico , Proteínas Recombinantes , Ensayos Antitumor por Modelo de Xenoinjerto
17.
World Neurosurg ; 142: 283-290, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32603865

RESUMEN

The history of neurosurgery in Texas is linked with the development over the past century of the Houston Methodist Hospital (HMH) from a 30-bed hospital in downtown Houston to an academic medical center with 900 beds in the Texas Medical Center. Neurosurgery at HMH has developed to meet the needs of the Houston Metropolitan Area, which has grown from 130,000 people in 1919 to 7 million people today. Neurosurgery at HMH has had steady growth and stable leadership with Dr. James Greenwood Jr. 1936-1980, Dr. Robert Grossman 1980-2013, and Dr. Gavin Britz 2013-present, as Chiefs of the Neurosurgical Service. HMH has been affiliated with 2 medical schools: Baylor College of Medicine 1950-2003 and Weill College of Medicine Cornell University 2004-present. Neurosurgical training began at HMH with the establishment of the Baylor College of Medicine Neurosurgery Residency Program with Dr. George Ehni as Program Director 1959-1979 and Dr. Robert Grossman as Program Director 1980-2006. Training has continued in the HMH residency program from 2006 to present with Dr. David Baskin as Program Director. As of 2019, 138 neurosurgical residents have been trained at HMH. The goals of delivering responsible patient care, advancing neurosurgical knowledge, and training the next generation of practitioners and teachers of neurosurgery have remained constant and have been met and remain the mission of the department.


Asunto(s)
Centros Médicos Académicos/historia , Neurocirujanos/historia , Neurocirugia/historia , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Procedimientos Neuroquirúrgicos/historia , Texas
18.
World Neurosurg ; 142: 171-175, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32593765

RESUMEN

BACKGROUND: Posterior reversible encephalopathy syndrome is manifested by a reversible neurologic deficit such as vision loss, encephalopathy, and a posterior location, typically the occipital lobes. It is commonly thought to be related to acute, severe hypertension. CASE DESCRIPTION: A 51-year-old woman presented with visual loss for several months, and a suprasellar mass was diagnosed. She underwent transsphenoidal surgery, which was complicated by cerebrospinal fluid leak, and she developed posterior reversible encephalopathy syndrome while undergoing postoperative cerebrospinal fluid drainage via lumbar catheter. Her visual acuity progressed to blindness, but blindness was reversed by discontinuation of lumbar drainage, tight blood pressure control, and high-dose steroid drip. CONCLUSIONS: To our knowledge, this is only the second case of posterior reversible encephalopathy syndrome following transsphenoidal surgery to be reported in the neurosurgical or ophthalmic English language literature.


Asunto(s)
Adenoma/cirugía , Neuroendoscopía/efectos adversos , Neoplasias Hipofisarias/cirugía , Síndrome de Leucoencefalopatía Posterior/etiología , Seno Esfenoidal/cirugía , Adenoma/diagnóstico por imagen , Corticoesteroides/uso terapéutico , Pérdida de Líquido Cefalorraquídeo/complicaciones , Pérdida de Líquido Cefalorraquídeo/diagnóstico por imagen , Femenino , Humanos , Persona de Mediana Edad , Neuroendoscopía/métodos , Neoplasias Hipofisarias/diagnóstico por imagen , Síndrome de Leucoencefalopatía Posterior/diagnóstico por imagen , Síndrome de Leucoencefalopatía Posterior/tratamiento farmacológico , Seno Esfenoidal/diagnóstico por imagen
19.
Cancers (Basel) ; 12(2)2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-32075134

RESUMEN

Various pathways can repair DNA alkylation by chemotherapeutic agents such as temozolomide (TMZ). The enzyme O6-methylguanine methyltransferase (MGMT) removes O6-methylated DNA adducts, leading to the failure of chemotherapy in resistant glioblastomas. Because of the anti-chemotherapeutic activities of MGMT previously described, estimating the levels of active MGMT in cancer cells can be a significant predictor of response to alkylating agents. Current methods to detect MGMT in cells are indirect, complicated, time-intensive, or utilize molecules that require complex and multistep chemistry synthesis. Our design simulates DNA repair by the transfer of a clickable propargyl group from O6-propargyl guanine to active MGMT and subsequent attachment of fluorescein-linked PEG linker via "click chemistry." Visualization of active MGMT levels reveals discrete active and inactive MGMT populations with biphasic kinetics for MGMT inactivation in response to TMZ-induced DNA damage.

20.
Cancers (Basel) ; 12(2)2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033192

RESUMEN

BACKGROUND: Rathke's Cleft Cysts (RCCs) are rare epithelial cysts arising from remnants of the Rathke pouch in the pituitary gland. A subset of these lesions enlarge and produce a mass effect with consequent hypopituitarism, and may result in visual loss. Moreover, some RCCs with a high intra-cystic protein content may mimic cystic pituitary adenoma, which makes their differential diagnosis ambiguous. Currently, medical professionals have no definitive way to distinguish RCCs from pituitary adenomas. Therefore, preoperative confirmation of RCCs would be of help to medical professionals for the management and proper surgical decision making. The goal of this study is to identify molecular markers in RCCs. METHODS: We characterized aqueous and chloroform extracts of surgically resected RCCs and pituitary adenomas using ex vivo 1H NMR spectroscopy. RESULTS: All RCCs exclusively showed the presence of mucopolysaccharides which are glycosaminoglycans (GAGs) made up of disaccharides of aminosugars and uronic sugars. CONCLUSION: GAGs can be used as metabolite marker for the detection of RCCs and this knowledge will lay the groundwork for the development of a non-invasive, in vivo magnetic resonance spectroscopy methodology for the differential diagnosis of RCCs and pituitary adenomas using clinical MRI scanners.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...