Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Curr Opin Neurobiol ; 89: 102916, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39293245

ABSTRACT

Neurodevelopmental disorders (NDDs) affect 15% of children and are usually associated with intellectual disability, seizures, and autistic behaviors, among other neurological presentations. Mutations in a wide spectrum of gene families alter key stages of human brain development, leading to defects in neural circuits or brain architecture. Studies in animal systems have provided important insights into the pathobiology of several NDDs. Human stem cell technologies provide a complementary system that allows functional manipulation of human brain cells during developmental stages that would otherwise be inaccessible during human fetal brain development. Therefore, stem cell-based models advance our understanding of human brain development by revealing human-specific mechanisms contributing to the broad pathogenesis of NDDs. We provide a comprehensive overview of the latest research on two and three-dimensional human stem cell-based models. First, we discuss convergent cellular and molecular phenotypes across different NDDs that have been revealed by human iPSC systems. Next, we examine the contribution of in vitro human neural systems to the development of promising therapeutic strategies. Finally, we explore the potential of stem cell systems to draw mechanistic insight for the study of sex dimorphism within NDDs.

4.
J Neurochem ; 2023 Nov 06.
Article in English | MEDLINE | ID: mdl-37929637

ABSTRACT

The metabolic demands of neuronal activity are both temporally and spatially dynamic, and neurons are particularly sensitive to disruptions in fuel and oxygen supply. Glucose is considered an obligate fuel for supporting brain metabolism. Although alternative fuels are often available, the extent of their contribution to central carbon metabolism remains debated. Differential fuel metabolism likely depends on cell type, location, and activity state, complicating its study. While biosensors provide excellent spatial and temporal information, they are limited to observations of only a few metabolites. On the other hand, mass spectrometry is rich in chemical information, but traditionally relies on cell culture or homogenized tissue samples. Here, we use mass spectrometry imaging (MALDI-MSI) to focus on the fuel metabolism of the dentate granule cell (DGC) layer in murine hippocampal slices. Using stable isotopes, we explore labeling dynamics at baseline, as well as in response to brief stimulation or fuel competition. We find that at rest, glucose is the predominant fuel metabolized through glycolysis, with little to no measurable contribution from glycerol or fructose. However, lactate/pyruvate, ß-hydroxybutyrate (ßHB), octanoate, and glutamine can contribute to TCA metabolism to varying degrees. In response to brief depolarization with 50 mM KCl, glucose metabolism was preferentially increased relative to the metabolism of alternative fuels. With an increased supply of alternative fuels, both lactate/pyruvate and ßHB can outcompete glucose for TCA cycle entry. While lactate/pyruvate modestly reduced glucose contribution to glycolysis, ßHB caused little change in glycolysis. This approach achieves broad metabolite coverage from a spatially defined region of physiological tissue, in which metabolic states are rapidly preserved following experimental manipulation. Using this powerful methodology, we investigated metabolism within the dentate gyrus not only at rest, but also in response to the energetic demand of activation, and in states of fuel competition.

5.
Nat Metab ; 5(10): 1820-1835, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37798473

ABSTRACT

Neuronal activity creates an intense energy demand that must be met by rapid metabolic responses. To investigate metabolic adaptations in the neuron-enriched dentate granule cell (DGC) layer within its native tissue environment, we employed murine acute hippocampal brain slices, coupled with fast metabolite preservation and followed by mass spectrometry (MS) imaging, to generate spatially resolved metabolomics and isotope-tracing data. Here we show that membrane depolarization induces broad metabolic changes, including increased glycolytic activity in DGCs. Increased glucose metabolism in response to stimulation is accompanied by mobilization of endogenous inosine into pentose phosphates via the action of purine nucleotide phosphorylase (PNP). The PNP reaction is an integral part of the neuronal response to stimulation, because inhibition of PNP leaves DGCs energetically impaired during recovery from strong activation. Performing MS imaging on brain slices bridges the gap between live-cell physiology and the deep chemical analysis enabled by MS.


Subject(s)
Dentate Gyrus , Neurons , Mice , Animals , Dentate Gyrus/physiology , Cell Membrane , Isotopes , Metabolomics
6.
Res Sq ; 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37546759

ABSTRACT

Neuronal activity creates an intense energy demand that must be met by rapid metabolic responses. To investigate metabolic adaptations in the neuron-enriched dentate granule cell (DGC) layer within its native tissue environment, we employed murine acute hippocampal brain slices coupled with fast metabolite preservation, followed by mass spectrometry imaging (MALDI-MSI) to generate spatially resolved metabolomics and isotope tracing data. Here we show that membrane depolarization induces broad metabolic changes, including increased glycolytic activity in DGCs. Increased glucose metabolism in response to stimulation is accompanied by mobilization of endogenous inosine into pentose phosphates, via the action of purine nucleotide phosphorylase (PNP). The PNP reaction is an integral part of the neuronal response to stimulation, as inhibiting PNP leaves DGCs energetically impaired during recovery from strong activation. Performing MSI on brain slices bridges the gap between live cell physiology and the deep chemical analysis enabled by mass spectrometry.

7.
Updates Surg ; 74(5): 1521-1531, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35986865

ABSTRACT

Surgical procedures are often impeded by bleeding and/or leakage of body fluids. These complications cannot always be resolved by conventional surgical techniques. Hemopatch® is a hemostatic patch that also functions as a sealant. Here we document the effectiveness and safety of Hemopatch® for routine procedures of multiple surgical disciplines. To this end, we performed a prospective, multicenter, single-arm, observational registry study. Patients were eligible if they had received Hemopatch® during an open or minimally invasive procedure in one of these specialties: hepatobiliary, cardiovascular, urological, neurological/spinal, general, or lung surgery. Patients were excluded if they had a known hypersensitivity to bovine proteins or brilliant blue, intraoperative pulsatile or severe bleeding and/or infection at the target application site (TAS). The primary endpoint for intraoperative effectiveness was hemostasis assessed as the percentage of patients achieving hemostasis within 2 min and the percentage of patients achieving hemostasis without re-bleeding at the time of surgical closure. The registry enrolled 621 patients at 23 study sites in six European countries. Six hundred twenty patients had completed follow-up information. Hemostasis within 2 min was achieved at 463 (74.5%) of all 621 TASs. Hemostasis without re-bleeding was observed at 620 (99.8%) TASs. Adverse events were reported in 64 patients (10.3%). This Hemopatch® registry shows that Hemopatch® efficiently establishes hemostasis and sealing in a variety of surgical specialties, including minimally invasive procedures. Furthermore, we provide evidence for the safety of Hemopatch® across all the specialties included in the registry. This study is registered at clinicaltrials.gov: NCT03392662.


Subject(s)
Hemostatics , Specialties, Surgical , Animals , Blood Loss, Surgical , Cattle , Hemostasis, Surgical/methods , Hemostatics/adverse effects , Humans , Prospective Studies , Registries , Treatment Outcome
8.
Glia ; 70(5): 820-841, 2022 05.
Article in English | MEDLINE | ID: mdl-35019164

ABSTRACT

Fecal-oral contamination promotes malnutrition pathology. Lasting consequences of early life malnutrition include cognitive impairment, but the underlying pathology and influence of gut microbes remain largely unknown. Here, we utilize an established murine model combining malnutrition and iterative exposure to fecal commensals (MAL-BG). The MAL-BG model was analyzed in comparison to malnourished (MAL mice) and healthy (CON mice) controls. Malnourished mice display poor spatial memory and learning plasticity, as well as altered microglia, non-neuronal CNS cells that regulate neuroimmune responses and brain plasticity. Chronic fecal-oral exposures shaped microglial morphology and transcriptional profile, promoting phagocytic features in MAL-BG mice. Unexpectedly, these changes occurred independently from significant cytokine-induced inflammation or blood-brain barrier (BBB) disruption, key gut-brain pathways. Metabolomic profiling of the MAL-BG cortex revealed altered polyunsaturated fatty acid (PUFA) profiles and systemic lipoxidative stress. In contrast, supplementation with an ω3 PUFA/antioxidant-associated diet (PAO) mitigated cognitive deficits within the MAL-BG model. These findings provide valued insight into the malnourished gut microbiota-brain axis, highlighting PUFA metabolism as a potential therapeutic target.


Subject(s)
Gastrointestinal Microbiome , Malnutrition , Animals , Cognition , Gastrointestinal Microbiome/physiology , Malnutrition/complications , Mice , Mice, Inbred C57BL , Microglia
9.
Glia ; 69(3): 567-578, 2021 03.
Article in English | MEDLINE | ID: mdl-32946147

ABSTRACT

Immunometabolism refers to the rearrangement of metabolic pathways in response to immune stimulation, and the ability of these metabolic pathways themselves to control immune functions. Many aspects of immunometabolism have been revealed through studies of peripheral immune cells. However, immunometabolic reprogramming of microglia, the resident immune cell of the central nervous system, and the consequential outcome on neuronal activity have remained difficult to unravel. Microglia are highly sensitive to subtle changes in their environment, limiting the techniques available to study their metabolic and inflammatory profiles. Here, using fluorescence lifetime imaging of endogenous NAD(P)H, we measure the metabolic activity of individual microglia within acute hippocampal slices. We observed an LPS-induced increase in aerobic glycolysis, which was blocked by the addition of 5 mM 2-deoxyglucose (2DG). This LPS-induced glycolysis in microglia was necessary for the stabilization of hypoxia inducible factor-1α (HIF-1α) and production of the proinflammatory cytokine, interleukin-1ß (IL-1ß). Upon release, IL-1ß acted via the neuronal interleukin-1 receptor to inhibit the formation of synaptic long-term potentiation (LTP) following high frequency stimulation. Remarkably, the addition of 2DG to blunt the microglial glycolytic increase also inhibited HIF-1α accumulation and IL-1ß production, and therefore rescued LTP in LPS-stimulated slices. Overall, these data reveal the importance of metabolic reprogramming in regulating microglial immune functions, with appreciable outcomes on cytokine release and neuronal activity.


Subject(s)
Long-Term Potentiation , Microglia , Cytokines/metabolism , Hippocampus/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism
10.
Trends Neurosci ; 43(11): 854-869, 2020 11.
Article in English | MEDLINE | ID: mdl-32958333

ABSTRACT

Immune cells react to their environment by flexibly reprogramming intracellular metabolic pathways that subsequently alter immune function, in a process called immunometabolism. However, in the CNS, the impact of metabolic reprogramming on microglia, neuroinflammation, and subsequently on brain function is poorly understood. As brain-resident macrophages, microglia are the CNS immune effectors and share similarities with peripheral immune cells. New tools for studying immunometabolism now allow the analysis of bioenergetic regulation with cellular resolution and, as a result, have uncovered previously unappreciated roles for microglial immunometabolism in shaping neuroinflammation. This review highlights evidence that microglia metabolism adapts to changes in brain energy homeostasis and that metabolic reprogramming regulates microglial polarization, thereby impacting pathological inflammatory responses in the brain.


Subject(s)
Inflammation , Microglia , Brain , Energy Metabolism , Humans
11.
Nat Commun ; 11(1): 1559, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214088

ABSTRACT

Microglia are highly motile cells that continuously monitor the brain environment and respond to damage-associated cues. While glucose is the main energy substrate used by neurons in the brain, the nutrients metabolized by microglia to support surveillance of the parenchyma remain unexplored. Here, we use fluorescence lifetime imaging of intracellular NAD(P)H and time-lapse two-photon imaging of microglial dynamics in vivo and in situ, to show unique aspects of the microglial metabolic signature in the brain. Microglia are metabolically flexible and can rapidly adapt to consume glutamine as an alternative metabolic fuel in the absence of glucose. During insulin-induced hypoglycemia in vivo or in aglycemia in acute brain slices, glutaminolysis supports the maintenance of microglial process motility and damage-sensing functions. This metabolic shift sustains mitochondrial metabolism and requires mTOR-dependent signaling. This remarkable plasticity allows microglia to maintain their critical surveillance and phagocytic roles, even after brain neuroenergetic homeostasis is compromised.


Subject(s)
Brain/immunology , Energy Metabolism/physiology , Microglia/metabolism , Animals , Brain/pathology , CX3C Chemokine Receptor 1/genetics , Cell Movement , Fatty Acids/metabolism , Glucose/deficiency , Glucose/metabolism , Glutamine/metabolism , Immunologic Surveillance , Mice , Mice, Transgenic , Microglia/cytology , Microglia/immunology , NAD/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
12.
Biomed Opt Express ; 10(9): 4381-4394, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31565496

ABSTRACT

Autofluorescence of endogenous molecules can provide valuable insights in both basic research and clinical applications. One such technique is fluorescence lifetime imaging (FLIM) of NAD(P)H, which serves as a correlate of glycolysis and electron transport chain rates in metabolically active tissue. A powerful advantage of NAD(P)H-FLIM is the ability to measure cell-specific metabolism within heterogeneous tissues. Cell-type specific identification is most commonly achieved with directed green fluorescent protein (GFP) expression. However, we demonstrate that NAD(P)H-FLIM should not be analyzed in GFP-expressing cells, as GFP molecules themselves emit photons in the blue spectrum with short fluorescence lifetimes when imaged using two-photon excitation at 750 nm. This is substantially different from the reported GFP emission wavelength and lifetime after two-photon excitation at 910 nm. These blue GFP photons are indistinguishable from free NAD(P)H by both emission spectra and fluorescence lifetime. Therefore, NAD(P)H-FLIM in GFP-expressing cells will lead to incorrect interpretations of metabolic rates, and thus, these techniques should not be combined.

13.
Cell Rep ; 27(10): 2895-2908.e4, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167136

ABSTRACT

Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.


Subject(s)
Brain/diagnostic imaging , Cyclic AMP/metabolism , Microglia/metabolism , Pseudopodia/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microglia/drug effects , Microtubules/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Pseudopodia/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction
14.
Dev Neurobiol ; 78(6): 593-603, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29271125

ABSTRACT

Investigations on the role of microglia in the brain have traditionally been focused on their contributions to disease states. However, recent observations have now convincingly shown that microglia in the healthy brain are not passive bystanders, but instead play a critical role in both central nervous system development and homeostasis of synaptic circuits in the adult. Here, we review the various mechanisms by which microglia impact neuronal communication in the healthy adult brain, both by sensing nearby synaptic responses and by actively modulating neuronal function. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 593-603, 2018.


Subject(s)
Brain/physiology , Microglia/physiology , Neurons/physiology , Animals , Cell Communication/physiology , Humans
15.
eNeuro ; 5(6)2018.
Article in English | MEDLINE | ID: mdl-30627639

ABSTRACT

Microglia are dynamic immune cells of the central nervous system, and their morphology is commonly used as a readout of cellular function. However, current morphological analysis techniques rely on either tracing of cells or two-dimensional projection analysis, which are time-consuming, subject to bias, and may ignore important three-dimensional (3D) information. Therefore, we have created 3DMorph, a MATLAB-based script that analyzes microglial morphology from 3D data. The program initially requires input of threshold levels, cell size expectations, and preferred methods of skeletonization. This makes 3DMorph easily scalable and adaptable to different imaging parameters or cell types. After these settings are defined, the program is completely automatic and can batch process files without user input. Output data includes cell volume, territorial volume, branch length, number of endpoints and branch points, and average distance between cells. We show that 3DMorph is accurate compared to manual tracing, with significantly decreased user input time. Importantly, 3DMorph is capable of processing in vivo microglial morphology, as well as other 3D branching cell types, from mouse cranial windows or acute hippocampal slices. Therefore, we present a novel, user-friendly, scalable, and semiautomatic method of analyzing cell morphology in 3 dimensions. This method should improve the accuracy of cell measurements, remove user bias between conditions, increase reproducibility between experimenters and labs, and reduce user input time. We provide this open source code on GitHub so that it is free and accessible to all investigators.


Subject(s)
Brain/cytology , Electronic Data Processing/methods , Microglia/cytology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Brain/diagnostic imaging , Brain/drug effects , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Size , Excitatory Amino Acid Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Neurons/cytology , Neurons/drug effects , Rats , Reproducibility of Results , Sodium Channel Blockers/pharmacology , Software , Tetrodotoxin/pharmacology
16.
Exp Neurol ; 283(Pt A): 1-15, 2016 09.
Article in English | MEDLINE | ID: mdl-27235933

ABSTRACT

The absence of axonal regeneration after spinal cord injury (SCI) has been attributed to the up-regulation of axon-repelling molecules, such as chondroitin sulfate proteoglycans (CSPGs) present in the glial scar that forms post-SCI. We previously identified the transcription factor SOX9 as a key up-regulator of CSPG production and also demonstrated that conditional Sox9 ablation leads to decreased CSPG levels and improved recovery of hind limb function after SCI. We herein demonstrate increased neural input onto spinal neurons caudal to the lesion in spinal cord injured Sox9 conditional knock out mice as indicated by increased levels of the presynaptic markers synaptophysin and vesicular glutamate transporter 1 (VGLUT1) compared to controls. Axonal sparing, long-range axonal regeneration and reactive sprouting were investigated as possible explanations for the increase in neural inputs caudal to the lesion and for the improved locomotor outcomes in spinal cord-injured Sox9 conditional knock out mice. Whereas retrograde tract-tracing studies failed to reveal any evidence for increased axonal sparing or for long-range regeneration in the Sox9 conditional knock out mice, anterograde tract-tracing experiments demonstrated increased reactive sprouting caudal to the lesion after SCI. Finally we demonstrate that application of a broad spectrum MMP inhibitor to reduce CSPG degradation in Sox9 conditional knock out mice prevents the improvements in locomotor recovery observed in untreated Sox9 conditional knock out mice. These results suggest that improved recovery of locomotor function in Sox9 conditional knock out mice after SCI is due to increased reactive sprouting secondary to reduced CSPG levels distal to the lesion.


Subject(s)
Locomotion/genetics , Recovery of Function/genetics , SOX9 Transcription Factor/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Axons/drug effects , Axons/pathology , Biotin/analogs & derivatives , Biotin/pharmacokinetics , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Dextrans/pharmacokinetics , Disease Models, Animal , Doxycycline/pharmacology , Doxycycline/therapeutic use , Edema/etiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Locomotion/physiology , Mice , Nerve Tissue Proteins/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recovery of Function/physiology , SOX9 Transcription Factor/genetics , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/genetics , Stilbamidines/pharmacokinetics , Synaptophysin/genetics , Synaptophysin/metabolism , Time Factors , Up-Regulation/genetics , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism
17.
Neurotherapeutics ; 10(4): 757-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24081781

ABSTRACT

Spinal cord injury results from an insult inflicted on the spinal cord that usually encompasses its 4 major functions (motor, sensory, autonomic, and reflex). The type of deficits resulting from spinal cord injury arise from primary insult, but their long-term severity is due to a multitude of pathophysiological processes during the secondary phase of injury. The failure of the mammalian spinal cord to regenerate and repair is often attributed to the very feature that makes the central nervous system special-it becomes so highly specialized to perform higher functions that it cannot effectively reactivate developmental programs to re-build novel circuitry to restore function after injury. Added to this is an extensive gliotic and immune response that is essential for clearance of cellular debris, but also lays down many obstacles that are detrimental to regeneration. Here, we discuss how the mature chromatin state of different central nervous system cells (neural, glial, and immune) may contribute to secondary pathophysiology, and how restoring silenced developmental gene expression by altering histone acetylation could stall secondary damage and contribute to novel approaches to stimulate endogenous repair.


Subject(s)
Epigenesis, Genetic , Nerve Regeneration/genetics , Spinal Cord Injuries/genetics , Spinal Cord/physiopathology , Humans , Spinal Cord Injuries/physiopathology
18.
J Neurosci ; 33(10): 4468-81, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23467362

ABSTRACT

Secreted protein acidic rich in cysteine (SPARC) is a matricellular protein that modulates the activity of growth factors, cytokines, and extracellular matrix to play multiple roles in tissue development and repair, such as cellular adhesion, migration, and proliferation. Throughout the CNS, SPARC is highly localized in mature ramified microglia, but its role in microglia--in development or during response to disease or injury--is not understood. In the postnatal brain, immature amoeboid myeloid precursors only induce SPARC expression after they cease proliferation and migration, and transform into mature, ramified resting microglia. SPARC null/CX3CR1-GFP reporter mice reveal that SPARC regulates the distribution and branching of mature microglia, with significant differences between cortical gray and white matter in both controls and SPARC nulls. Following ischemic and excitotoxic lesion, reactive, hypertrophic microglia rapidly downregulate and release SPARC at the lesion, concomitant with reactive, hypertrophic perilesion astrocytes upregulating SPARC. After photothrombotic stroke in the forelimb sensorimotor cortex, SPARC nulls demonstrate enhanced microgliosis in and around the lesion site, which accompanies significantly enhanced functional recovery by 32 d after lesion. Microglia from SPARC nulls also intrinsically proliferate at a greater rate in vitro--an enhanced effect that can be rescued by the addition of exogenous SPARC. SPARC is thus a novel regulator of microglial proliferation and structure, and, in addition to regulating glioma progression, may play an important role in differently regulating the gray and white matter microglial responses to CNS lesion--and modulating behavioral recovery--after injury.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/pathology , Cerebral Cortex/pathology , Gliosis/etiology , Glycoproteins/metabolism , Recovery of Function/physiology , Tumor Suppressor Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Brain Infarction/etiology , Brain Infarction/pathology , Brain Ischemia/etiology , CX3C Chemokine Receptor 1 , Calcium-Binding Proteins/metabolism , Cell Count , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Size , Cells, Cultured , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Female , Forelimb/physiopathology , Galectin 3/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Genotype , Glial Fibrillary Acidic Protein/metabolism , Glycoproteins/deficiency , Glycoproteins/pharmacology , Green Fluorescent Proteins/genetics , Intracranial Thrombosis/complications , Lectins/metabolism , Male , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/physiology , Motor Skills/drug effects , Motor Skills/physiology , Mutation/genetics , N-Methylaspartate/toxicity , Olfactory Bulb/injuries , Osteonectin , Proliferating Cell Nuclear Antigen/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Chemokine/genetics , Time Factors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/pharmacology , NF-kappaB-Inducing Kinase
19.
Am J Surg ; 202(3): e31-3, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21871977

ABSTRACT

Incising the external oblique muscle aponeurosis is an important part of the components separation technique for the repair of large incisional hernias. Endoscopically assisted section has been suggested to prevent complications of extensive skin flap formation. We used a simplified method for incising the external oblique aponeurosis, using a modified Collin Hartmann retractor, in 14 patients for the repair of large incisional hernias. Eight women and 6 men, with a mean (± standard deviation) age of 61.9 ± 14.9 years, underwent surgery. The median transverse diameter of the defect was 8.6 ± 3.0 cm. No postoperative morbidity occurred except 1 case of skin necrosis. One patient had a recurrence. Sectioning the external oblique aponeurosis during the components separation method using the technique described is a simple, safe, and economic approach that can prevent the complications described after the original techniques.


Subject(s)
Abdominal Muscles/surgery , Abdominal Wall/surgery , Hernia, Ventral/surgery , Laparotomy/methods , Adult , Aged , Fasciotomy , Female , Humans , Laparotomy/adverse effects , Laparotomy/instrumentation , Male , Middle Aged , Recurrence , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL