Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Stem Cell Reports ; 18(12): 2300-2312, 2023 12 12.
Article in English | MEDLINE | ID: mdl-37977142

ABSTRACT

Human fetal tissue and cells derived from fetal tissue are crucial for biomedical research. Fetal tissues and cells are used to study both normal development and developmental disorders. They are broadly applied in vaccine development and production. Further, research using cells from fetal tissue is instrumental for studying many infectious diseases, including a broad range of viruses. These widespread applications underscore the value of fetal tissue research and reflect an important point: cells derived from fetal tissues have capabilities that cells from other sources do not. In many cases, increased functionality of cells derived from fetal tissues arises from increased proliferative capacity, ability to survive in culture, and developmental potential that is attenuated in adult tissues. This review highlights important, representative applications of fetal tissue for science and medicine.


Subject(s)
Fetal Research , Fetus , Adult , Humans
2.
Mol Brain ; 16(1): 75, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37924159

ABSTRACT

Non-familial Alzheimer's disease (AD) occurring before 65 years of age is commonly referred to as early-onset Alzheimer's disease (EOAD) and constitutes ~ 5-6% of all AD cases (Mendez et al. in Continuum 25:34-51, 2019). While EOAD exhibits the same clinicopathological changes such as amyloid plaques, neurofibrillary tangles (NFTs), brain atrophy, and cognitive decline (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022; Caldwell et al. in Mol Brain 15:83, 2022) as observed in the more prevalent late-onset AD (LOAD), EOAD patients tend to have more severe cognitive deficits, including visuospatial, language, and executive dysfunction (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022). Patient-derived induced pluripotent stem cells (iPSCs) have been used to model and study penetrative, familial AD (FAD) mutations in APP, PSEN1, and PSEN2 (Valdes et al. in Research Square 1-30, 2022; Caldwell et al. in Sci Adv 6:1-16, 2020) but have been seldom used for sporadic forms of AD that display more heterogeneous disease mechanisms. In this study, we sought to characterize iPSC-derived neurons from EOAD patients via RNA sequencing. A modest difference in expression profiles between EOAD patients and non-demented control (NDC) subjects resulted in a limited number of differentially expressed genes (DEGs). Based on this analysis, we provide evidence that iPSC-derived neuron model systems, likely due to the loss of EOAD-associated epigenetic signatures arising from iPSC reprogramming, may not be ideal models for studying sporadic AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Neurons/pathology
3.
Methods Mol Biol ; 2561: 105-133, 2023.
Article in English | MEDLINE | ID: mdl-36399267

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disorder and a mechanistically complex disease. For the last decade, human models of AD using induced pluripotent stem cells (iPSCs) have emerged as a powerful way to understand disease pathogenesis in relevant human cell types. In this review, we summarize the state of the field and how this technology can apply to studies of both familial and sporadic studies of AD. We discuss patient-derived iPSCs, genome editing, differentiation of neural cell types, and three-dimensional organoids, and speculate on the future of this type of work for increasing our understanding of, and improving therapeutic development for, this devastating disease.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Neurons/pathology , Neuroglia/pathology , Organoids/pathology
4.
J Neurosci ; 41(49): 10034-10053, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34663629

ABSTRACT

Traumatic brain injury (TBI) results in disrupted brain function following impact from an external force and is a risk factor for sporadic Alzheimer's disease (AD). Although neurologic symptoms triggered by mild traumatic brain injuries (mTBI), the most common form of TBI, typically resolve rapidly, even an isolated mTBI event can increase the risk to develop AD. Aberrant accumulation of amyloid ß peptide (Aß), a cleaved fragment of amyloid precursor protein (APP), is a key pathologic outcome designating the progression of AD following mTBI and has also been linked to impaired axonal transport. However, relationships among mTBI, amyloidogenesis, and axonal transport remain unclear, in part because of the dearth of human models to study the neuronal response following mTBI. Here, we implemented a custom-microfabricated device to deform neurons derived from human-induced pluripotent stem cells, derived from a cognitively unimpaired male individual, to mimic the mild stretch experienced by neurons during mTBI. Although no cell lethality or cytoskeletal disruptions were observed, mild stretch was sufficient to stimulate rapid amyloidogenic processing of APP. This processing led to abrupt cessation of APP axonal transport and progressive formation of aberrant axonal accumulations that contained APP, its processing machinery, and amyloidogenic fragments. Consistent with this sequence of events, stretch-induced defects were abrogated by reducing amyloidogenesis either pharmacologically or genetically. In sum, we have uncovered a novel and manipulable stretch-induced amyloidogenic pathway directly responsible for APP axonal transport dysregulation. Our findings may help to understand and ultimately mitigate the risk of developing AD following mTBI.SIGNIFICANCE STATEMENT Mild traumatic brain injury is a risk factor for sporadic Alzheimer's disease (AD). Increased amyloid ß peptide generation after injury may drive this risk. Here, by using a custom-built device to impose mild stretch to human neurons, we found that stretch triggers amyloid precursor protein (APP) cleavage, and thus amyloid ß peptide generation, consequently disrupting APP axonal transport. Compellingly, protecting APP from cleavage was sufficient to spare axonal transport dysregulation and the consequent aberrant axonal accumulation of APP. Supporting such protective mechanism, the expression of the AD-protective APPA673T genetic variant conferred protection against stretch-induced APP axonal transport phenotypes. Our data reveal potential subcellular pathways contributing to the development of AD-associated phenotypes following mild traumatic brain injury, and putative strategies for intervening in these pathways.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axonal Transport/physiology , Neurons/metabolism , Neurons/pathology , Alzheimer Disease/etiology , Brain Concussion/complications , Brain Concussion/metabolism , Brain Concussion/pathology , Cell Culture Techniques/methods , Humans , Induced Pluripotent Stem Cells , Male
6.
Cell Stem Cell ; 28(9): 1533-1548.e6, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33910058

ABSTRACT

Sporadic Alzheimer's disease (AD) exclusively affects elderly people. Using direct conversion of AD patient fibroblasts into induced neurons (iNs), we generated an age-equivalent neuronal model. AD patient-derived iNs exhibit strong neuronal transcriptome signatures characterized by downregulation of mature neuronal properties and upregulation of immature and progenitor-like signaling pathways. Mapping iNs to longitudinal neuronal differentiation trajectory data demonstrated that AD iNs reflect a hypo-mature neuronal identity characterized by markers of stress, cell cycle, and de-differentiation. Epigenetic landscape profiling revealed an underlying aberrant neuronal state that shares similarities with malignant transformation and age-dependent epigenetic erosion. To probe for the involvement of aging, we generated rejuvenated iPSC-derived neurons that showed no significant disease-related transcriptome signatures, a feature that is consistent with epigenetic clock and brain ontogenesis mapping, which indicate that fibroblast-derived iNs more closely reflect old adult brain stages. Our findings identify AD-related neuronal changes as age-dependent cellular programs that impair neuronal identity.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Aged , Aging , Fibroblasts , Humans , Neurons
8.
Prog Mol Biol Transl Sci ; 177: 109-122, 2021.
Article in English | MEDLINE | ID: mdl-33453937

ABSTRACT

Recent data establish multiple defects in endocytic functions as early events initiating various neurodegenerative disorders, including Alzheimer's disease (AD). The genetic landscape resulting from genome-wide association studies (GWAS) reveals changes in post-endocytic trafficking of amyloid precursor protein (APP) in neurons leading to an increase in amyloidogenic processing, deficits in amyloid beta (Aß) clearance, increases in intracellular Aß, and other endosomal pathogenic phenotypes. Multiple genetic factors regulate each segment of endosomal and post-endosomal trafficking. Intriguingly, several studies indicate endosomal dysfunctions preceding Aß pathology and tau phosphorylation. In this chapter we highlight the role of various GWAS-identified endosomal and post-endosomal gene products in initiating AD pathologies. We also summarize the functions of various genetic modifiers of post-endocytic trafficking of APP that may work as targets for therapeutic intervention in AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Endosomes/metabolism , Genome-Wide Association Study , Humans , Protein Transport
10.
Mol Biol Cell ; 32(3): 247-259, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33296223

ABSTRACT

Amyloid beta (Aß) is a major component of amyloid plaques, which are a key pathological hallmark found in the brains of Alzheimer's disease (AD) patients. We show that statins are effective at reducing Aß in human neurons from nondemented control subjects, as well as subjects with familial AD and sporadic AD. Aß is derived from amyloid precursor protein (APP) through sequential proteolytic cleavage by BACE1 and γ-secretase. While previous studies have shown that cholesterol metabolism regulates APP processing to Aß, the mechanism is not well understood. We used iPSC-derived neurons and bimolecular fluorescence complementation assays in transfected cells to elucidate how altering cholesterol metabolism influences APP processing. Altering cholesterol metabolism using statins decreased the generation of sAPPß and increased levels of full-length APP (flAPP), indicative of reduced processing of APP by BACE1. We further show that statins decrease flAPP interaction with BACE1 and enhance APP dimerization. Additionally, statin-induced changes in APP dimerization and APP-BACE1 are dependent on cholesterol binding to APP. Our data indicate that statins reduce Aß production by decreasing BACE1 interaction with flAPP and suggest that this process may be regulated through competition between APP dimerization and APP cholesterol binding.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/physiology , Aspartic Acid Endopeptidases/metabolism , Cholesterol/metabolism , Dimerization , HEK293 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/drug effects , Neurons/physiology , Protein Binding
11.
Nat Rev Neurosci ; 21(1): 21-35, 2020 01.
Article in English | MEDLINE | ID: mdl-31780819

ABSTRACT

The global epidemic of Alzheimer disease (AD) is worsening, and no approved treatment can revert or arrest progression of this disease. AD pathology is characterized by the accumulation of amyloid-ß (Aß) plaques and tau neurofibrillary tangles in the brain. Genetic data, as well as autopsy and neuroimaging studies in patients with AD, indicate that Aß plaque deposition precedes cortical tau pathology. Because Aß accumulation has been considered the initial insult that drives both the accumulation of tau pathology and tau-mediated neurodegeneration in AD, the development of AD therapeutics has focused mostly on removing Aß from the brain. However, striking preclinical evidence from AD mouse models and patient-derived human induced pluripotent stem cell models indicates that tau pathology can progress independently of Aß accumulation and arises downstream of genetic risk factors for AD and aberrant metabolic pathways. This Review outlines novel insights from preclinical research that implicate apolipoprotein E, the endocytic system, cholesterol metabolism and microglial activation as Aß-independent regulators of tau pathology. These factors are discussed in the context of emerging findings from clinical pathology, functional neuroimaging and other approaches in humans. Finally, we discuss the implications of these new insights for current Aß-targeted strategies and highlight the emergence of novel therapeutic strategies that target processes upstream of both Aß and tau.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Alzheimer Disease/therapy , Animals , Apolipoproteins E/metabolism , Cholesterol/metabolism , Endocytosis , Humans , Microglia/metabolism , Plaque, Amyloid/pathology
12.
Nat Genet ; 51(12): 1691-1701, 2019 12.
Article in English | MEDLINE | ID: mdl-31740836

ABSTRACT

In the mammalian genome, the clustered protocadherin (cPCDH) locus provides a paradigm for stochastic gene expression with the potential to generate a unique cPCDH combination in every neuron. Here we report a chromatin-based mechanism that emerges during the transition from the naive to the primed states of cell pluripotency and reduces, by orders of magnitude, the combinatorial potential in the human cPCDH locus. This mechanism selectively increases the frequency of stochastic selection of a small subset of cPCDH genes after neuronal differentiation in monolayers, 10-month-old cortical organoids and engrafted cells in the spinal cords of rats. Signs of these frequent selections can be observed in the brain throughout fetal development and disappear after birth, except in conditions of delayed maturation such as Down's syndrome. We therefore propose that a pattern of limited cPCDH-gene expression diversity is maintained while human neurons still retain fetal-like levels of maturation.


Subject(s)
Cadherins/genetics , Chromatin/genetics , Down Syndrome/pathology , Induced Pluripotent Stem Cells/cytology , Neurons/physiology , Adult , Animals , Astrocytes/cytology , Astrocytes/physiology , Brain/cytology , Brain/embryology , Cell Differentiation , Cell Line , Down Syndrome/genetics , Gene Expression Regulation , Histones/genetics , Humans , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Mice , Middle Aged , Neurons/cytology , Promoter Regions, Genetic , Rats , Single-Cell Analysis , Spinal Cord/cytology , Spinal Cord/transplantation , Transplantation, Heterologous
13.
Cell Stem Cell ; 24(3): 363-375.e9, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30686764

ABSTRACT

Genetic, epidemiologic, and biochemical evidence suggests that predisposition to Alzheimer's disease (AD) may arise from altered cholesterol metabolism, although the molecular pathways that may link cholesterol to AD phenotypes are only partially understood. Here, we perform a phenotypic screen for pTau accumulation in AD-patient iPSC-derived neurons and identify cholesteryl esters (CE), the storage product of excess cholesterol, as upstream regulators of Tau early during AD development. Using isogenic induced pluripotent stem cell (iPSC) lines carrying mutations in the cholesterol-binding domain of APP or APP null alleles, we found that while CE also regulate Aß secretion, the effects of CE on Tau and Aß are mediated by independent pathways. Efficacy and toxicity screening in iPSC-derived astrocytes and neurons showed that allosteric activation of CYP46A1 lowers CE specifically in neurons and is well tolerated by astrocytes. These data reveal that CE independently regulate Tau and Aß and identify a druggable CYP46A1-CE-Tau axis in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cholesterol/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL
14.
Science ; 363(6424): 207, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30655416
15.
J Biol Chem ; 293(29): 11341-11357, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29858247

ABSTRACT

Mounting evidence suggests that alterations in cholesterol homeostasis are involved in Alzheimer's disease (AD) pathogenesis. Amyloid precursor protein (APP) or multiple fragments generated by proteolytic processing of APP have previously been implicated in the regulation of cholesterol metabolism. However, the physiological function of APP in regulating lipoprotein homeostasis in astrocytes, which are responsible for de novo cholesterol biosynthesis and regulation in the brain, remains unclear. To address this, here we used CRISPR/Cas9 genome editing to generate isogenic APP-knockout (KO) human induced pluripotent stem cells (hiPSCs) and differentiated them into human astrocytes. We found that APP-KO astrocytes have reduced cholesterol and elevated levels of sterol regulatory element-binding protein (SREBP) target gene transcripts and proteins, which were both downstream consequences of reduced lipoprotein endocytosis. To elucidate which APP fragments regulate cholesterol homeostasis and to examine whether familial AD mutations in APP affect lipoprotein metabolism, we analyzed an isogenic allelic series harboring the APP Swedish and APP V717F variants. Only astrocytes homozygous for the APP Swedish (APPSwe/Swe) mutation, which had reduced full-length APP (FL APP) due to increased ß-secretase cleavage, recapitulated the APP-KO phenotypes. Astrocytic internalization of ß-amyloid (Aß), another ligand for low-density lipoprotein (LDL) receptors, was also impaired in APP-KO and APPSwe/Swe astrocytes. Finally, impairing cleavage of FL APP through ß-secretase inhibition in APPSwe/Swe astrocytes reversed the LDL and Aß endocytosis defects. In conclusion, FL APP is involved in the endocytosis of LDL receptor ligands and is required for proper cholesterol homeostasis and Aß clearance in human astrocytes.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Amyloid beta-Protein Precursor/genetics , Astrocytes/cytology , CRISPR-Cas Systems , Cell Line , Endocytosis , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
16.
Stem Cell Reports ; 10(3): 1046-1058, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29503090

ABSTRACT

Developing effective therapeutics for complex diseases such as late-onset, sporadic Alzheimer's disease (SAD) is difficult due to genetic and environmental heterogeneity in the human population and the limitations of existing animal models. Here, we used hiPSC-derived neurons to test a compound that stabilizes the retromer, a highly conserved multiprotein assembly that plays a pivotal role in trafficking molecules through the endosomal network. Using this human-specific system, we have confirmed previous data generated in murine models and show that retromer stabilization has a potentially beneficial effect on amyloid beta generation from human stem cell-derived neurons. We further demonstrate that manipulation of retromer complex levels within neurons affects pathogenic TAU phosphorylation in an amyloid-independent manner. Taken together, our work demonstrates that retromer stabilization is a promising candidate for therapeutic development in AD and highlights the advantages of testing novel compounds in a human-specific, neuronal system.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Phosphorylation/physiology , tau Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Endosomes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neurons/metabolism , Protein Transport/physiology
17.
Stem Cell Reports ; 8(4): 1101-1111, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410643

ABSTRACT

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) offers the possibility of studying the molecular mechanisms underlying human diseases in cell types difficult to extract from living patients, such as neurons and cardiomyocytes. To date, studies have been published that use small panels of iPSC-derived cell lines to study monogenic diseases. However, to study complex diseases, where the genetic variation underlying the disorder is unknown, a sizable number of patient-specific iPSC lines and controls need to be generated. Currently the methods for deriving and characterizing iPSCs are time consuming, expensive, and, in some cases, descriptive but not quantitative. Here we set out to develop a set of simple methods that reduce cost and increase throughput in the characterization of iPSC lines. Specifically, we outline methods for high-throughput quantification of surface markers, gene expression analysis of in vitro differentiation potential, and evaluation of karyotype with markedly reduced cost.


Subject(s)
Genetic Variation , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/metabolism , Karyotyping/methods , Myocytes, Cardiac/metabolism , Neurons/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Cost-Benefit Analysis , Genotype , High-Throughput Screening Assays/economics , High-Throughput Screening Assays/instrumentation , Humans , Induced Pluripotent Stem Cells/cytology , Karyotyping/economics , Myocytes, Cardiac/cytology , Neurons/cytology , Phenotype
18.
Stem Cell Reports ; 8(4): 1086-1100, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410642

ABSTRACT

Large-scale collections of induced pluripotent stem cells (iPSCs) could serve as powerful model systems for examining how genetic variation affects biology and disease. Here we describe the iPSCORE resource: a collection of systematically derived and characterized iPSC lines from 222 ethnically diverse individuals that allows for both familial and association-based genetic studies. iPSCORE lines are pluripotent with high genomic integrity (no or low numbers of somatic copy-number variants) as determined using high-throughput RNA-sequencing and genotyping arrays, respectively. Using iPSCs from a family of individuals, we show that iPSC-derived cardiomyocytes demonstrate gene expression patterns that cluster by genetic background, and can be used to examine variants associated with physiological and disease phenotypes. The iPSCORE collection contains representative individuals for risk and non-risk alleles for 95% of SNPs associated with human phenotypes through genome-wide association studies. Our study demonstrates the utility of iPSCORE for examining how genetic variants influence molecular and physiological traits in iPSCs and derived cell lines.


Subject(s)
Arrhythmias, Cardiac/genetics , Databases, Factual , Genetic Association Studies , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Multigene Family , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Racial Groups
19.
Cell Rep ; 17(3): 759-773, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732852

ABSTRACT

We investigated early phenotypes caused by familial Alzheimer's disease (fAD) mutations in isogenic human iPSC-derived neurons. Analysis of neurons carrying fAD PS1 or APP mutations introduced using genome editing technology at the endogenous loci revealed that fAD mutant neurons had previously unreported defects in the recycling state of endocytosis and soma-to-axon transcytosis of APP and lipoproteins. The endocytosis reduction could be rescued through treatment with a ß-secretase inhibitor. Our data suggest that accumulation of ß-CTFs of APP, but not Aß, slow vesicle formation from an endocytic recycling compartment marked by the transcytotic GTPase Rab11. We confirm previous results that endocytosis is affected in AD and extend these to uncover a neuron-specific defect. Decreased lipoprotein endocytosis and transcytosis to the axon suggest that a neuron-specific impairment in endocytic axonal delivery of lipoproteins and other key materials might compromise synaptic maintenance in fAD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Induced Pluripotent Stem Cells/metabolism , Lipoproteins, LDL/metabolism , Mutation/genetics , Neurons/metabolism , Transcytosis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Axons/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Presenilin-1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , rab GTP-Binding Proteins/metabolism
20.
Am J Crit Care ; 24(5): 440-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26330437

ABSTRACT

BACKGROUND: Many patients each year require prolonged mechanical ventilation. Inflammatory processes may prevent successful weaning, and evidence indicates that mechanical ventilation induces oxidative stress in the diaphragm, resulting in atrophy and contractile dysfunction of diaphragmatic myofibers. Antioxidant supplementation might mitigate the harmful effects of the oxidative stress induced by mechanical ventilation. OBJECTIVE: To test the clinical effectiveness of antioxidant supplementation in reducing the duration of mechanical ventilation. METHODS: A randomized, prospective, placebo-controlled double-blind design was used to test whether enterally administered antioxidant supplementation would decrease the duration of mechanical ventilation, all-cause mortality, and length of stay in the intensive care unit and hospital. Patients received vitamin C 1000 mg plus vitamin E 1000 IU, vitamin C 1000 mg plus vitamin E 1000 IU plus N-acetylcysteine 400 mg, or placebo solution as a bolus injection via their enteral feeding tube every 8 hours. RESULTS: Clinical and statistically significant differences in duration of mechanical ventilation were seen among the 3 groups (Mantel-Cox log rank statistic = 5.69, df = 1, P = .017). The 3 groups did not differ significantly in all-cause mortality during hospitalization or in the length of stay in the intensive care unit or hospital. CONCLUSIONS: Enteral administration of antioxidants is a simple, safe, inexpensive, and effective intervention that decreases the duration of mechanical ventilation in critically ill adults.


Subject(s)
Antioxidants/therapeutic use , Critical Care/methods , Inflammation/drug therapy , Inflammation/etiology , Oxidative Stress/drug effects , Respiration, Artificial/adverse effects , Adult , Aged , Aged, 80 and over , Ascorbic Acid/therapeutic use , Critical Illness , Cystine/analogs & derivatives , Cystine/therapeutic use , Double-Blind Method , Female , Humans , Length of Stay , Male , Middle Aged , Prospective Studies , Time Factors , Treatment Outcome , Vitamin E/therapeutic use , Vitamins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...