Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Clin Psychol Rev ; 110: 102428, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657292

ABSTRACT

Cognitive reappraisal, an adaptive emotion regulation strategy that involves subjectively reinterpreting stressful and adverse experiences in a more positive manner, can enhance personal resilience. Personal resilience is a constellation of attributes that facilitate successful coping and an expeditious return to adaptive functioning after exposure to stress or adversity. This meta-analysis evaluated the association between cognitive reappraisal and personal resilience. A systematic and exhaustive search identified 64 independent samples from 55 studies (N = 29,824) that examined the correlation between cognitive reappraisal and personal resilience. A random-effects model revealed a positive summary effect (r = 0.47, p < .001), indicating that higher cognitive reappraisal was associated with higher personal resilience. Six potential meta-moderators were tested: culture, age, name of the cognitive reappraisal measure, name of the personal resilience measure, study design, and publication period. After two extreme effect size outliers were omitted, tests of publication bias did not reveal any publication bias in this line of research. This quantitative synthesis offers compelling evidence showing that cognitive reappraisal skills operate as a protective strategy against stress and adversity and, therefore, enhance personal resilience. The protective benefits of cognitive reappraisal in relation to personal resilience are relatively robust, as the correlations were statistically significant for all subgroups in the meta-moderation analyses.


Subject(s)
Resilience, Psychological , Humans , Cognition/physiology , Adaptation, Psychological/physiology , Emotional Regulation/physiology , Stress, Psychological/psychology
2.
Mol Genet Metab Rep ; 38: 101025, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38125072

ABSTRACT

Background: LBSL is a mitochondrial disorder caused by mutations in the mitochondrial aspartyl-tRNA synthetase gene DARS2, resulting in a distinctive pattern on brain magnetic resonance imaging (MRI) and spectroscopy. Clinical presentation varies from severe infantile to chronic, slowly progressive neuronal deterioration in adolescents or adults. Most individuals with LBSL are compound heterozygous for one splicing defect in an intron 2 mutational hotspot and a second defect that could be a missense, non-sense, or splice site mutation or deletion resulting in decreased expression of the full-length protein. Aim: To present a new family with two affected members with LBSL and report a novel DARS2 mutation. Results: An 8-year-old boy (Patient 1) was referred due to headaches and abnormal MRI, suggestive of LBSL. Genetic testing revealed a previously reported c.492 + 2 T > C mutation in the DARS2 gene. Sanger sequencing uncovered a novel variant c.228-17C > G in the intron 2 hotspot. Family studies found the same genetic changes in an asymptomatic 4-year-old younger brother (Patient 2), who was found on follow-up to have an abnormal MRI. mRNA extracted from patients' fibroblasts showed that the c.228-17C > G mutation caused skipping of exon 3 resulting in lower DARS2 mRNA level. Complete absence of DARS2 protein was also found in both patients. Summary: We present a new family with two children affected with LBSL and describe a novel mutation in the DARS2 intron 2 hotspot. Despite findings of extensive white matter disease in the brain and spine, the proband in this family presented only with headaches, while the younger sibling, who also had extensive white matter changes, was asymptomatic. Our in-vitro results confirmed skipping of exon 3 in patients and family members carrying the intron 2 variant, which is consistent with previous reported mutations in intron 2 hotspots. DARS2 mRNA and protein levels were also reduced in both patients, further supporting the pathogenicity of the novel variant.

3.
J Med Genet ; 58(5): 314-325, 2021 05.
Article in English | MEDLINE | ID: mdl-32518176

ABSTRACT

BACKGROUND: The nucleotide binding protein-like (NUBPL) gene was first reported as a cause of mitochondrial complex I deficiency (MIM 613621, 618242) in 2010. To date, only eight patients have been reported with this mitochondrial disorder. Five other patients were recently reported to have NUBPL disease but their clinical picture was different from the first eight patients. Here, we report clinical and genetic findings in five additional patients (four families). METHODS: Whole exome sequencing was used to identify patients with compound heterozygous NUBPL variants. Functional studies included RNA-Seq transcript analyses, missense variant biochemical analyses in a yeast model (Yarrowia lipolytica) and mitochondrial respiration experiments on patient fibroblasts. RESULTS: The previously reported c.815-27T>C branch-site mutation was found in all four families. In prior patients, c.166G>A [p.G56R] was always found in cis with c.815-27T>C, but only two of four families had both variants. The second variant found in trans with c.815-27T>C in each family was: c.311T>C [p.L104P] in three patients, c.693+1G>A in one patient and c.545T>C [p.V182A] in one patient. Complex I function in the yeast model was impacted by p.L104P but not p.V182A. Clinical features include onset of neurological symptoms at 3-18 months, global developmental delay, cerebellar dysfunction (including ataxia, dysarthria, nystagmus and tremor) and spasticity. Brain MRI showed cerebellar atrophy. Mitochondrial function studies on patient fibroblasts showed significantly reduced spare respiratory capacity. CONCLUSION: We report on five new patients with NUBPL disease, adding to the number and phenotypic variability of patients diagnosed worldwide, and review prior reported patients with pathogenic NUBPL variants.


Subject(s)
Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Adolescent , Brain/diagnostic imaging , Child , DNA Mutational Analysis , Female , Humans , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Diseases/physiopathology , Pedigree , RNA-Seq , Exome Sequencing , Young Adult
4.
Mol Genet Metab ; 126(1): 53-63, 2019 01.
Article in English | MEDLINE | ID: mdl-30473481

ABSTRACT

Primary mitochondrial complex I deficiency is the most common defect of the mitochondrial respiratory chain. It is caused by defects in structural components and assembly factors of this large protein complex. Mutations in the assembly factor NDUFAF5 are rare, with only five families reported to date. This study provides clinical, biochemical, molecular and functional data for four unrelated additional families, and three novel pathogenic variants. Three cases presented in infancy with lactic acidosis and classic Leigh syndrome. One patient, however, has a milder phenotype, with symptoms starting at 27 months and a protracted clinical course with improvement and relapsing episodes. She is homozygous for a previously reported mutation, p.Met279Arg and alive at 19 years with mild neurological involvement, normal lactate but abnormal urine organic acids. We found the same mutation in one of our severely affected patients in compound heterozygosity with a novel p.Lys52Thr mutation. Both patients with p.Met279Arg are of Taiwanese descent and had severe hyponatremia. Our third and fourth patients, both Caucasian, shared a common, newly described, missense mutation p.Lys109Asn which we show induces skipping of exon 3. Both Caucasian patients were compound heterozygotes, one with a previously reported Ashkenazi founder mutation while the other was negative for additional exonic variants. Whole genome sequencing followed by RNA studies revealed a novel deep intronic variant at position c.223-907A>C inducing an exonic splice enhancer. Our report adds significant new information to the mutational spectrum of NDUFAF5, further delineating the phenotypic heterogeneity of this mitochondrial defect.


Subject(s)
Electron Transport Complex I/deficiency , Leigh Disease/genetics , Methyltransferases/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mutation , Phenotype , Adolescent , Biopsy , Child , Child, Preschool , Electron Transport Complex I/genetics , Female , Humans , Infant , Male , Pedigree , Skin/pathology , Exome Sequencing , Whole Genome Sequencing , Young Adult
5.
Mol Genet Metab ; 119(1-2): 91-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27448789

ABSTRACT

In humans, mitochondrial DNA (mtDNA) depletion syndromes are a group of genetically and clinically heterogeneous autosomal recessive disorders that arise as a consequence of defects in mtDNA replication or nucleotide synthesis. Clinical manifestations are variable and include myopathic, encephalomyopathic, neurogastrointestinal or hepatocerebral phenotypes. Through clinical exome sequencing, we identified a homozygous missense variant (c.533C>T; p.Pro178Leu) in mitochondrial transcription factor A (TFAM) segregating in a consanguineous kindred of Colombian-Basque descent in which two siblings presented with IUGR, elevated transaminases, conjugated hyperbilirubinemia and hypoglycemia with progression to liver failure and death in early infancy. Results of the liver biopsy in the proband revealed cirrhosis, micro- and macrovesicular steatosis, cholestasis and mitochondrial pleomorphism. Electron microscopy of muscle revealed abnormal mitochondrial morphology and distribution while enzyme histochemistry was underwhelming. Electron transport chain testing in muscle showed increased citrate synthase activity suggesting mitochondrial proliferation, while respiratory chain activities were at the lower end of normal. mtDNA content was reduced in liver and muscle (11% and 21% of normal controls respectively). While Tfam mRNA expression was upregulated in primary fibroblasts, Tfam protein level was significantly reduced. Furthermore, functional investigations of the mitochondria revealed reduced basal respiration and spare respiratory capacity, decreased mtDNA copy number and markedly reduced nucleoids. TFAM is essential for transcription, replication and packaging of mtDNA into nucleoids. Tfam knockout mice display embryonic lethality secondary to severe mtDNA depletion. In this report, for the first time, we associate a homozygous variant in TFAM with a novel mtDNA depletion syndrome.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , Liver Failure/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Animals , DNA Replication/genetics , DNA, Mitochondrial/metabolism , Female , Homozygote , Humans , Infant, Newborn , Liver/metabolism , Liver/physiopathology , Liver Failure/physiopathology , Male , Mice , Mice, Knockout , Mitochondrial Diseases/physiopathology , Mutation, Missense , Neonatal Screening , Exome Sequencing
6.
J Vis Exp ; (112)2016 06 10.
Article in English | MEDLINE | ID: mdl-27341536

ABSTRACT

This paper describes how to use a custom manufactured, commercially available enclosed cell culture system for basic and preclinical research. Biosafety cabinets (BSCs) and incubators have long been the standard for culturing and expanding cell lines for basic and preclinical research. However, as the focus of many stem cell laboratories shifts from basic research to clinical translation, additional requirements are needed of the cell culturing system. All processes must be well documented and have exceptional requirements for sterility and reproducibility. In traditional incubators, gas concentrations and temperatures widely fluctuate anytime the cells are removed for feeding, passaging, or other manipulations. Such interruptions contribute to an environment that is not the standard for cGMP and GLP guidelines. These interruptions must be minimized especially when cells are utilized for therapeutic purposes. The motivation to move from the standard BSC and incubator system to a closed system is that such interruptions can be made negligible. Closed systems provide a work space to feed and manipulate cell cultures and maintain them in a controlled environment where temperature and gas concentrations are consistent. This way, pluripotent and multipotent stem cells can be maintained at optimum health from the moment of their derivation all the way to their eventual use in therapy.


Subject(s)
Neural Stem Cells , Pluripotent Stem Cells , Cell Culture Techniques , Humans , Incubators , Laboratories , Reproducibility of Results
8.
Mol Ther Methods Clin Dev ; 2: 14068, 2015.
Article in English | MEDLINE | ID: mdl-26052536

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is an inherited α-L-iduronidase (IDUA, I) deficiency in which glycosaminoglycan (GAG) accumulation causes progressive multisystem organ dysfunction, neurological impairment, and death. Current MPS I mouse models, based on a NOD/SCID (NS) background, are short-lived, providing a very narrow window to assess the long-term efficacy of therapeutic interventions. They also develop thymic lymphomas, making the assessment of potential tumorigenicity of human stem cell transplantation problematic. We therefore developed a new MPS I model based on a NOD/SCID/Il2rγ (NSG) background. This model lives longer than 1 year and is tumor-free during that time. NSG MPS I (NSGI) mice exhibit the typical phenotypic features of MPS I including coarsened fur and facial features, reduced/abnormal gait, kyphosis, and corneal clouding. IDUA is undetectable in all tissues examined while GAG levels are dramatically higher in most tissues. NSGI brain shows a significant inflammatory response and prominent gliosis. Neurological MPS I manifestations are evidenced by impaired performance in behavioral tests. Human neural and hematopoietic stem cells were found to readily engraft, with human cells detectable for at least 1 year posttransplantation. This new MPS I model is thus suitable for preclinical testing of novel pluripotent stem cell-based therapy approaches.

9.
Stem Cells Transl Med ; 3(11): 1275-86, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25273538

ABSTRACT

The autism spectrum disorders (ASDs) comprise a set of neurodevelopmental disorders that are, at best, poorly understood but are the fastest growing developmental disorders in the United States. Because animal models of polygenic disorders such as the ASDs are difficult to validate, the derivation of induced pluripotent stem cells (iPSCs) by somatic cell reprogramming offers an alternative strategy for identifying the cellular mechanisms contributing to ASDs and the development of new treatment options. Access to statistically relevant numbers of ASD patient cell lines, however, is still a limiting factor for the field. We describe a new resource with more than 200 cell lines (fibroblasts, iPSC clones, neural stem cells, glia) from unaffected volunteers and patients with a wide range of clinical ASD diagnoses, including fragile X syndrome. We have shown that both normal and ASD-specific iPSCs can be differentiated toward a neural stem cell phenotype and terminally differentiated into action-potential firing neurons and glia. The ability to evaluate and compare data from a number of different cell lines will facilitate greater insight into the cause or causes and biology of the ASDs and will be extremely useful for uncovering new therapeutic and diagnostic targets. Some drug treatments have already shown promise in reversing the neurobiological abnormalities in iPSC-based models of ASD-associated diseases. The ASD Stem Cell Resource at the Children's Hospital of Orange County will continue expanding its collection and make all lines available on request with the goal of advancing the use of ASD patient cells as disease models by the scientific community.


Subject(s)
Child Development Disorders, Pervasive , Induced Pluripotent Stem Cells , Models, Biological , Multifactorial Inheritance , Tissue Banks , Action Potentials/genetics , Adolescent , Cell Differentiation/genetics , Cell Line , Child , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/metabolism , Child Development Disorders, Pervasive/pathology , Child, Preschool , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Stem Cells
10.
J Neurosci Res ; 91(10): 1247-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893392

ABSTRACT

Robust strategies for developing patient-specific, human, induced pluripotent stem cell (iPSC)-based therapies of the brain require an ability to derive large numbers of highly defined neural cells. Recent progress in iPSC culture techniques includes partial-to-complete elimination of feeder layers, use of defined media, and single-cell passaging. However, these techniques still require embryoid body formation or coculture for differentiation into neural stem cells (NSCs). In addition, none of the published methodologies has employed all of the advances in a single culture system. Here we describe a reliable method for long-term, single-cell passaging of PSCs using a feeder-free, defined culture system that produces confluent, adherent PSCs that can be differentiated into NSCs. To provide a basis for robust quality control, we have devised a system of cellular nomenclature that describes an accurate genotype and phenotype of the cells at specific stages in the process. We demonstrate that this protocol allows for the efficient, large-scale, cGMP-compliant production of transplantable NSCs from all lines tested. We also show that NSCs generated from iPSCs produced with the process described are capable of forming both glia defined by their expression of S100ß and neurons that fire repetitive action potentials.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Cell Differentiation/physiology , Flow Cytometry , Humans , Immunohistochemistry , Neurons/cytology , Neurons/physiology , Neurons/transplantation , Patch-Clamp Techniques
11.
Cancer Cell Int ; 12(1): 41, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22995409

ABSTRACT

BACKGROUND: The cancer stem cell (CSC) hypothesis posits that deregulated neural stem cells (NSCs) form the basis of brain tumors such as glioblastoma multiforme (GBM). GBM, however, usually forms in the cerebral white matter while normal NSCs reside in subventricular and hippocampal regions. We attempted to characterize CSCs from a rare form of glioblastoma multiforme involving the neurogenic ventricular wall. METHODS: We described isolating CSCs from a GBM involving the lateral ventricles and characterized these cells with in vitro molecular biomarker profiling, cellular behavior, ex vivo and in vivo techniques. RESULTS: The patient's MRI revealed a heterogeneous mass with associated edema, involving the left subventricular zone. Histological examination of the tumor established it as being a high-grade glial neoplasm, characterized by polygonal and fusiform cells with marked nuclear atypia, amphophilic cytoplasm, prominent nucleoli, frequent mitotic figures, irregular zones of necrosis and vascular hyperplasia. Recurrence of the tumor occurred shortly after the surgical resection. CD133-positive cells, isolated from the tumor, expressed stem cell markers including nestin, CD133, Ki67, Sox2, EFNB1, EFNB2, EFNB3, Cav-1, Musashi, Nucleostemin, Notch 2, Notch 4, and Pax6. Biomarkers expressed in differentiated cells included Cathepsin L, Cathepsin B, Mucin18, Mucin24, c-Myc, NSE, and TIMP1. Expression of unique cancer-related transcripts in these CD133-positive cells, such as caveolin-1 and -2, do not appear to have been previously reported in the literature. Ex vivo organotypic brain slice co-culture showed that the CD133+ cells behaved like tumor cells. The CD133-positive cells also induced tumor formation when they were stereotactically transplanted into the brains of the immune-deficient NOD/SCID mice. CONCLUSIONS: This brain tumor involving the neurogenic lateral ventricular wall was comprised of tumor-forming, CD133-positive cancer stem cells, which are likely the driving force for the rapid recurrence of the tumor in the patient.

12.
Methods Mol Biol ; 767: 107-23, 2011.
Article in English | MEDLINE | ID: mdl-21822870

ABSTRACT

Culturing human embryonic stem cells (hESCs) requires a significant commitment of time and resources. It takes weeks to establish a culture, and the cultures require daily attention. Once hESC cultures are established, they can, with skill and the methods described, be kept in continuous culture for many years. hESC lines were originally derived using very similar culture medium and conditions as those developed for the derivation and culture of mouse ESC lines. However, these methods were suboptimal for hESCs and have evolved considerably in the years since the first hESC lines were derived. Compared with mouse ESCs, hESCs are very difficult to culture - they grow slowly, and most importantly, since we have no equivalent assays for germline competence, we cannot assume that the cells that we have in our culture dishes are either stable or pluripotent. This makes it far more critical to assay the cells frequently using the characterization methods, such as karyotyping, immunocytochemistry, gene expression analysis, and flow cytometry, provided in this manual.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Animals , Cell Count , Cells, Cultured , Collagenases/metabolism , Colony-Forming Units Assay , Cryopreservation , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Humans , Mice , Microscopy, Phase-Contrast , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
13.
Methods Mol Biol ; 767: 137-46, 2011.
Article in English | MEDLINE | ID: mdl-21822872

ABSTRACT

This protocol describes the culture of human pluripotent stem cells (PSCs) under feeder-free conditions in a commercially available, chemically defined, growth medium, using Matrigel as a substrate and the enzyme solution Accutase for single-cell passaging. This system is strikingly different from traditional PSC culture, where the cells are co-cultured with feeder cells and in medium containing serum replacement. PSCs cultured in this new system have a different morphology than those cultured on feeder cells but retain their characteristic pluripotency. This feeder-free PSC culture system is conceptually similar to feeder-free systems that use mouse embryonic fibroblast (MEF)-conditioned medium (MEF-CM) and Matrigel substratum. Instead of MEF-CM, a very complex and undefined medium, this new system uses StemPro SFM, a chemically defined medium that permits enzymatic passaging with Accutase to disaggregate the colonies into single cells. Accutase passaging has been used in conjunction with Stempro in our hands for 20+ passages without detectable karyotypic abnormalities. We will also review techniques for adapting cultures previously grown on MEFs, routine passaging of the cells, and cryopreservation.


Subject(s)
Cell Culture Techniques/methods , Culture Media/pharmacology , Enzymes/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Proliferation/drug effects , Cell Shape/drug effects , Coculture Techniques , Collagen/pharmacology , Cryopreservation , Drug Combinations , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Humans , Laminin/pharmacology , Mice , Pluripotent Stem Cells/drug effects , Proteoglycans/pharmacology
14.
Methods Mol Biol ; 767: 391-8, 2011.
Article in English | MEDLINE | ID: mdl-21822890

ABSTRACT

Embryoid body (EB) formation is a traditional method of inducing differentiation of pluripotent stem cells (PSCs). It is a routine in vitro test of pluripotency as well as the first stage in many differentiation protocols targeted toward the production of a specific lineage or cellular population, as in neural differentiation (see Chapters 29 and 30). The induction of differentiation via EB formation is fairly straightforward. However, depending on the specific PSC culture conditions - substrate, feeders, medium, and eventual cell type of interest - various methods are applied in order to most routinely obtain healthy EB cultures.


Subject(s)
Cell Culture Techniques/methods , Embryoid Bodies/cytology , Feeder Cells/cytology , Pluripotent Stem Cells/cytology , Cell Adhesion , Cell Aggregation , Centrifugation , Embryoid Bodies/metabolism , Feeder Cells/metabolism , Humans , Pluripotent Stem Cells/metabolism
15.
Methods ; 45(2): 142-58, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18593611

ABSTRACT

Human pluripotent stem cells have the unique properties of being able to proliferate indefinitely in their undifferentiated state and to differentiate into any somatic cell type. These cells are thus posited to be extremely useful for furthering our understanding of both normal and abnormal human development, providing a human cell preparation that can be used to screen for new reagents or therapeutic agents, and generating large numbers of differentiated cells that can be used for transplantation purposes. Critical among the applications for the latter are diseases and injuries of the nervous system, medical approaches to which have been, to date, primarily palliative in nature. Differentiation of human pluripotent stem cells into cells of the neural lineage, therefore, has become a central focus of a number of laboratories. This has resulted in the description in the literature of several dozen methods for neural cell differentiation from human pluripotent stem cells. Among these are methods for the generation of such divergent neural cells as dopaminergic neurons, retinal neurons, ventral motoneurons, and oligodendroglial progenitors. In this review, we attempt to fully describe most of these methods, breaking them down into five basic subdivisions: (1) starting material, (2) induction of loss of pluripotency, (3) neural induction, (4) neural maintenance and expansion, and (5) neuronal/glial differentiation. We also show data supporting the concept that undifferentiated human pluripotent stem cells appear to have an innate neural differentiation potential. In addition, we evaluate data comparing and contrasting neural stem cells differentiated from human pluripotent stem cells with those derived directly from the human brain.


Subject(s)
Cell Differentiation/physiology , Neurons/cytology , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Lineage/physiology , Cell Proliferation , Cell Separation/methods , Culture Media , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Neurons/metabolism , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...