Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 247
Filter
1.
Elife ; 122024 Sep 17.
Article in English | MEDLINE | ID: mdl-39287504

ABSTRACT

The integrated stress response (ISR) is a conserved pathway in eukaryotic cells that is activated in response to multiple sources of cellular stress. Although acute activation of this pathway restores cellular homeostasis, intense or prolonged ISR activation perturbs cell function and may contribute to neurodegeneration. DNL343 is an investigational CNS-penetrant small-molecule ISR inhibitor designed to activate the eukaryotic initiation factor 2B (eIF2B) and suppress aberrant ISR activation. DNL343 reduced CNS ISR activity and neurodegeneration in a dose-dependent manner in two established in vivo models - the optic nerve crush injury and an eIF2B loss of function (LOF) mutant - demonstrating neuroprotection in both and preventing motor dysfunction in the LOF mutant mouse. Treatment with DNL343 at a late stage of disease in the LOF model reversed elevation in plasma biomarkers of neuroinflammation and neurodegeneration and prevented premature mortality. Several proteins and metabolites that are dysregulated in the LOF mouse brains were normalized by DNL343 treatment, and this response is detectable in human biofluids. Several of these biomarkers show differential levels in CSF and plasma from patients with vanishing white matter disease (VWMD), a neurodegenerative disease that is driven by eIF2B LOF and chronic ISR activation, supporting their potential translational relevance. This study demonstrates that DNL343 is a brain-penetrant ISR inhibitor capable of attenuating neurodegeneration in mouse models and identifies several biomarker candidates that may be used to assess treatment responses in the clinic.


Subject(s)
Eukaryotic Initiation Factor-2B , Animals , Mice , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-2B/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/prevention & control , Stress, Physiological/drug effects , Disease Models, Animal , Male , Humans , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Female , Acetamides , Cyclohexylamines
2.
Neuroradiology ; 66(9): 1553-1564, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38886214

ABSTRACT

PURPOSE: To characterize Vanishing White Matter Disease (VWM) cases from a Brazilian University Tertiary hospital, focusing on brain magnetic resonance image (MRI) aspects, clinical and molecular data. METHODS: Medical records and brain MRI of 13 genetically confirmed VWM patients were reviewed. Epidemiological data such as age at symptom onset, gender and main symptoms were analyzed, along with genetic mutations and MRI characteristics, such as the distribution of white matter lesions and atrophy. RESULTS: The majority of patients were female, with the age of symptom onset ranging from 1 year and 6 months to 40 years. All mutations were identified in the EIF2B5 gene, the most prevalent being c.338G > A (p.Arg113His), and a novel mutation related to the disease was discovered, c.1051G > A (p.Gly351Ser). Trauma or infection were significant triggers. The most frequent symptoms were ataxia and limb spasticity. All MRI scans displayed deep white matter involvement, cystic degeneration, with U-fibers relatively spared and a predilection for the frontoparietal region. Lesions in the corpus callosum and posterior fossa were present in all patients. Follow-up exams revealed the evolution of white matter lesions and cerebral atrophy, which correlated with clinical deterioration. CONCLUSIONS: VWM affects various age groups, with a significant clinical and genetic variability. A novel mutation associated with the disease is highlighted. MRI reveals a typical pattern of white matter involvement, characterized by diffuse lesions in the periventricular and deep regions, with subsequent extension to the subcortical areas, accompanied by cystic degeneration, and plays a crucial role in diagnosis and follow-up.


Subject(s)
Leukoencephalopathies , Magnetic Resonance Imaging , Humans , Female , Male , Brazil , Adult , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Child , Adolescent , Magnetic Resonance Imaging/methods , Infant , Child, Preschool , Mutation , Young Adult , Eukaryotic Initiation Factor-2B/genetics
3.
Biosci Rep ; 44(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38873976

ABSTRACT

The heterotrimeric eIF2 complex consists of a core eIF2γ subunit to which binds eIF2α and eIF2ß subunits and plays an important role in delivering the Met-tRNAiMet to the 40S ribosome and start codon selection. The intricacies of eIF2ß-γ interaction in promoting Met-tRNAiMet binding are not clearly understood. Previously, the zinc-binding domain (ZBD) eIF2ßS264Y mutation was reported to cause Met-tRNAiMet binding defect due to the intrinsic GTPase activity. We showed that the eIF2ßS264Y mutation has eIF2ß-γ interaction defect. Consistently, the eIF2ßT238A intragenic suppressor mutation restored the eIF2ß-γ and Met-tRNAiMet binding. The eIF2ß-ZBD residues Asn252Asp and Arg253Ala mutation caused Met-tRNAiMet binding defect that was partially rescued by the eIF2ßT238A mutation, suggesting the eIF2ß-ZBD modulates Met-tRNAiMet binding. The suppressor mutation rescued the translation initiation fidelity defect of the eIF2γN135D SW-I mutation and eIF2ßF217A/Q221A double mutation in the HTH domain. The eIF2ßT238A suppressor mutation could not rescue the eIF2ß binding defect of the eIF2γV281K mutation; however, combining the eIF2ßS264Y mutation with the eIF2γV281K mutation was lethal. In addition to the previously known interaction of eIF2ß with the eIF2γ subunit via its α1-helix, the eIF2ß-ZBD also interacts with the eIF2γ subunit via guanine nucleotide-binding interface; thus, the eIF2ß-γ interacts via two distinct binding sites.


Subject(s)
Protein Binding , Binding Sites , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/chemistry , Mutation , RNA, Transfer, Met/metabolism , RNA, Transfer, Met/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Zinc/metabolism
4.
Ideggyogy Sz ; 77(5-6): 207-211, 2024 May 30.
Article in Hungarian | MEDLINE | ID: mdl-38829246

ABSTRACT

Background - Leukodystrophies, a hete­ro­­ge­neous group of brain and spinal cord dis­orders, often pose challenges in es­tab­li­shing molecular etiology. Vanishing White Matter Disease (VWMD) is a rare sub­type of leu­ko­dys­trophies presenting with characteristic clinical and MRI features, ne­ver­theless, achieving diag­nostic certainty requires genetic studies.

Case presentation - Our patient is a nine year old girl, who developed progressive gait difficulties at around 3-4 years of age. Her brain MRI showed confluent lesions with in­­creased signal intensity in the cerebral and cerebellar white matter on T2/FLAIR se­quen­ces, within which hypointense regions ap­peared with signal intensity resembling that of the cerebrospinal fluid on T1 sequences. Whole exome sequencing identified a homozygous likely pathogenic variant within the EIF2B5 gene in the proband, which was present in a heterozygous state in both asymptomatic parents. Having the clinical and molecular genetic diagnosis established, we explored therapeutic possibilities for the patient.

Conclusion - VWMD is a severe form of leukodystrophies with little or no disease modifying therapy available until recently. A better understanding of its molecular pathogenesis offers some hope for new inventive therapies. 

.


Subject(s)
Eukaryotic Initiation Factor-2B , Leukoencephalopathies , Mutation , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , Female , Eukaryotic Initiation Factor-2B/genetics , Child , Magnetic Resonance Imaging , White Matter/diagnostic imaging , White Matter/pathology
5.
BMC Neurol ; 24(1): 201, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872124

ABSTRACT

BACKGROUND: Leukoencephalopathy with vanishing white matter (VWM) is an autosomal recessive disorder affecting the white matter of the brain. It typically manifests during childhood, with clinical features including sudden and severe neurological deterioration triggered by stressors such as febrile illness, minor head trauma, or stressful events. Adult-onset cases of VWM are exceptionally uncommon. CASE PRESENTATION: In this case, we present an adult patient who exhibited late-onset progressive VWM characterized by ataxia, postural instability, cognitive impairment, and emotional disturbances. Comprehensive screening for endocrine, metabolic, tumor, and immunologic disorders yielded normal or negative results. Brain imaging revealed diffuse and confluent hyperintensity in the white matter on T2-weighted images, along with periventricular cavitations. Genetic testing confirmed the diagnosis of VWM, identifying two heterozygous variants in the eukaryotic translation initiation factor 2B subunit γ (EIF2B3) gene: a pathogenic variant, c.1037 T > C (p.I346T), and a variant of undetermined significance, c.22A > T (p.M8L). Upon a 2-year follow-up, the patient's symptoms deteriorated rapidly following a COVID-19 infection. CONCLUSIONS: In conclusion, we have presented a case of classical adult-onset VWM. Since there are no cures or definitive treatments for the disease, it's extremely important to focus on early diagnosis and the prevention of stressors to avoid acute deterioration.


Subject(s)
Eukaryotic Initiation Factor-2B , Leukoencephalopathies , Humans , Eukaryotic Initiation Factor-2B/genetics , Leukoencephalopathies/genetics , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , White Matter/diagnostic imaging , White Matter/pathology , Male , Female , COVID-19/genetics , COVID-19/complications , Heterozygote , Middle Aged
6.
Cell Mol Life Sci ; 81(1): 234, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789799

ABSTRACT

Vanishing white matter (VWM) is a leukodystrophy caused by biallelic pathogenic variants in eukaryotic translation initiation factor 2B. To date, it remains unclear which factors contribute to VWM pathogenesis. Here, we investigated the basis of VWM pathogenesis using the 2b5ho mouse model. We first mapped the temporal proteome in the cerebellum, corpus callosum, cortex, and brainstem of 2b5ho and wild-type (WT) mice. Protein changes observed in 2b5ho mice were then cross-referenced with published proteomic datasets from VWM patient brain tissue to define alterations relevant to the human disease. By comparing 2b5ho mice with their region- and age-matched WT counterparts, we showed that the proteome in the cerebellum and cortex of 2b5ho mice was already dysregulated prior to pathology development, whereas proteome changes in the corpus callosum only occurred after pathology onset. Remarkably, protein changes in the brainstem were transient, indicating that a compensatory mechanism might occur in this region. Importantly, 2b5ho mouse brain proteome changes reflect features well-known in VWM. Comparison of the 2b5ho mouse and VWM patient brain proteomes revealed shared changes. These could represent changes that contribute to the disease or even drive its progression in patients. Taken together, we show that the 2b5ho mouse brain proteome is affected in a region- and time-dependent manner. We found that the 2b5ho mouse model partly replicates the human disease at the protein level, providing a resource to study aspects of VWM pathogenesis by highlighting alterations from early to late disease stages, and those that possibly drive disease progression.


Subject(s)
Disease Models, Animal , Leukoencephalopathies , Proteome , Proteomics , White Matter , Animals , Mice , Humans , Proteome/metabolism , Leukoencephalopathies/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , White Matter/metabolism , White Matter/pathology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-2B/genetics , Brain/metabolism , Brain/pathology , Mice, Inbred C57BL , Cerebellum/metabolism , Cerebellum/pathology
7.
J Med Chem ; 67(7): 5758-5782, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38511649

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a key component of the integrated stress response (ISR), which regulates protein synthesis and stress granule formation in response to cellular insult. Modulation of the ISR has been proposed as a therapeutic strategy for treatment of neurodegenerative diseases such as vanishing white matter (VWM) disease and amyotrophic lateral sclerosis (ALS) based on its ability to improve cellular homeostasis and prevent neuronal degeneration. Herein, we report the small-molecule discovery campaign that identified potent, selective, and CNS-penetrant eIF2B activators using both structure- and ligand-based drug design. These discovery efforts culminated in the identification of DNL343, which demonstrated a desirable preclinical drug profile, including a long half-life and high oral bioavailability across preclinical species. DNL343 was progressed into clinical studies and is currently undergoing evaluation in late-stage clinical trials for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Leukoencephalopathies , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Mutation , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Brain/metabolism , Leukoencephalopathies/metabolism
8.
Mol Ther ; 32(6): 1701-1720, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38549375

ABSTRACT

Leukoencephalopathy with vanishing white matter (VWM) is a progressive incurable white matter disease that most commonly occurs in childhood and presents with ataxia, spasticity, neurological degeneration, seizures, and premature death. A distinctive feature is episodes of rapid neurological deterioration provoked by stressors such as infection, seizures, or trauma. VWM is caused by autosomal recessive mutations in one of five genes that encode the eukaryotic initiation factor 2B complex, which is necessary for protein translation and regulation of the integrated stress response. The majority of mutations are in EIF2B5. Astrocytic dysfunction is central to pathophysiology, thereby constituting a potential therapeutic target. Herein we characterize two VWM murine models and investigate astrocyte-targeted adeno-associated virus serotype 9 (AAV9)-mediated EIF2B5 gene supplementation therapy as a therapeutic option for VWM. Our results demonstrate significant rescue in body weight, motor function, gait normalization, life extension, and finally, evidence that gene supplementation attenuates demyelination. Last, the greatest rescue results from a vector using a modified glial fibrillary acidic protein (GFAP) promoter-AAV9-gfaABC(1)D-EIF2B5-thereby supporting that astrocytic targeting is critical for disease correction. In conclusion, we demonstrate safety and early efficacy through treatment with a translatable astrocyte-targeted gene supplementation therapy for a disease that has no cure.


Subject(s)
Astrocytes , Dependovirus , Disease Models, Animal , Eukaryotic Initiation Factor-2B , Genetic Therapy , Genetic Vectors , Leukoencephalopathies , Animals , Dependovirus/genetics , Mice , Leukoencephalopathies/therapy , Leukoencephalopathies/genetics , Leukoencephalopathies/etiology , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Astrocytes/metabolism , Astrocytes/pathology , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Humans
9.
Mol Ther ; 32(5): 1328-1343, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38454603

ABSTRACT

Vanishing white matter (VWM) is a fatal leukodystrophy caused by recessive mutations in subunits of the eukaryotic translation initiation factor 2B. Currently, there are no effective therapies for VWM. Here, we assessed the potential of adenine base editing to correct human pathogenic VWM variants in mouse models. Using adeno-associated viral vectors, we delivered intein-split adenine base editors into the cerebral ventricles of newborn VWM mice, resulting in 45.9% ± 5.9% correction of the Eif2b5R191H variant in the cortex. Treatment slightly increased mature astrocyte populations and partially recovered the integrated stress response (ISR) in female VWM animals. This led to notable improvements in bodyweight and grip strength in females; however, locomotor disabilities were not rescued. Further molecular analyses suggest that more precise editing (i.e., lower rates of bystander editing) as well as more efficient delivery of the base editors to deep brain regions and oligodendrocytes would have been required for a broader phenotypic rescue. Our study emphasizes the potential, but also identifies limitations, of current in vivo base-editing approaches for the treatment of VWM or other leukodystrophies.


Subject(s)
Dependovirus , Disease Models, Animal , Eukaryotic Initiation Factor-2B , Gene Editing , Leukoencephalopathies , Phenotype , Animals , Mice , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/therapy , Leukoencephalopathies/pathology , Dependovirus/genetics , Humans , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Female , Mutation , Genetic Therapy/methods , White Matter/pathology , White Matter/metabolism , Astrocytes/metabolism
10.
CNS Neurosci Ther ; 30(2): e14600, 2024 02.
Article in English | MEDLINE | ID: mdl-38357857

ABSTRACT

AIM: Characterize Growth Differentiation Factor 15 (GDF15) as a secreted biomarker of the integrated stress response (ISR) within the central nervous system (CNS). METHODS: We determined GDF15 levels utilizing in vitro and in vivo neuronal systems wherein the ISR was activated. Primarily, we used the murine model of vanishing white matter disease (VWMD), a neurological disease driven by persistent ISR in the CNS, to establish a link between levels of GDF15 in the cerebrospinal fluid (CSF) and ISR gene expression signature in the CNS. GDF15 was also determined in the CSF of VWM patients. RESULTS: GDF15 expression was increased concomitant to ISR activation in stress-induced primary astrocytes as well as in retinal ganglion cells following optic nerve crush, while treatment with 2Bact, a specific eIF2B activator, suppressed both the ISR and GDF15. In the VWMD model, CSF GDF15 levels corresponded with the magnitude of the ISR and were reduced by 2BAct. In VWM patients, mean CSF GDF15 was elevated >20-fold as compared to healthy controls, whereas plasma GDF15 was undifferentiated. CONCLUSIONS: These data suggest that CSF GDF15 is a dynamic marker of ISR activation in the CNS and may serve as a pharmacodynamic biomarker for ISR-modulating therapies.


Subject(s)
Growth Differentiation Factor 15 , Leukoencephalopathies , Humans , Mice , Animals , Growth Differentiation Factor 15/genetics , Leukoencephalopathies/genetics , Central Nervous System/metabolism , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Biomarkers
11.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38171595

ABSTRACT

The leukodystrophy vanishing white matter (VWM) is characterized by chronic and episodic acute neurological deterioration. Curative treatment is presently unavailable. Pathogenic variants in the genes encoding eukaryotic initiation factor 2B (eIF2B) cause VWM and deregulate the integrated stress response (ISR). Previous studies in VWM mouse models showed that several ISR-targeting compounds ameliorate clinical and neuropathological disease hallmarks. It is unclear which ISR components are suitable therapeutic targets. In this study, effects of 4-phenylbutyric acid, tauroursodeoxycholic acid, or pridopidine (PDPD), with ISR targets upstream or downstream of eIF2B, were assessed in VWM mice. In addition, it was found that the composite ataxia score represented motor decline of VWM mice more accurately than the previously used neuroscore. 4-phenylbutyric acid and tauroursodeoxycholic acid did not improve VWM disease hallmarks, whereas PDPD had subtle beneficial effects on motor skills. PDPD alone does not suffice as treatment in VWM mice but may be considered for combination therapy. Also, treatments aimed at ISR components upstream of eIF2B do not improve chronic neurological deterioration; effects on acute episodic decline remain to be investigated.


Subject(s)
Eukaryotic Initiation Factor-2B , White Matter , Mice , Animals , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , White Matter/pathology , Motor Skills , Disease Models, Animal
12.
J Genet ; 1022023.
Article in English | MEDLINE | ID: mdl-37674283

ABSTRACT

Vanishing of white matter (VWM) is a hereditary heterogeneous brain disorder that most often affects children. However, the onset of the disease varies from childhood to adulthood. VWM is caused by mutations in one of the five genes encoding subunits of the eukaryotic initiation factor eIF2B. In the current study, we aimed to determine the genetic cause of VWM in a large consanguineous Iranian family with three affected members. Next-generation sequencing was conducted on the proband to determine the underlying cause of VWM. The identified variant was validated by PCR-Sanger sequencing in the patient and was also segregated in his parents and two other affected members of the pedigree. The potential functional effects of this mutation within EIF2B5 were predicted by in silico analysis. We have also reviewed all EIF2B5 disease-causing variants and available clinical features of each patient reported in HGMD Professional 2022.2. A novel homozygous variant c.746T>G [p.Ile249Ser] was detected in EIF2B5 which was co-segregated with the disease in all affected family members in an autosomal recessive manner. All employed in silico prediction tools and 3D structure analysis for the novel mutation also supported the pathogenicity of this variant. Our study not only expanded the spectrum of the pathogenic variants in EIF2B5 but also presented a literature review on EIF2B5-related conditions that provide a comprehensive picture of the genetic nature of this gene and phenotypic variability in patients.


Subject(s)
Leukoencephalopathies , Child , Humans , Adolescent , Young Adult , Iran , Consanguinity , Leukoencephalopathies/genetics , Mutation, Missense , Mutation , Eukaryotic Initiation Factor-2B/genetics
13.
J Neurol ; 270(9): 4219-4234, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37171481

ABSTRACT

BACKGROUND: Leukodystrophy with vanishing white matter (LVWM) is an autosomal recessive disease with typical pediatric-onset caused by mutations in one of the five EIF2B genes. Adult-onset (AO) cases are rare. METHODS: In this observational study, we reviewed clinical and laboratory information of the patients with AO-LVWM assessed at two referral centers in Italy and Portugal from Jan-2007 to Dec-2019. RESULTS: We identified 18 patients (13 females) with AO-LVWM caused by EIF2B5 or EIF2B3 mutations. Age of neurological onset ranged from 16 to 60 years, with follow-ups occurring from 2 to 37 years. Crucial symptoms were cognitive and motor decline. In three patients, stroke-like events were the first manifestation; in another, bladder dysfunction remained the main complaint across decades. Brain MRI showed white matter (WM) rarefaction in all cases, except two. Diffusion-weighted imaging documented focal hyperintensity in the acute stage of stroke-like events. 1H-spectroscopy primarily showed N-acetyl-aspartate reduction; 18fluorodeoxyglucose-PET revealed predominant frontoparietal hypometabolism; evoked potential studies demonstrated normal-to-reduced amplitudes; neuro-ophthalmological assessment showed neuroretinal thinning, and b-wave reduction on full-field electroretinogram. Interestingly, we found an additional patient with LVWM-compatible phenotype and monoallelic variants in two distinct eIF2B genes, EIF2B1 and EIF2B2. CONCLUSIONS: AO-LVWM presents varying clinical manifestations at onset, including stroke-like events. WM rarefaction is the most consistent diagnostic clue even in the latest onset cases. Spectroscopy and electrophysiological features are compatible with axon, rather than myelin, damage. Cerebral glucose metabolic abnormalities and retinal alterations can be present. LVWM might also be caused by a digenic inheritance affecting the eIF2B complex.


Subject(s)
Demyelinating Diseases , Leukoencephalopathies , Lysosomal Storage Diseases , Neurodegenerative Diseases , Stroke , White Matter , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult , Eukaryotic Initiation Factor-2B/genetics , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Magnetic Resonance Imaging , Mutation/genetics , Observational Studies as Topic , White Matter/diagnostic imaging
14.
Proc Natl Acad Sci U S A ; 120(15): e2207898120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37014850

ABSTRACT

Breast cancer (BC) metastasis involves cancer stem cells (CSCs) and their regulation by micro-RNAs (miRs), but miR targeting of the translation machinery in CSCs is poorly explored. We therefore screened miR expression levels in a range of BC cell lines, comparing non-CSCs to CSCs, and focused on miRs that target translation and protein synthesis factors. We describe a unique translation regulatory axis enacted by reduced expression of miR-183 in breast CSCs, which we show targets the eIF2Bδ subunit of guanine nucleotide exchange factor eIF2B, a regulator of protein synthesis and the integrated stress response (ISR) pathway. We report that reduced expression of miR-183 greatly increases eIF2Bδ protein levels, preventing strong induction of the ISR and eIF2α phosphorylation, by preferential interaction with P-eIF2α. eIF2Bδ overexpression is essential for BC cell invasion, metastasis, maintenance of metastases, and breast CSC expansion in animal models. Increased expression of eIF2Bδ, a site of action of the drug ISRIB that also prevents ISR signaling, is essential for breast CSC maintenance and metastatic capacity.


Subject(s)
MicroRNAs , Neoplasms , Animals , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Guanine Nucleotide Exchange Factors , Neoplastic Stem Cells/metabolism
15.
Antioxid Redox Signal ; 39(4-6): 351-373, 2023 08.
Article in English | MEDLINE | ID: mdl-36943285

ABSTRACT

Significance: Organisms adapt to changing environments by engaging cellular stress response pathways that serve to restore proteostasis and enhance survival. A primary adaptive mechanism is the integrated stress response (ISR), which features phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2). Four eIF2α kinases respond to different stresses, enabling cells to rapidly control translation to optimize management of resources and reprogram gene expression for stress adaptation. Phosphorylation of eIF2 blocks its guanine nucleotide exchange factor, eIF2B, thus lowering the levels of eIF2 bound to GTP that is required to deliver initiator transfer RNA (tRNA) to ribosomes. While bulk messenger RNA (mRNA) translation can be sharply lowered by heightened phosphorylation of eIF2α, there are other gene transcripts whose translation is unchanged or preferentially translated. Among the preferentially translated genes is ATF4, which directs transcription of adaptive genes in the ISR. Recent Advances and Critical Issues: This review focuses on how eIF2α kinases function as first responders of stress, the mechanisms by which eIF2α phosphorylation and other stress signals regulate the exchange activity of eIF2B, and the processes by which the ISR triggers differential mRNA translation. To illustrate the synergy between stress pathways, we describe the mechanisms and functional significance of communication between the ISR and another key regulator of translation, mammalian/mechanistic target of rapamycin complex 1 (mTORC1), during acute and chronic amino acid insufficiency. Finally, we discuss the pathological conditions that stem from aberrant regulation of the ISR, as well as therapeutic strategies targeting the ISR to alleviate disease. Future Directions: Important topics for future ISR research are strategies for modulating this stress pathway in disease conditions and drug development, molecular processes for differential translation and the coordinate regulation of GCN2 and other stress pathways during physiological and pathological conditions. Antioxid. Redox Signal. 39, 351-373.


Subject(s)
Eukaryotic Initiation Factor-2B , Eukaryotic Initiation Factor-2 , Animals , Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphorylation , Gene Expression Regulation , Stress, Physiological , Mammals/metabolism
16.
CNS Neurosci Ther ; 29(4): 1049-1066, 2023 04.
Article in English | MEDLINE | ID: mdl-36650674

ABSTRACT

INTRODUCTION: Leukoencephalopathy with vanishing white matter (VWM) is a rare autosomal recessive leukoencephalopathy resulting from mutations in EIF2B1-5, which encode subunits of eukaryotic translation initiation factor 2B (eIF2B). Studies have found that eIF2B mutation has a certain influence on embryonic brain development. So far, the effect of the eIF2B mutations on the dynamic process of brain development is not fully understood yet. AIMS: Three-dimensional brain organoid technology has promoted the study of human nervous system developmental diseases in recent years, providing a potential platform for elucidating the pathological mechanism of neurodevelopmental diseases. In this study, we aimed to investigate the effects of eIF2B mutation on the differentiation and development of different nerve cells during dynamic brain development process using 3D brain organoids. RESULTS: We constructed eIF2B mutant and wild-type brain organoid model with induced pluripotent stem cell (iPSC). Compared with the wild type, the mutant brain organoids were significantly smaller, accompanied by increase in apoptosis, which might be resulted from overactivation of unfolded protein response (UPR). Neuronal development was delayed in early stage, but with normal superficial neuronal differentiation in later stage. eIF2B mutations resulted in immature astrocytes with increased expression of GFAPδ, nestin, and αB-crystallin, and there were increased oligodendrocyte progenitor cells, decreased mature oligodendrocytes, and sparse myelin in mutant cerebral organoids in the later stage. CONCLUSION: we constructed the first eIF2B mutant cerebral organoids to explore the dynamic brain development process, which provides a platform for further research on the specific pathogenesis of VWM.


Subject(s)
Induced Pluripotent Stem Cells , Leukoencephalopathies , White Matter , Humans , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Brain/metabolism , White Matter/pathology , Mutation/genetics
17.
Curr Opin Immunol ; 78: 102251, 2022 10.
Article in English | MEDLINE | ID: mdl-36242870

ABSTRACT

The interferon-regulated kinase PKR (protein kinase RNA-activated) is a potent innate immune factor against a broad range of viruses. Being part of the integrated stress response (ISR), its restrictive effect is predominantly exerted by phosphorylating the eukaryotic translation-initiation factor eIF2, thereby turning it into an inhibitor of translation-initiation factor eIF2B. A plethora of viruses are known to evade the shutdown of cellular mRNA translation by interfering either with PKR activation or with eIF2 phosphorylation. Recently, a novel PKR evasion strategy was described: proteins from three taxonomically distinct RNA viruses allow for full PKR activation and eIF2 phosphorylation in the infected cell, but protect eIF2B from inhibition by phosphorylated eIF2, thus enabling mRNA translation in the presence of an activated ISR.


Subject(s)
Eukaryotic Initiation Factor-2B , Eukaryotic Initiation Factor-2 , Immunity, Innate , Virus Diseases , Humans , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Immunity, Innate/genetics , Immunity, Innate/physiology , Interferons , Protein Kinases/immunology , Protein Kinases/metabolism , RNA, Messenger , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
18.
Neurochem Res ; 47(12): 3747-3760, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36198922

ABSTRACT

Leukoencephalopathy with vanishing white matter (VWM) is an inherited leukoencephalopathy characterized by progressive rarefaction of cerebral white matter. Dysfunction of patient astrocyte plays a central role in the pathogenesis, while the immaturity of oligodendrocyte is probably secondary. How eIF2B mutant astrocytes affect the maturation and myelination of oligodendrocyte precursor cells (OPCs) is unclear yet. We used induced pluripotent stem cells (iPSCs) derived from our patient with EIF2B5 mutations to differentiate into astrocytes (AS) and OPCs, and aimed to verify that patient astrocytes inhibited the differentiation of OPCs by abnormalities of secreted proteins. eIF2B mutant astrocytes and astrocyte-conditioned medium (ACM) both inhibited the maturation of OPCs. It was revealed that 13 promising proteins exhibited a similar up- or downregulation by the PRM method correlated well with TMT results. eIF2B mutant astrocytes may secrete abnormal extracellular matrix (HA, LAMA4, BGN, FBN1, VASN, PCOLCE, MFAP4), cytokines (IL-6, CRABP1, ISG15), growth factors (PDGF-AA, CNTF, IGF-II, sFRP1, SERPINF1) and increased FABP7, which might lead to the differentiation and maturation disorder of OPCs. We analyzed the astrocyte-conditioned medium to find the key secretory molecules affecting the differentiation and maturation of OPCs, which provides potential clues for further research on the mechanism of VWM.


Subject(s)
Induced Pluripotent Stem Cells , Leukoencephalopathies , White Matter , Humans , Astrocytes/metabolism , Culture Media, Conditioned/pharmacology , Eukaryotic Initiation Factor-2B/genetics , Induced Pluripotent Stem Cells/pathology , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , White Matter/metabolism
19.
Ann Clin Transl Neurol ; 9(8): 1147-1162, 2022 08.
Article in English | MEDLINE | ID: mdl-35778832

ABSTRACT

OBJECTIVE: Vanishing white matter (VWM) is a leukodystrophy, characterized by stress-sensitive neurological deterioration and premature death. It is currently without curative treatment. It is caused by bi-allelic pathogenic variants in the genes encoding eukaryotic initiation factor 2B (eIF2B). eIF2B is essential for the regulation of the integrated stress response (ISR), a physiological response to cellular stress. Preclinical studies on VWM mouse models revealed that deregulated ISR is key in the pathophysiology of VWM and an effective treatment target. Guanabenz, an α2-adrenergic agonist, attenuates the ISR and has beneficial effects on VWM neuropathology. The current study aimed at elucidating guanabenz's disease-modifying potential and mechanism of action in VWM mice. Sephin1, an ISR-modulating guanabenz analog without α2-adrenergic agonistic properties, was included to separate effects on the ISR from α2-adrenergic effects. METHODS: Wild-type and VWM mice were subjected to placebo, guanabenz or sephin1 treatments. Effects on clinical signs, neuropathology, and ISR deregulation were determined. Guanabenz's and sephin1's ISR-modifying effects were tested in cultured cells that expressed or lacked the α2-adrenergic receptor. RESULTS: Guanabenz improved clinical signs, neuropathological hallmarks, and ISR regulation in VWM mice, but sephin1 did not. Guanabenz's effects on the ISR in VWM mice were not replicated in cell cultures and the contribution of α2-adrenergic effects on the deregulated ISR could therefore not be assessed. INTERPRETATION: Guanabenz proved itself as a viable treatment option for VWM. The exact mechanism through which guanabenz exerts its ameliorating impact on VWM requires further studies. Sephin1 is not simply a guanabenz replacement without α2-adrenergic effects.


Subject(s)
Guanabenz , White Matter , Adrenergic Agents , Animals , Eukaryotic Initiation Factor-2B/genetics , Guanabenz/analogs & derivatives , Guanabenz/pharmacology , Mice , White Matter/pathology
20.
Ital J Pediatr ; 48(1): 128, 2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35897042

ABSTRACT

BACKGROUND: Leukoencephalopathy with vanishing white matter (VWM) is an autosomal recessive neurological disease. The physiopathology of disease is still little understood, but it seems to involve impairment in maturation of astrocytes; as a consequence white matter is more prone to cellular stress. Disease is caused by mutations in five genes encoding subunits of the translation initiation factor eIF2B. We know five different types of VWM syndrome classified based different ages of onset (prenatal, infantile, childhood, juvenile and adult onset). CASE PRESENTATION: We report the case of a 4-month-old boy with early seizure onset, recurrent hypoglycemia and post mortem diagnosis of vanishing white matter disease (VMD). At the admission he presented suspected critical episodes, resolved after intravenous administration of benzodiazepines. The brain MRI showed total absence of myelination that suggested hypomyelination leukoencephalopathy. The whole exome sequencing (WES) revealed a variant of EIF2B2 gene (p. Val308Met) present in homozygosity. In this case report we also describe the clinical evolution of seizures, in fact the epileptic seizures had a polymorphic aspect, from several complex partial seizures secondarily generalized to status epilepticus. CONCLUSION: Infantile and early childhood onset forms are associated with chronic progressive neurological signs, with episodes of rapid neurological worsening, and poor prognosis, with death in few months or years. Clinical presentation of epilepsy is poorly documented and do not include detailed information about the type, time of onset and severity of seizures. No therapeutic strategies for VWM disease have been reported.


Subject(s)
Epilepsy , Leukoencephalopathies , White Matter , Child, Preschool , Eukaryotic Initiation Factor-2B/genetics , Humans , Infant , Leukoencephalopathies/diagnosis , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Magnetic Resonance Imaging , Male , Mutation , Seizures , White Matter/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL