Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1328748, 2024.
Article in English | MEDLINE | ID: mdl-38572474

ABSTRACT

Background: In observational studies, the relationship between coffee intake and bone mineral density (BMD) is contradictory. However, residual confounding tends to bias the results of these studies. Therefore, we used a two-sample Mendelian randomization (MR) approach to further investigate the potential causal relationship between the two. Methods: Genetic instrumental variables (IVs) associated with coffee intake were derived from genome-wide association studies (GWAS) of the Food Frequency Questionnaire (FFQ) in 428,860 British individuals and matched using phenotypes in PhenoScanner. Summarized data on BMD were obtained from 537,750 participants, including total body BMD (TB-BMD), TB-BMD in five age brackets ≥60, 45-60, 30-45, 15-30, and 0-15 years, and BMD in four body sites: the lumbar spine, the femoral neck, the heel, and the ultradistal forearm. We used inverse variance weighting (IVW) methods as the primary analytical method for causal inference. In addition, several sensitivity analyses (MR-Egger, Weighted median, MR-PRESSO, Cochran's Q test, and Leave-one-out test) were used to test the robustness of the results. Results: After Bonferroni correction, Coffee intake has a potential positive correlation with total body BMD (effect estimate [Beta]: 0.198, 95% confidence interval [Cl]: 0.05-0.35, P=0.008). In subgroup analyses, coffee intake was potentially positively associated with TB-BMD (45-60, 30-45 years) (Beta: 0.408, 95% Cl: 0.12-0.69, P=0.005; Beta: 0.486, 95% Cl: 0.12-0.85, P=0.010). In addition, a significant positive correlation with heel BMD was also observed (Beta: 0.173, 95% Cl: 0.08-0.27, P=0.002). The results of the sensitivity analysis were generally consistent. Conclusion: The results of the present study provide genetic evidence for the idea that coffee intake is beneficial for bone density. Further studies are needed to reveal the biological mechanisms and offer solid support for clinical guidelines on osteoporosis prevention.


Subject(s)
Bone Density , Coffee , Humans , Bone Density/genetics , Genome-Wide Association Study , Mendelian Randomization Analysis , Femur Neck
2.
Int Orthop ; 48(2): 547-553, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37782331

ABSTRACT

PURPOSE: The search for more effective and safe treatment methods for cervical spondylotic radiculopathy (CSR) has led to the rapid development and increasing popularity of minimally invasive posterior cervical foraminotomy (MI-PCF). This study aims to compare two important approaches for MI-PCF surgery: the channel-assisted cervical key hole technology combined with ultrasonic bone osteotome (CKH-UBO) and posterior percutaneous endoscopic cervical foraminotomy (PPECF). METHODS: Data from patients treated with single-level CKH-UBO (n = 35) or PPECF (n = 40) were analyzed. Clinical outcomes, including visual analogue scale (VAS) scores for neck and arm pain, Neck Disability Index (NDI), and modified Macnab criteria, were assessed preoperatively, as well as at three days, three months, and one year postoperatively. RESULTS: The percentages of patients with excellent and good outcomes were 97.14% and 92.5%, respectively. The average surgical time in the CKH-UBO group was significantly shorter than in the PPECF group (p < 0.001), while the average incision length in the PPECF group was significantly smaller than in the CKH-UBO group. There were no significant differences between the two groups in terms of blood loss, hospital stay, and clinical outcomes at three days, three months, and 12 months postoperatively. CONCLUSION: CKH-UBO can achieve the same surgical outcomes as PPECF for the treatment of CSR. However, CKH-UBO saves more time but requires patients to undergo larger incisions.


Subject(s)
Foraminotomy , Radiculopathy , Spondylosis , Humans , Foraminotomy/adverse effects , Foraminotomy/methods , Retrospective Studies , Ultrasonics , Treatment Outcome , Cervical Vertebrae/diagnostic imaging , Cervical Vertebrae/surgery , Spondylosis/surgery , Radiculopathy/surgery , Diskectomy/methods
3.
Front Immunol ; 14: 1291206, 2023.
Article in English | MEDLINE | ID: mdl-38173728

ABSTRACT

Background: Observational studies have shown that changes in circulating cytokine/growth factor levels occur throughout the initiation and progression of ankylosing spondylitis (AS), yet whether they are etiologic or downstream effects remains unclear. In this study, we performed a summarized-level bidirectional Mendelian randomization (MR) analysis to shed light on the causal relationship between the two. Methods: Genetic instrumental-variables (IVs) associated with circulating cytokine/growth factor levels were derived from a genome-wide association study (GWAS) of 8,293 European individuals, whereas summary data for the AS were obtained from a FinnGen GWAS of 166,144 participants. We used the inverse-variance-weighted (IVW) method as the main analysis for causal inference. Furthermore, several sensitivity analyses (MR-Egger, weighted median, MR-PRESSO and Cochran's Q test) were utilized to examine the robustness of the results. Finally, reverse MR analysis was performed to assess reverse causality between AS and circulating cytokine/growth factor levels. Results: After Bonferroni correction, circulating levels of Cutaneous T-cell attracting (CTACK) and Monocyte specific chemokine 3 (MCP-3) were positively associated with a higher risk of AS (odds ratio [OR]: 1.224, 95% confidence interval [95% Cl]: 1.022 ~ 1.468, P = 0.028; OR: 1.250, 95% Cl: 1.016 ~ 1.539, P = 0.035). In addition, elevated circulating levels of Basic fibroblast growth factor (FGF-basic), Granulocyte colony-stimulating factor (G-CSF) and MCP-3 was considered a consequence of AS disease (ß = 0.023, P = 0.017; ß = 0.017, P = 0.025; ß = 0.053, P = 0.025). The results of the sensitivity analysis were generally consistent. Conclusion: The present study supplies genetic evidence for the relationship between circulating cytokine levels and AS. Targeted interventions of specific cytokines may help to reduce the risk of AS initiation and progression.


Subject(s)
Cytokines , Spondylitis, Ankylosing , Humans , Genome-Wide Association Study , Mendelian Randomization Analysis , Spondylitis, Ankylosing/genetics , Granulocyte Colony-Stimulating Factor
4.
Front Neurol ; 13: 998173, 2022.
Article in English | MEDLINE | ID: mdl-36299275

ABSTRACT

Background: The purpose of this study was to compare the safety and efficacy of unilateral vs. bilateral pedicle screw fixation (BPSF) for lumbar degenerative diseases. Methods: Electronic databases including PubMed, Web of science, the Cochrane Library, Scopus, MEDLINE, EMBASE, EBSCO were searched by computer. The deadline was set for June 1, 2022. This study included all high-quality randomized controlled trials (RCTs), prospective clinical controlled studies (PRO), and retrospective studies (Retro) that compared unilateral and bilateral pedicle screw fixation in the treatment of lumbar degenerative diseases. Revman5.3 software was used for meta-analysis after two researchers independently screened the literature, extracted data, and assessed the risk of bias in the study. Results: Fourteen studies with a total of 1,086 patients were included. Compared with BPSF, unilateral pedicle screw fixation (UPSF) has shorter operation time and hospital time, and less blood loss and operation cost, operation time [SMD = -1.75, 95% CI (-2.46 to -1.03), P < 0.00001], hospital time [SMD = -1.10, 95% CI (-1.97 to -0.22), P = 0.01], Blood loss [SMD = -1.62, 95% CI (-2.42 to -0.82), P < 0.0001], operation cost [SMD = -14.03, 95% CI (-20.08 to -7.98), P < 0.00001], the ODI after bilateral pedicle screw fixation was lower, and the degree of lumbar dysfunction was lighter, [SMD = 0.19, 95% CI (0.05-0.33), P = 0.007], better fusion effect, fusion rate [RR=0.95, 95% CI (0.91-1.00), P = 0.04]. VAS-Low back pain [SMD = 0.07, 95% CI (-0.07-0.20), P = 0.35], VAS-Leg pain [SMD = 0.18, 95% CI (-0.00-0.36), P = 0.05], SF-36 [SMD = 0.00, 95% CI (-0.30-0.30), P = 1.00], complications rate [RR = 0.94, 95% CI (0.9154-1.63), P = 0.82], the overall difference was not statistically significant. Conclusions: Currently limited evidence suggests that UPSF significantly reduces blood loss, significantly shortens the operative time and hospital stay, and reduces blood loss and costs. After BPSF, the ODI was lower, the degree of lumbar spine dysfunction was lower, and the fusion rate was significantly higher. The VAS, SF-36, and complications scores of the two groups were comparable, and there was no significant clinical difference.

5.
Exp Ther Med ; 15(3): 2961-2967, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29599833

ABSTRACT

Post-menopausal osteoporosis is one of the most common bone diseases in women. The aim of the present study was to predict the diagnostic function modules from a differential co-expression gene network in order to enhance the current understanding of the biological processes and to promote the early prevention and intervention of post-menopausal osteoporosis. The diagnostic function modules were extracted from a differential co-expression network by the established protein-protein interaction (PPI) network analysis. First, significant genes were identified from the differential co-expression network, which were regarded as seed genes. Starting from the seed genes, the sub-networks in this disease, referred to as diagnostic function modules, were exhaustively searched and prioritized through a snowball sampling strategy to identify genes to accurately predict clinical outcomes. In addition, crucial function inference was performed for each diagnostic function module. Based on the microarray and PPI data, the differential co-expression network was constructed, which contained 1,607 genes and 4,197 interactions. A total of 110 seed genes were identified, and nine diagnostic modules that accurately distinguished post-menopausal osteoporosis from healthy controls were screened out from these seed genes. The diagnostic modules may be associated with five functional pathways with emphasis on metabolism. A total of nine diagnostic functional modules screened in the present study may be considered as potential targets for predicting the clinical outcomes of post-menopausal osteoporosis, and may contribute to the early diagnosis and therapy of osteoporosis.

6.
PLoS Genet ; 12(5): e1006083, 2016 05.
Article in English | MEDLINE | ID: mdl-27203582

ABSTRACT

The Huntington's disease (HD) protein, huntingtin (HTT), is a large protein consisting of 3144 amino acids and has conserved N-terminal sequences that are followed by a polyglutamine (polyQ) repeat. Loss of Htt is known to cause embryonic lethality in mice, whereas polyQ expansion leads to adult neuronal degeneration. Whether N-terminal HTT is essential for neuronal development or contributes only to late-onset neurodegeneration remains unknown. We established HTT knock-in mice (N160Q-KI) expressing the first 208 amino acids of HTT with 160Q, and they show age-dependent HTT aggregates in the brain and neurological phenotypes. Importantly, the N-terminal mutant HTT also preferentially accumulates in the striatum, the brain region most affected in HD, indicating the importance of N-terminal HTT in selective neuropathology. That said, homozygous N160Q-KI mice are also embryonic lethal, suggesting that N-terminal HTT alone is unable to support embryonic development. Using Htt knockout neurons, we found that loss of Htt selectively affects the survival of developing neuronal cells, but not astrocytes, in culture. This neuronal degeneration could be rescued by a truncated HTT lacking the first 237 amino acids, but not by N-terminal HTT (1-208 amino acids). Also, the rescue effect depends on the region in HTT known to be involved in intracellular trafficking. Thus, the N-terminal HTT region may not be essential for the survival of developing neurons, but when carrying a large polyQ repeat, can cause selective neuropathology. These findings imply a possible therapeutic benefit of removing the N-terminal region of HTT containing the polyQ repeat to treat the neurodegeneration in HD.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Peptides/genetics , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gene Knock-In Techniques , Humans , Huntingtin Protein/metabolism , Huntingtin Protein/therapeutic use , Huntington Disease/pathology , Huntington Disease/therapy , Mice , Mutation , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/therapeutic use , Neurons/metabolism , Neurons/pathology , Peptides/metabolism , Peptides/therapeutic use , Phenotype
8.
Mol Neurodegener ; 10: 42, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26334913

ABSTRACT

BACKGROUND: TAR DNA-binding protein 43 (TDP-43) is a nuclear protein, but it is redistributed in the neuronal cytoplasm in both amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Because small transgenic animal models often lack cytoplasmic TDP-43, how the cytoplasmic accumulation of TDP-43 contributes to these diseases remains unclear. The current study is aimed at studying the mechanism of cytoplasmic pathology of TDP-43. RESULTS: We established transgenic pigs expressing mutant TDP-43 (M337V). This pig model shows severe phenotypes and early death. We found that transgenic TDP-43 is also distributed in the cytoplasm of neuronal cells in the spinal cord and brain. Transgenic TDP-43 interacts with PSF, an RNA splicing factor that associates with NeuN to regulate neuronal RNA splicing. The interaction of TDP-43, PSF and NeuN causes PSF and NeuN mislocalize into the neuronal cytoplasm in transgenic pigs. Consistently, abnormal PSF-related neuronal RNA splicing is seen in TDP-43 transgenic pigs. The cytoplasmic localization of PSF and NeuN as well as abnormal PSF-related neuronal RNA splicing was also found in ALS patient brains. CONCLUSION: Our findings from a large mammalian model suggest that cytoplasmic mutant TDP-43 could reduce the nuclear function of RNA splicing factors, contributing to neuropathology.


Subject(s)
DNA-Binding Proteins/genetics , RNA Splicing , TDP-43 Proteinopathies/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Central Nervous System/metabolism , Central Nervous System/pathology , Cytoplasm/metabolism , DNA-Binding Proteins/physiology , Disease Models, Animal , Humans , Motor Neurons/pathology , Muscular Atrophy/etiology , Nerve Tissue Proteins/metabolism , Nuclear Transfer Techniques , PTB-Associated Splicing Factor , Phenotype , Protein Interaction Mapping , Protein Transport , RNA-Binding Proteins/metabolism , Species Specificity , Sus scrofa , TDP-43 Proteinopathies/pathology
9.
J Neurosci ; 35(21): 8345-58, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26019347

ABSTRACT

Parkinson's disease (PD) is an age-dependent neurodegenerative disease that often occurs in those over age 60. Although rodents and small animals have been used widely to model PD and investigate its pathology, their short life span makes it difficult to assess the aging-related pathology that is likely to occur in PD patient brains. Here, we used brain tissues from rhesus monkeys at 2-3, 7-8, and >15 years of age to examine the expression of Parkin, PINK1, and α-synuclein, which are known to cause PD via loss- or gain-of-function mechanisms. We found that α-synuclein is increased in the older monkey brains, whereas Parkin and PINK1 are decreased or remain unchanged. Because of the gain of toxicity of α-synuclein, we performed stereotaxic injection of lentiviral vectors expressing mutant α-synuclein (A53T) into the substantia nigra of monkeys and found that aging also increases the accumulation of A53T in neurites and its associated neuropathology. A53T also causes more extensive reactive astrocytes and axonal degeneration in monkey brain than in mouse brain. Using monkey brain tissues, we found that A53T interacts with neurofascin, an adhesion molecule involved in axon subcellular targeting and neurite outgrowth. Aged monkey brain tissues show an increased interaction of neurofascin with A53T. Overexpression of A53T causes neuritic toxicity in cultured neuronal cells, which can be attenuated by transfected neurofascin. These findings from nonhuman primate brains reveal age-dependent pathological and molecular changes that could contribute to the age-dependent neuropathology in PD.


Subject(s)
Aging/genetics , Aging/pathology , Brain/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , alpha-Synuclein/genetics , Aging/metabolism , Animals , Brain/metabolism , Haplorhini , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Nerve Degeneration/metabolism , alpha-Synuclein/biosynthesis
10.
Hum Mol Genet ; 24(8): 2308-17, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25552648

ABSTRACT

Parkinson's disease (PD) is an age-dependent neurodegenerative disease that can be caused by genetic mutations in α-synuclein (α-syn) or duplication of wild-type α-syn; PD is characterized by the deposition of α-syn aggregates, indicating a gain of toxicity from accumulation of α-syn. Although the major neuropathologic feature of PD is the degeneration of dopaminergic (DA) neurons in the substantia nigra, non-motor symptoms including anxiety, cognitive defect and sleep disorder precede the onset of motor impairment, and many clinical symptoms of PD are not caused by degeneration of DA neurons. Non-human primate models of PD are important for revealing the early pathology in PD and identifying effective treatments. We established transgenic PD rhesus monkeys that express mutant α-syn (A53T). Six transgenic A53T monkeys were produced via lentiviral vector expressing A53T in fertilized monkey eggs and subsequent embryo transfer to surrogates. Transgenic A53T is expressed in the monkey brain and causes age-dependent non-motor symptoms, including cognitive defects and anxiety phenotype, without detectable sleeping disorders. The transgenic α-syn monkeys demonstrate the specific early symptoms caused by mutant α-syn and provide insight into treatment of early PD.


Subject(s)
Disease Models, Animal , Macaca mulatta , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Brain/metabolism , Brain/pathology , Dopaminergic Neurons/metabolism , Female , Humans , Macaca mulatta/genetics , Macaca mulatta/metabolism , Male , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity
11.
J Neurosci ; 34(25): 8411-22, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948797

ABSTRACT

Huntington's disease (HD) belongs to a family of neurodegenerative diseases caused by misfolded proteins and shares the pathological hallmark of selective accumulation of misfolded proteins in neuronal cells. Polyglutamine expansion in the HD protein, huntingtin (Htt), causes selective neurodegeneration that is more severe in the striatum and cortex than in other brain regions, but the mechanism behind this selectivity is unknown. Here we report that in HD knock-in mice, the expression levels of mutant Htt (mHtt) are higher in brain tissues than in peripheral tissues. However, the expression of N-terminal mHtt via stereotaxic injection of viral vectors in mice also results in greater accumulation of mHtt in the striatum than in muscle. We developed an in vitro assay that revealed that extracts from the striatum and cortex promote the formation of high-molecular weight (HMW) mHtt compared with the relatively unaffected cerebellar and peripheral tissue extracts. Inhibition of ubiquitin-activating enzyme E1 (Ube1) increased the levels of HMW mHtt in the relatively unaffected tissues. Importantly, the expression levels of Ube1 are lower in brain tissues than peripheral tissues and decline in the nuclear fraction with age, which is correlated with the increased accumulation of mHtt in the brain and neuronal nuclei during aging. Our findings suggest that decreased targeting of misfolded Htt to the proteasome for degradation via Ube1 may underlie the preferential accumulation of toxic forms of mHtt in the brain and its selective neurodegeneration.


Subject(s)
Brain Chemistry/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ubiquitin-Activating Enzymes/physiology , Animals , Enzyme Activation/genetics , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntingtin Protein , Male , Mice , Mutation , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Tissue Distribution/genetics , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics
12.
Proc Natl Acad Sci U S A ; 111(15): 5706-11, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706802

ABSTRACT

Ubiquitination of misfolded proteins, a common feature of many neurodegenerative diseases, is mediated by different lysine (K) residues in ubiquitin and alters the levels of toxic proteins. In Huntington disease, polyglutamine expansion causes N-terminal huntingtin (Htt) to misfold, inducing neurodegeneration. Here we report that shorter N-terminal Htt fragments are more stable than longer fragments and find differential ubiquitination via K63 of ubiquitin. Aging decreases proteasome-mediated Htt degradation, at the same time increasing K63-mediated ubiquitination and subsequent Htt aggregation in HD knock-in mice. The association of Htt with the K48-specific E3 ligase, Ube3a, is decreased in aged mouse brain. Overexpression of Ube3a in HD mouse brain reduces K63-mediated ubiquitination and Htt aggregation, enhancing its degradation via the K48 ubiquitin-proteasome system. Our findings suggest that aging-dependent Ube3a levels result in differential ubiquitination and degradation of Htt fragments, thereby contributing to the age-related neurotoxicity of mutant Htt.


Subject(s)
Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Age Factors , Animals , Cycloheximide , Fluorescent Antibody Technique , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntingtin Protein , Immunoprecipitation , Mice
13.
Cell Res ; 24(4): 464-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24577199

ABSTRACT

Mutations in the human copper/zinc superoxide dismutase 1 (hSOD1) gene cause familial amyotrophic lateral sclerosis (ALS). It remains unknown whether large animal models of ALS mimic more pathological events seen in ALS patients via novel mechanisms. Here, we report the generation of transgenic pigs expressing mutant G93A hSOD1 and showing hind limb motor defects, which are germline transmissible, and motor neuron degeneration in dose- and age-dependent manners. Importantly, in the early disease stage, mutant hSOD1 did not form cytoplasmic inclusions, but showed nuclear accumulation and ubiquitinated nuclear aggregates, as seen in some ALS patient brains, but not in transgenic ALS mouse models. Our findings revealed that SOD1 binds PCBP1, a nuclear poly(rC) binding protein, in pig brain, but not in mouse brain, suggesting that the SOD1-PCBP1 interaction accounts for nuclear SOD1 accumulation and that species-specific targets are key to ALS pathology in large mammals and in humans.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Animals, Genetically Modified , Disease Models, Animal , Superoxide Dismutase/genetics , Swine , Alanine/genetics , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , DNA-Binding Proteins , Glycine/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mice , Phenotype , RNA-Binding Proteins , Species Specificity , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Swine/genetics
14.
Hum Mol Genet ; 23(10): 2678-93, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24381309

ABSTRACT

Mutations in TAR DNA-binding protein 43 (TDP-43) are associated with familial forms of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Although recent studies have revealed that mutant TDP-43 in neuronal and glial cells is toxic, how mutant TDP-43 causes primarily neuronal degeneration in an age-dependent manner remains unclear. Using adeno-associated virus (AAV) that expresses mutant TDP-43 (M337V) ubiquitously, we found that mutant TDP-43 accumulates preferentially in neuronal cells in the postnatal mouse brain. We then ubiquitously or selectively expressed mutant TDP-43 in neuronal and glial cells in the striatum of adult mouse brains via stereotaxic injection of AAV vectors and found that it also preferentially accumulates in neuronal cells. Expression of mutant TDP-43 in neurons in the striatum causes more severe degeneration, earlier death and more robust symptoms in mice than expression of mutant TDP-43 in glial cells; however, aging increases the expression of mutant TDP-43 in glial cells, and expression of mutant TDP-43 in older mice caused earlier onset of phenotypes and more severe neuropathology than that in younger mice. Although expression of mutant TDP-43 in glial cells via stereotaxic injection does not lead to robust neurological phenotypes, systemic inhibition of the proteasome activity via MG132 in postnatal mice could exacerbate glial TDP-43-mediated toxicity and cause mice to die earlier. Consistently, this inhibition increases the expression of mutant TDP-43 in glial cells in mouse brains. Thus, the differential accumulation of mutant TDP-43 in neuronal versus glial cells contributes to the preferential toxicity of mutant TDP-43 in neuronal cells and age-dependent pathology.


Subject(s)
Brain/pathology , DNA-Binding Proteins/genetics , Neuroglia/metabolism , Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , HEK293 Cells , Humans , Leupeptins/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , PC12 Cells , Proteasome Inhibitors/pharmacology , Proteolysis , Rats
15.
J Cell Biol ; 202(7): 1123-38, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24081492

ABSTRACT

Many genetic mouse models of Huntington's disease (HD) have established that mutant huntingtin (htt) accumulates in various subcellular regions to affect a variety of cellular functions, but whether and how synaptic mutant htt directly mediates HD neuropathology remains to be determined. We generated transgenic mice that selectively express mutant htt in the presynaptic terminals. Although it was not overexpressed, synaptic mutant htt caused age-dependent neurological symptoms and early death in mice as well as defects in synaptic neurotransmitter release. Mass spectrometry analysis of synaptic fractions and immunoprecipitation of synapsin-1 from HD CAG150 knockin mouse brains revealed that mutant htt binds to synapsin-1, a protein whose phosphorylation is critical for neurotransmitter release. We found that polyglutamine-expanded exon1 htt binds to the C-terminal region of synapsin-1 to reduce synapsin-1 phosphorylation. Our findings point to a critical role for synaptic htt in the neurological symptoms of HD, providing a new therapeutic target.


Subject(s)
Brain/pathology , Huntington Disease/etiology , Mutation/genetics , Nerve Tissue Proteins/physiology , Presynaptic Terminals/pathology , Synapsins/metabolism , Age Factors , Animals , Behavior, Animal , Blotting, Western , Brain/metabolism , Chromatography, Liquid , Dopamine/metabolism , Exons/genetics , Female , Fluorescent Antibody Technique , Glutamic Acid/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Peptides/genetics , Phosphorylation , Presynaptic Terminals/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Synapsins/genetics , Synaptic Transmission , Synaptosomal-Associated Protein 25/genetics , Tandem Mass Spectrometry , gamma-Aminobutyric Acid/metabolism
16.
J Neurosci ; 33(19): 8172-84, 2013 May 08.
Article in English | MEDLINE | ID: mdl-23658157

ABSTRACT

Mutations in the Abelson helper integration site-1 (AHI1) gene result in N-terminal Ahi1 fragments and cause Joubert syndrome, an autosomal recessive brain malformation disorder associated with delayed development. How AHI1 mutations lead to delayed development remains unclear. Here we report that full-length, but not N-terminal, Ahi1 binds Hap1, a huntingtin-associated protein that is essential for the postnatal survival of mice and that this binding is regulated during neuronal differentiation by nerve growth factor. Nerve growth factor induces dephosphorylation of Hap1A and decreases its association with Ahi1, correlating with increased Hap1A distribution in neurite tips. Consistently, Ahi1 associates with phosphorylated Hap1A in cytosolic, but not in synaptosomal, fractions isolated from mouse brain, suggesting that Ahi1 functions mainly in the soma of neurons. Mass spectrometry analysis of cytosolic Ahi1 immunoprecipitates reveals that Ahi1 also binds Cend1 (cell cycle exit and neuronal differentiation protein 1)/BM88, a neuronal protein that mediates neuronal differentiation and is highly expressed in postnatal mouse brain. Loss of Ahi1 reduces the levels of Cend1 in the hypothalamus of Ahi1 KO mice, which show retarded growth during postnatal days. Overexpressed Ahi1 can stabilize Cend1 in cultured cells. Furthermore, overexpression of Cend1 can rescue the neurite extension defects of hypothalamic neurons from Ahi1 KO mice. Our findings suggest that Cend1 is involved in Ahi1-associated hypothalamic neuronal differentiation in early development, giving us fresh insight into the mechanism behind the delayed development in Joubert syndrome.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Proto-Oncogene Proteins/deficiency , Adaptor Proteins, Vesicular Transport , Age Factors , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Hindlimb Suspension/physiology , Humans , Hypothalamus/cytology , Hypothalamus/growth & development , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/physiology , Neurons/ultrastructure , Phosphorylation/drug effects , Rats , Swimming , Transfection
17.
Cell Mol Life Sci ; 69(8): 1305-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21544547

ABSTRACT

Hap1 was originally identified as a neuronal protein that interacts with huntingtin, the Huntington's disease (HD) protein. Later studies revealed that Hap1 participates in intracellular trafficking in neuronal cells and that this trafficking function can be adversely affected by mutant huntingtin. Hap1 is also present in pancreatic ß-cells and other endocrine cells; however, the role of Hap1 in these endocrine cells remains unknown. Using the Cre-loxP system, we generated conditional Hap1 knockout mice to selectively deplete the expression of Hap1 in mouse pancreatic ß-cells. Mutant mice with Hap1 deficiency in pancreatic ß-cells had impaired glucose tolerance and decreased insulin release in response to intraperitoneally injected glucose. Using cultured pancreatic ß-cell lines and isolated mouse pancreatic islets, we confirmed that decreasing Hap1 could reduce glucose-mediated insulin release. Electron microscopy suggested that there was a reduced number of insulin-containing vesicles docked at the plasma membrane of pancreatic islets in Hap1 mutant mice following intraperitoneal glucose injection. Glucose treatment decreased the phosphorylation of Hap1A in cultured ß-cells and in mouse pancreatic tissues. Moreover, this glucose treatment increased Hap1's association with kinesin light chain and dynactin p150, both of which are involved in microtubule-dependent trafficking. These studies suggest that Hap1 is important for insulin release from ß-cells via dephosphorylation that can regulate its intracellular trafficking function.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Line , Glucose/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Phosphorylation
18.
Hum Mol Genet ; 20(7): 1424-37, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21245084

ABSTRACT

An expanded polyglutamine tract (>37 glutamines) in the N-terminal region of huntingtin (htt) causes htt to accumulate in the nucleus, leading to transcriptional dysregulation in Huntington disease (HD). In HD knock-in mice that express full-length mutant htt at the endogenous level, mutant htt preferentially accumulates in the nuclei of striatal neurons, which are affected most profoundly in HD. The mechanism underlying this preferential nuclear accumulation of mutant htt in striatal neurons remains unknown. Here, we report that serine 16 (S16) in htt is important for the generation of small N-terminal fragments that are able to accumulate in the nucleus and form aggregates. Phosphorylation of N-terminal S16 in htt promotes the nuclear accumulation of small N-terminal fragments and reduces the interaction of N-terminal htt with the nuclear pore complex protein Tpr. Mouse brain striatal tissues show increased S16 phosphorylation and a decreased association between mutant N-terminal htt and Tpr. These findings provide mechanistic insight into the nuclear accumulation of mutant htt and the selective neuropathology of HD, revealing potential therapeutic targets for treating this disease.


Subject(s)
Corpus Striatum/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Animals , Corpus Striatum/pathology , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/therapy , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Neurons/physiology , Nuclear Pore/genetics , Nuclear Pore/pathology , Nuclear Proteins/genetics , Phosphorylation , Polyglutamic Acid/genetics , Polyglutamic Acid/metabolism , Protein Structure, Tertiary
19.
Mol Brain ; 3: 33, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-21044321

ABSTRACT

Huntington's disease results from expansion of a glutamine repeat (>36 glutamines) in the N-terminal region of huntingtin (htt) and is characterized by preferential neurodegeneration in the striatum of the brain. N171-82Q mice that express N-terminal 171 amino acids of htt with an 82-glutamine repeat show severe neurological phenotypes and die early, suggesting that N-terminal mutant htt is pathogenic. In addition, various cellular factors and genetic modifiers are found to modulate the cytotoxicity of mutant htt. Understanding the contribution of these factors to HD pathogenesis will help identify therapeutics for this disease. To investigate the role of interleukin type 1 (IL-1), a cytokine that has been implicated in various neurological diseases, in HD neurological symptoms, we crossed N171-82Q mice to type I IL-1 receptor (IL-1RI) knockout mice. Mice lacking IL-1RI and expressing N171-82Q show more severe neurological symptoms than N171-82Q or IL-1RI knockout mice, suggesting that lack of IL-1RI can promote the neuronal toxicity of mutant htt. Lack of IL-1RI also increases the accumulation of transgenic mutant htt in the striatum in N171-82Q mice. Since IL-1RI signaling mediates both toxic and protective effects on neurons, its basal function and protective effects may be important for preventing the neuropathology seen in HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/physiopathology , Nerve Tissue Proteins , Nuclear Proteins , Phenotype , Receptors, Interleukin-1 Type I/metabolism , Animals , Brain/anatomy & histology , Brain/metabolism , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Huntingtin Protein , Huntington Disease/pathology , Male , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Interleukin-1 Type I/genetics , Rotarod Performance Test , Signal Transduction/physiology
20.
Hum Mol Genet ; 19(20): 3983-94, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20660116

ABSTRACT

Neurodegeneration is a hallmark of many neurological diseases, including Alzheimer's, Parkinson's and the polyglutamine diseases, which are all caused by misfolded proteins that accumulate in neuronal cells of the brain. Although apoptosis is believed to contribute to neurodegeneration in these cases, genetic mouse models of these diseases often fail to replicate apoptosis and overt neurodegeneration in the brain. Using nuclear transfer, we generated transgenic Huntington's disease (HD) pigs that express N-terminal (208 amino acids) mutant huntingtin with an expanded polyglutamine tract (105Q). Postnatal death, dyskinesia and chorea-like movement were observed in some transgenic pigs that express mutant huntingtin. Importantly, the transgenic HD pigs, unlike mice expressing the same transgene, displayed typical apoptotic neurons with DNA fragmentation in their brains. Also, expression of mutant huntingtin resulted in more neurons with activated caspase-3 in transgenic pig brains than that in transgenic mouse brains. Our findings suggest that species differences determine neuropathology and underscore the importance of large mammalian animals for modeling neurological disorders.


Subject(s)
Brain/pathology , Disease Models, Animal , Huntington Disease , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Swine, Miniature , Animals , Animals, Genetically Modified/metabolism , Apoptosis , Brain/metabolism , Caspase 3/biosynthesis , DNA Fragmentation , Gene Expression , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurodegenerative Diseases , Neurons/pathology , Proteostasis Deficiencies , Species Specificity , Swine/genetics , Swine/metabolism , Swine, Miniature/genetics , Swine, Miniature/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...