Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
bioRxiv ; 2024 May 17.
Article in English | MEDLINE | ID: mdl-38798341

ABSTRACT

TDP43 is an RNA/DNA binding protein increasingly recognized for its role in neurodegenerative conditions including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). As characterized by its aberrant nuclear export and cytoplasmic aggregation, TDP43 proteinopathy is a hallmark feature in over 95% of ALS/FTD cases, leading to the formation of detrimental cytosolic aggregates and a reduction in nuclear functionality within neurons. Building on our prior work linking TDP43 proteinopathy to the accumulation of DNA double-strand breaks (DSBs) in neurons, the present investigation uncovers a novel regulatory relationship between TDP43 and DNA mismatch repair (MMR) gene expressions. Here, we show that TDP43 depletion or overexpression directly affects the expression of key MMR genes. Alterations include MLH1, MSH2, MSH3, MSH6, and PMS2 levels across various primary cell lines, independent of their proliferative status. Our results specifically establish that TDP43 selectively influences the expression of MLH1 and MSH6 by influencing their alternative transcript splicing patterns and stability. We furthermore find aberrant MMR gene expression is linked to TDP43 proteinopathy in two distinct ALS mouse models and post-mortem brain and spinal cord tissues of ALS patients. Notably, MMR depletion resulted in the partial rescue of TDP43 proteinopathy-induced DNA damage and signaling. Moreover, bioinformatics analysis of the TCGA cancer database reveals significant associations between TDP43 expression, MMR gene expression, and mutational burden across multiple cancers. Collectively, our findings implicate TDP43 as a critical regulator of the MMR pathway and unveil its broad impact on the etiology of both neurodegenerative and neoplastic pathologies.

2.
Neurobiol Dis ; 192: 106414, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253209

ABSTRACT

Alteration in protein citrullination (PC), a common posttranslational modification (PTM), contributes to pathogenesis in various inflammatory disorders. We previously reported that PC and protein arginine deiminase 2 (PAD2), the predominant enzyme isoform that catalyzes this PTM in the central nervous system (CNS), are altered in mouse models of amyotrophic lateral sclerosis (ALS). We now demonstrate that PAD2 expression and PC are altered in human postmortem ALS spinal cord and motor cortex compared to controls, increasing in astrocytes while trending lower in neurons. Furthermore, PC is enriched in protein aggregates that contain the myelin proteins PLP and MBP in ALS. These results confirm our findings in ALS mouse models and suggest that altered PAD2 and PC contribute to neurodegeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Citrullination , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/metabolism , Gliosis/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Myelin Proteins/metabolism , Myelin Sheath/pathology , Protein Aggregates , Protein-Arginine Deiminase Type 2/metabolism , Protein-Arginine Deiminases/metabolism , Proteins/metabolism , Spinal Cord/pathology
3.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555655

ABSTRACT

ALS-linked mutations induce aberrant conformations within the SOD1 protein that are thought to underlie the pathogenic mechanism of SOD1-mediated ALS. Although clinical trials are underway for gene silencing of SOD1, these approaches reduce both wild-type and mutated forms of SOD1. Here, we sought to develop anti-SOD1 nanobodies with selectivity for mutant and misfolded forms of human SOD1 over wild-type SOD1. Characterization of two anti-SOD1 nanobodies revealed that these biologics stabilize mutant SOD1 in vitro. Further, SOD1 expression levels were enhanced and the physiological subcellular localization of mutant SOD1 was restored upon co-expression of anti-SOD1 nanobodies in immortalized cells. In human motor neurons harboring the SOD1 A4V mutation, anti-SOD1 nanobody expression promoted neurite outgrowth, demonstrating a protective effect of anti-SOD1 nanobodies in otherwise unhealthy cells. In vitro assays revealed that an anti-SOD1 nanobody exhibited selectivity for human mutant SOD1 over endogenous murine SOD1, thus supporting the preclinical utility of anti-SOD1 nanobodies for testing in animal models of ALS. In sum, the anti-SOD1 nanobodies developed and presented herein represent viable biologics for further preclinical testing in human and mouse models of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Single-Domain Antibodies , Humans , Mice , Animals , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Single-Domain Antibodies/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Protein Folding , Motor Neurons/metabolism , Neuronal Outgrowth , Mutation
4.
Acta Neuropathol Commun ; 10(1): 135, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36076282

ABSTRACT

Increased protein citrullination (PC) and dysregulated protein arginine deiminase (PAD) activity have been observed in several neurodegenerative diseases. PC is a posttranslational modification catalyzed by the PADs. PC converts peptidyl-arginine to peptidyl-citrulline, thereby reducing the positive charges and altering structure and function of proteins. Of the five PADs, PAD2 is the dominant isoform in the central nervous system (CNS). Abnormal PC and PAD dysregulation are associated with numerous pathological conditions, including inflammatory diseases and neurodegeneration. Animal model studies have shown therapeutic efficacy from inhibition of PADs, thus suggesting a role of PC in pathogenesis. To determine whether PC contribute to amyotrophic lateral sclerosis (ALS), a deadly neurodegenerative disease characterized by loss of motor neurons, paralysis, and eventual death, we investigated alterations of PC and PAD2 in two different transgenic mouse models of ALS expressing human mutant SOD1G93A and PFN1C71G, respectively. PC and PAD2 expression are altered dynamically in the spinal cord during disease progression in both models. PC and PAD2 increase progressively in astrocytes with the development of reactive astrogliosis, while decreasing in neurons. Importantly, in the spinal cord white matter, PC accumulates in protein aggregates that contain the myelin proteins PLP and MBP. PC also accumulates progressively in insoluble protein fractions during disease progression. Finally, increased PC and PAD2 expression spatially correlate with areas of the CNS with the most severe motor neuron degeneration. These results suggest that altered PC is an integral part of the neurodegenerative process and potential biomarkers for disease progression in ALS. Moreover, increased PC may contribute to disease-associated processes such as myelin protein aggregation, myelin degeneration, and astrogliosis.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/pathology , Animals , Citrullination , Disease Models, Animal , Disease Progression , Gliosis/pathology , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Myelin Proteins , Myelin Sheath/pathology , Neurodegenerative Diseases/pathology , Profilins/metabolism , Protein Aggregates , Spinal Cord/pathology , Superoxide Dismutase/genetics
5.
PLoS One ; 17(2): e0255710, 2022.
Article in English | MEDLINE | ID: mdl-35113871

ABSTRACT

Modestly increased expression of transactive response DNA binding protein (TDP-43) gene have been reported in amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and other neuromuscular diseases. However, whether this modest elevation triggers neurodegeneration is not known. Although high levels of TDP-43 overexpression have been modeled in mice and shown to cause early death, models with low-level overexpression that mimic the human condition have not been established. In this study, transgenic mice overexpressing wild type TDP-43 at less than 60% above the endogenous CNS levels were constructed, and their phenotypes analyzed by a variety of techniques, including biochemical, molecular, histological, behavioral techniques and electromyography. The TDP-43 transgene was expressed in neurons, astrocytes, and oligodendrocytes in the cortex and predominantly in astrocytes and oligodendrocytes in the spinal cord. The mice developed a reproducible progressive weakness ending in paralysis in mid-life. Detailed analysis showed ~30% loss of large pyramidal neurons in the layer V motor cortex; in the spinal cord, severe demyelination was accompanied by oligodendrocyte injury, protein aggregation, astrogliosis and microgliosis, and elevation of neuroinflammation. Surprisingly, there was no loss of lower motor neurons in the lumbar spinal cord despite the complete paralysis of the hindlimbs. However, denervation was detected at the neuromuscular junction. These results demonstrate that low-level TDP-43 overexpression can cause diverse aspects of ALS, including late-onset and progressive motor dysfunction, neuroinflammation, and neurodegeneration. Our findings suggest that persistent modest elevations in TDP-43 expression can lead to ALS and other neurological disorders involving TDP-43 proteinopathy. Because of the predictable and progressive clinical paralytic phenotype, this transgenic mouse model will be useful in preclinical trial of therapeutics targeting neurological disorders associated with elevated levels of TDP-43.


Subject(s)
Amyotrophic Lateral Sclerosis
6.
Niger J Physiol Sci ; 37(2): 255-260, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-38243556

ABSTRACT

Physiology remains one of the core disciplines on which all biological and medical sciences were founded. In Nigeria, it is known that most students study Physiology at the undergraduate level by chance and not by choice and end up performing poorly, which could be mainly due to low awareness and knowledge of the discipline, its opportunities, and prospects. Therefore, this study investigated the awareness, attitude, and knowledge about physiology among senior secondary school students in Southwest Nigeria. A cross-sectional of 544 students in science-based senior secondary schools in south-west Nigeria were sampled. Our results showed a high level of awareness, with television being the dominant medium of information. However, knowledge of Physiology was low, while most of the students also showed interest in knowing more about it. Although gender does not seem to influence the level of knowledge, females had a better attitude towards learning about physiology. Similarly, residence did not affect attitude, howbeit associated with the level of knowledge. In conclusion, the high awareness and low knowledge observed in this study would give insights to educate students at the early stages of education about the opportunity and prospects of Physiology and other science-related disciplines.


Subject(s)
Health Knowledge, Attitudes, Practice , Students , Female , Humans , Nigeria , Cross-Sectional Studies , Schools , Surveys and Questionnaires
7.
J Basic Clin Physiol Pharmacol ; 33(3): 355-362, 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33873260

ABSTRACT

OBJECTIVES: The human body physiology rapidly changes and adapt to several environmental stimuli, including light. Abnormal artificial light exposures have been shown to affect sleep cycle, cognition, and mood. Although studies have reported inconsistent effects of short-term or constant long-term light exposures, human exposures to artificial lights occur at varying, unpredictable times and duration daily. Here, we studied the effects of long-term unpredictable light exposure on learning, memory, oxidative status, and associated cytokines in rats. METHODS: Artificial lighting was provided using an array of white light-emitting diodes coupled to a microcontroller that switches them on or off at unpredictable times and duration (light intensity = 200 ± 20 lx). Within the last eight days of 40 days exposure, animals were subjected to open field test, Morris water maze, and novel object recognition behavioral paradigms. Brain levels of malondialdehyde (MDA), superoxide dismutase (SOD), catalase, reduced glutathione (GSH), glutathione S-transferase (GST), tumor necrosis factor-alpha (TNF-α), and vascular endothelial growth factor (VEGF) were assayed. RESULTS: Exposed rats showed impaired spatial learning and memory (p<0.05), but no changes in object recognition memory or locomotor activity. Oxidative stress analyses also revealed significant changes in the concentrations of MDA, SOD, catalase, and GSH levels (p<0.05), not GST. Similarly, there was an increased TNF-α expression (p<0.05), not VEGF. CONCLUSIONS: We conclude that oxidative stress is involved in memory impairment in rats exposed to prolonged unpredictable lights, which again suggests the detrimental effects of extended light exposure on the nervous system.


Subject(s)
Spatial Memory , Tumor Necrosis Factor-alpha , Animals , Catalase/metabolism , Hippocampus/metabolism , Maze Learning , Memory Disorders/etiology , Oxidative Stress , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
8.
Mol Neurobiol ; 58(5): 2396-2406, 2021 May.
Article in English | MEDLINE | ID: mdl-33421017

ABSTRACT

Proper development of neuronal cells is important for brain functions, and impairment of neuronal development may lead to neuronal disorders, implying that improvement in neuronal development may be a therapeutic direction for these diseases. Huntington's disease (HD) is a neurodegenerative disease characterized by impairment of neuronal structures, ultimately leading to neuronal death and dysfunctions of the central nervous system. Based on previous studies, fibroblast growth factor 9 (FGF9) may provide neuroprotective functions in HD, and FGFs may enhance neuronal development and neurite outgrowth. However, whether FGF9 can provide neuronal protective functions through improvement of neuronal morphology in HD is still unclear. Here, we study the effects of FGF9 on neuronal length in HD and attempt to understand the related working mechanisms. Taking advantage of striatal cell lines from HD knock-in mice, we found that FGF9 increases total neuronal length and upregulates several structural and synaptic proteins under HD conditions. In addition, activation of nuclear factor kappa B (NF-kB) signaling by FGF9 was observed to be significant in HD cells, and blockage of NF-kB leads to suppression of these structural and synaptic proteins induced by FGF9, suggesting the involvement of NF-kB signaling in these effects of FGF9. Taken these results together, FGF9 may enhance total neuronal length through upregulation of NF-kB signaling, and this mechanism could serve as an important mechanism for neuroprotective functions of FGF9 in HD.


Subject(s)
Corpus Striatum/drug effects , Fibroblast Growth Factor 9/pharmacology , Huntington Disease/metabolism , NF-kappa B/metabolism , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Cell Line , Corpus Striatum/metabolism , Disease Models, Animal , Mice , Neurons/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
9.
Life Sci ; 267: 118952, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33383048

ABSTRACT

AIMS: Huntington's disease (HD) is a neurodegenerative disease that causes deficits in neurite outgrowth, which suggests that enhancement of neurite outgrowth is a potential direction by which to improve HD. Our previous publications showed that fibroblast growth factor 9 (FGF9) provides anti-apoptosis and anti-oxidative functions in striatal cell models of HD through the extracellular signal-regulated kinases (ERK) pathway, and FGF9 also stimulates cytoskeletons to enhance neurite outgrowth via nuclear factor kappa B (NF-kB) signaling. In this study, we further demonstrate the importance of the ERK pathway for the neurite outgrowth induced by FGF9 in HD striatal models. MATERIALS AND METHODS: FGF9 was treated with ERK (U0126) or NF-kB (BAY11-7082) inhibitors in STHdhQ7/Q7 and STHdhQ111/Q111 striatal knock-in cell lines to examine neurite outgrowth, cytoskeletal markers, and synaptic proteins via immunofluorescence staining and Western blotting. NF-kB activity was analyzed by NF-kB promoter reporter assay. KEY FINDINGS: Here, we show that suppression of ERK signaling significantly inhibits FGF9-induced neurite outgrowth, cytoskeletal markers, and synaptic proteins in HD striatal cells. In addition, we also show suppression of ERK signaling significantly decreases FGF9-induced NF-kB activation, whereas suppression of NF-kB does not decrease FGF9-induced ERK signaling. These results suggest that FGF9 activates ERK signaling first, stimulates NF-kB upregulation, and then enhances neurite outgrowth in HD striatal cells. SIGNIFICANCE: We elucidate the more detailed mechanisms of neurite outgrowth enhanced by FGF9 in these HD striatal cells. This study may provide insights into targeting neurite outgrowth for HD therapy.


Subject(s)
Fibroblast Growth Factor 9/metabolism , Fibroblast Growth Factor 9/pharmacology , MAP Kinase Signaling System/drug effects , Neurites/metabolism , Animals , Butadienes/pharmacology , Cell Line , Cells, Cultured , Corpus Striatum/metabolism , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 9/antagonists & inhibitors , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Mice , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neurites/drug effects , Neuronal Outgrowth/physiology , Nitriles/pharmacology , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Recombinant Proteins/pharmacology , Signal Transduction , Sulfones/pharmacology
10.
Free Radic Biol Med ; 130: 256-266, 2019 01.
Article in English | MEDLINE | ID: mdl-30391672

ABSTRACT

Huntington's disease (HD) is a heritable neurodegenerative disorder, and has been characterized as an increase of oxidative stress in brain regions. In our previous results, we showed fibroblast growth factor 9 (FGF9) provides neuroprotective functions to suppress cell death in HD striatal cells dominantly through ERK signalling. However, whether the working mechanism of FGF9 is related to anti-oxidative stress in HD is still unknown. In this study, STHdhQ7/Q7 (Q7) and STHdhQ111/Q111 (Q111) striatal knock-in cell lines were used to examine the neuroprotective effects of FGF9 against oxidative stress in HD. Results show that FGF9 alleviates oxidative stress induced by starvation in Q7 and Q111 cells. The treatment of FGF9 not only induces upregulation and activation of nuclear factor erythroid 2-like 2 (Nrf2), a critical transcription factor for anti-oxidative stress, but also further upregulates its downstream targets, such as superoxide dismutase 2, gamma-glutamylcysteine synthetase and glutathione reductase. Furthermore, blockage of the Nrf2 pathway abolishes the anti-oxidative functions of FGF9, and inhibition of ERK signalling reduces the activation of the FGF9-Nrf2 pathway, resulting in higher level of oxidative stress in HD cells. These results support the neuroprotective effects of FGF9 against oxidative stress through the ERK-Nrf2 pathway, and imply one of potential strategies for therapy of HD.


Subject(s)
Antioxidants/pharmacology , Brain/metabolism , Fibroblast Growth Factor 9/genetics , Huntington Disease/drug therapy , NF-E2-Related Factor 2/genetics , Animals , Brain/pathology , Cell Line , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione Reductase/genetics , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , MAP Kinase Signaling System/genetics , Mice , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Superoxide Dismutase/genetics , Visual Cortex/metabolism , Visual Cortex/pathology
11.
Cell Physiol Biochem ; 48(2): 605-617, 2018.
Article in English | MEDLINE | ID: mdl-30021209

ABSTRACT

BACKGROUND/AIMS: Huntington's disease (HD) is a heritable neurodegenerative disorder, and there is no cure for HD to date. A type of fibroblast growth factor (FGF), FGF9, has been reported to play prosurvival roles in other neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. However, the effects of FGF9 on HD is still unknown. With many similarities in the cellular and pathological mechanisms that eventually cause cell death in neurodegenerative diseases, we hypothesize that FGF9 might provide neuroprotective functions in HD. METHODS: In this study, STHdhQ7/Q7 (WT) and STHdhQ111/Q111 (HD) striatal knock-in cell lines were used to evaluate the neuroprotective effects of FGF9. Cell proliferation, cell death and neuroprotective markers were determined via the MTT assay, propidium iodide staining and Western blotting, respectively. The signaling pathways regulated by FGF9 were demonstrated using Western blotting. Additionally, HD transgenic mouse models were used to further confirm the neuroprotective effects of FGF9 via ELISA, Western blotting and immunostaining. RESULTS: Results show that FGF9 not only enhances cell proliferation, but also alleviates cell death as cells under starvation stress. In addition, FGF9 significantly upregulates glial cell line-derived neurotrophic factor (GDNF) and an anti-apoptotic marker, Bcl-xL, and decreases the expression level of an apoptotic marker, cleaved caspase 3. Furthermore, FGF9 functions through ERK, AKT and JNK pathways. Especially, ERK pathway plays a critical role to influence the effects of FGF9 toward cell survival and GDNF production. CONCLUSIONS: These results not only show the neuroprotective effects of FGF9, but also clarify the critical mechanisms in HD cells, further providing an insight for the therapeutic potential of FGF9 in HD.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 9/pharmacology , MAP Kinase Signaling System/drug effects , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Caspase 3/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Transgenic , Nitriles/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Visual Cortex/cytology , Visual Cortex/drug effects , Visual Cortex/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL