Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 49
1.
PLoS One ; 19(3): e0299042, 2024.
Article En | MEDLINE | ID: mdl-38427657

Epigallocatechin gallate (EGCG) is a polyphenolic component of green tea that has anti-oxidative and anti-inflammatory effects in neurons. Ischemic stroke is a major neurological disease that causes irreversible brain disorders. It increases the intracellular calcium concentration and induces apoptosis. The regulation of intracellular calcium concentration is important to maintain the function of the nervous system. Hippocalcin is a neuronal calcium sensor protein that controls intracellular calcium concentration. We investigated whether EGCG treatment regulates the expression of hippocalcin in stroke animal model and glutamate-induced neuronal damage. We performed middle cerebral artery occlusion (MCAO) to induce cerebral ischemia. EGCG (50 mg/kg) or phosphate buffered saline was injected into the abdominal cavity just before MCAO surgery. The neurobehavioral tests were performed 24 h after MCAO surgery and cerebral cortex tissue was collected. MCAO damage induced severe neurobehavioral disorders, increased infarct volume, and decreased the expression of hippocalcin in the cerebral cortex. However, EGCG treatment improved these deficits and alleviated the decrease in hippocalcin expression in cerebral cortex. In addition, EGCG dose-dependently alleviated neuronal cell death and intracellular calcium overload in glutamate-exposed neurons. Glutamate exposure reduced hippocalcin expression, decreased Bcl-2 expression, and increased Bax expression. However, EGCG treatment mitigated these changes caused by glutamate toxicity. EGCG also attenuated the increase in caspase-3 and cleaved caspase-3 expressions caused by glutamate exposure. The effect of EGCG was more pronounced in non-transfected cells than in hippocalcin siRNA-transfected cells. These findings demonstrate that EGCG protects neurons against glutamate toxicity through the regulation of Bcl-2 family proteins and caspase-3. It is known that hippocalcin exerts anti-apoptotic effect through the modulation of apoptotic pathway. Thus, we can suggest evidence that EGCG has a neuroprotective effect by regulating hippocalcin expression in ischemic brain damage and glutamate-exposed cells.


Catechin , Ischemic Stroke , Neuroprotective Agents , Animals , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , Catechin/analogs & derivatives , Glutamic Acid/metabolism , Hippocalcin/genetics , Hippocalcin/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Disease Models, Animal
2.
Theranostics ; 12(17): 7450-7464, 2022.
Article En | MEDLINE | ID: mdl-36438486

Rationale: Hepatocellular carcinoma (HCC) is one of the most severe cancers worldwide, with few effective targeted therapies for HCC. Lipid metabolic reprogramming is emerged as a hallmark of cancer metabolism that guides response to antitumoral therapies. Such lipid metabolic alteration in cancers is critically regulated by the mammalian target of rapamycin mTOR, which is considered as a promising therapeutic target. Despite efforts, mTOR inhibitors (mTORi) have produced limited response clinically, partly due to incomplete knowledge of mTORC1 addiction in cancers. Methods: CRISPR-Cas9 system was used to establish Hpcal1 null mice. The liver cancer model in mice was generated using Hpcal1-deficient mice with diethylnitrosamine (DEN) /CCL4 or MYC/Trp53-/- via hydrodynamic tail-vein injection. RNA-sequencing (RNA-seq) was used to identify potential signaling pathways. The expression of HPCAL1 and mTOR signaling were determined using quantitative polymerase chain reaction (qPCR), western blot and immunohistochemistry. The role of Hpcal1 in liver tumorigenesis and its response to mTORi was assessed by CCK-8 measurements, colony formation assay and in mouse model. Results: In this study, we identified hippocalcin-like protein 1 (HPCAL1) as an important negative regulator of de novo lipid biosynthesis and mTOR signaling activation, limiting liver tumorigenesis and establishing a metabolic vulnerability of HCC in mice. Genetic loss of HPCAL1 rendered HCC mTORC1-addicted and sensitive to mTORi AZD-8055 in vitro and in vivo. Importantly, HPCAL1 expression was inversely correlated with the levels of mTOR phosphorylation and several critical lipid biosynthesis enzymes in human specimens. Mechanistically, HPCAL1 directly bound to RuvB Like AAA ATPase 1 (RUVBL1), inhibiting the assembly of TEL2-TTI1-TTI2 (TTT)-RUVBL complex and subsequent leading the mTOR signaling suppression. Conclusion: We uncover a metabolic vulnerability and mTOR addiction in HCC with HPCAL1 loss that provides a selective therapeutic window for HCC with mTORC1 hyperactivation using mTORi.


Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , ATPases Associated with Diverse Cellular Activities/metabolism , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic , DNA Helicases/metabolism , Hippocalcin/metabolism , Lipid Metabolism , Lipids , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mammals/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Cell Oncol (Dordr) ; 45(1): 179-191, 2022 Feb.
Article En | MEDLINE | ID: mdl-35102488

BACKGROUND: Hippocalcin-like 1 (HPCAL1), a neuronal calcium sensor protein family member, has been reported to regulate cancer growth. As yet, however, the biological functions of HPCAL1 and its molecular mechanisms have not been investigated in non-small cell lung carcinoma (NSCLC). METHODS: HPCAL1 expression in NSCLC samples was detected using immunohistochemistry, Western blotting and RT-PCR. The anticancer effects of HPCAL1 knockdown were determined by MTT, soft agar, cell cycle, oxygen consumption and reactive oxygen species assays. The effect of HPCAL1 knockdown on in vivo tumor growth was assessed using NSCLC cancer patient-derived xenograft models. Potentially interacting protein partners of HPCAL1 were identified using IP-MS/MS, immunoprecipitation and Western blotting assays. Metabolic alterations resulting from HPCAL1 knockdown were investigated using non-targeted metabolomics and RNA sequencing analyses. RESULTS: We found that HPCAL1 is highly expressed in NSCLC tissues and is positively correlated with low survival rates and AJCC clinical staging in lung cancer patients. Knockdown of HPCAL1 strongly increased oxygen consumption rates and the production of reactive oxygen species. HPCAL1 knockdown also inhibited NSCLC cell growth and patient-derived NSCLC tumor growth in vivo. Mechanistically, we found that HPCAL1 can directly bind to LDHA and enhance SRC-mediated phosphorylation of LDHA at tyrosine 10. The metabolomics and RNA sequencing analyses indicated that HPCAL1 knockdown reduces amino acid levels and induces fatty acid synthesis through regulating the expression of metabolism-related genes. Additionally, rescued cells expressing wild-type or mutant LDHA in HPCAL1 knockdown cells suggest that LDHA may serve as the main substrate of HPCAL1. CONCLUSIONS: Our data indicate that the effect of HPCAL1 knockdown on reducing SRC-mediated LDHA activity attenuates NSCLC growth. Our findings reveal novel biological functions and a mechanism underlying the role of HPCAL1 in NSCLC growth in vitro and in vivo.


Carcinoma, Non-Small-Cell Lung , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms , Neurocalcin/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Hippocalcin/genetics , Hippocalcin/metabolism , Humans , Lung Neoplasms/pathology , Tandem Mass Spectrometry
4.
Neuropediatrics ; 52(5): 377-382, 2021 10.
Article En | MEDLINE | ID: mdl-33511595

BACKGROUND: Recent research suggested an hippocalcin (HPCA)-related form of DYT2-like autosomal recessive dystonia. Two reports highlight a broad spectrum of the clinical phenotype. Here, we describe a novel HPCA gene variant in a pediatric patient and two affected relatives. METHODS: Whole exome sequencing was applied after a thorough clinical and neurological examination of the index patient and her family members. Results of neuropsychological testing were analyzed. RESULTS: Whole exome sequencing revealed a novel homozygous missense variant in the HPCA gene [c.182C>T p.(Ala61Val)] in our pediatric patient and the two affected family members. Clinically, the cases presented with dystonia, dysarthria, and jerky movements. We observed a particular cognitive profile with executive dysfunctions in our patient, which corresponds to the cognitive deficits that have been observed in the patients previously described. CONCLUSION: We present a novel genetic variant of the HPCA gene associated with autosomal recessive dystonia in a child with childhood-onset dystonia supporting its clinical features. Furthermore, we propose specific HPCA-related cognitive changes in homozygous carriers, underlining the importance of undertaking a systematic assessment of cognition in HPCA-related dystonia.


Dystonia , Dystonic Disorders , Child , Cognition , Dystonia/genetics , Dystonic Disorders/genetics , Female , Hippocalcin/genetics , Hippocalcin/metabolism , Humans , Mutation
5.
Neuroscience ; 430: 47-62, 2020 03 15.
Article En | MEDLINE | ID: mdl-31982469

Calcium acts as a second messenger that mediates physiologic functions, such as metabolism, cell proliferation, and apoptosis. Hippocalcin is a neuronal calcium sensor protein that regulates intracellular calcium concentration. Moreover, it prevents neuronal cell death from oxidative stress. Quercetin has excellent antioxidant properties and preventative effects. We studied modulation of hippocalcin expression by quercetin treatment in cerebral ischemic injury and glutamate-induced neuronal cell damage. Focal cerebral ischemia was induced by permanent middle cerebral artery occlusion (pMCAO). Male Sprague-Dawley rats were injected with vehicle or quercetin (10 mg/kg) 1 h prior to pMCAO, and cerebral cortical tissues were isolated 24 h after pMCAO. Quercetin improved pMCAO-induced neuronal movement deficit and infarction. pMCAO induced a decrease in hippocalcin expression in the cerebral cortex. However, quercetin treatment attenuated this pMCAO-induced decrease. In cultured hippocampal cells, glutamate excitotoxicity dramatically increased the intracellular calcium concentration, whereas quercetin alleviated intracellular calcium overload. Moreover, Western blot and immunocytochemical studies showed reduction of hippocalcin expression in glutamate-exposed cells. Quercetin prevented this glutamate-induced decrease. Furthermore, caspase-3 expression in hippocalcin siRNA transfection conditions is higher than caspase-3 expression in un-transfection conditions. Quercetin treatment attenuated the increase of caspase-3. Taken together, these results suggest that quercetin exerts a preventative effect through attenuation of intracellular calcium overload and restoration of down-regulated hippocalcin expression during ischemic injury.


Brain Injuries , Brain Ischemia , Neuroprotective Agents , Animals , Brain Ischemia/drug therapy , Calcium/metabolism , Hippocalcin/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Male , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley
6.
Neurobiol Dis ; 132: 104529, 2019 12.
Article En | MEDLINE | ID: mdl-31301343

A recent report of autosomal-recessive primary isolated dystonia (DYT2 dystonia) identified mutations in HPCA, a gene encoding a neuronal calcium sensor protein, hippocalcin (HPCA), as the cause of this disease. However, how mutant HPCA leads to neuronal dysfunction remains unknown. Using a multidisciplinary approach, we demonstrated the failure of dystonic N75K HPCA mutant to decode short bursts of action potentials and theta rhythms in hippocampal neurons by its Ca2+-dependent translocation to the plasma membrane. This translocation suppresses neuronal activity via slow afterhyperpolarization (sAHP) and we found that the N75K mutant could not control sAHP during physiologically relevant neuronal activation. Simulations based on the obtained experimental results directly demonstrated an increased excitability in neurons expressing N75K mutant instead of wild type (WT) HPCA. In conclusion, our study identifies sAHP as a downstream cellular target perturbed by N75K mutation in DYT2 dystonia, demonstrates its impact on neuronal excitability, and suggests a potential therapeutic strategy to efficiently treat DYT2.


Action Potentials/physiology , Calcium Signaling/physiology , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/physiopathology , Hippocalcin/genetics , Mutation/physiology , Animals , Animals, Newborn , Cells, Cultured , Dystonia Musculorum Deformans/metabolism , Female , HEK293 Cells , Hippocalcin/metabolism , Hippocampus/cytology , Hippocampus/physiology , Humans , Male , Rats , Rats, Wistar
7.
Neurobiol Dis ; 127: 114-130, 2019 07.
Article En | MEDLINE | ID: mdl-30763678

Parkinson's disease (PD) is the second most frequent neurodegenerative disorder in the old population. Among its monogenic variants, a frequent cause is a mutation in the Parkin gene (Prkn). Deficient function of Parkin triggers ubiquitous mitochondrial dysfunction and inflammation in the brain, but it remains unclear how selective neural circuits become vulnerable and finally undergo atrophy. We attempted to go beyond previous work, mostly done in peripheral tumor cells, which identified protein targets of Parkin activity, an ubiquitin E3 ligase. Thus, we now used aged Parkin-knockout (KO) mouse brain for a global quantification of ubiquitylated peptides by mass spectrometry (MS). This approach confirmed the most abundant substrate to be VDAC3, a mitochondrial outer membrane porin that modulates calcium flux, while uncovering also >3-fold dysregulations for neuron-specific factors. Ubiquitylation decreases were prominent for Hippocalcin (HPCA), Calmodulin (CALM1/CALML3), Pyruvate Kinase (PKM2), sodium/potassium-transporting ATPases (ATP1A1/2/3/4), the Rab27A-GTPase activating protein alpha (TBC1D10A) and an ubiquitin ligase adapter (DDB1), while strong increases occurred for calcium transporter ATP2C1 and G-protein subunits G(i)/G(o)/G(Tr). Quantitative immunoblots validated elevated abundance for the electrogenic pump ATP1A2, for HPCA as neuron-specific calcium sensor, which stimulates guanylate cyclases and modifies axonal slow afterhyperpolarization (sAHP), and for the calcium-sensing G-protein GNA11. We assessed if compensatory molecular regulations become insufficient over time, leading to functional deficits. Patch clamp experiments in acute Parkin-KO brain slices indeed revealed alterations of the electrophysiological properties in aged noradrenergic locus coeruleus (LC) neurons. LC neurons of aged Parkin-KO brain showed an acceleration of the spontaneous pacemaker frequency, a reduction in sAHP and shortening of action potential duration, without modulation of KCNQ potassium currents. These findings indicate altered calcium-dependent excitability in a PARK2 model of PD, mediated by diminished turnover of potential Parkin targets such as ATP1A2 and HPCA. The data also identified further novel Parkin substrate candidates like SIRT2, OTUD7B and CUL5. Our elucidation of neuron-specific mechanisms of PD pathogenesis helps to explain the known exceptional susceptibility of noradrenergic and dopaminergic projections to alterations of calcium homeostasis and its mitochondrial buffering.


Adrenergic Neurons/metabolism , Brain/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Hippocalcin/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Mass Spectrometry , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Patch-Clamp Techniques , Ubiquitin-Protein Ligases/genetics , Voltage-Dependent Anion Channels/metabolism
8.
Mol Neurobiol ; 56(5): 3069-3078, 2019 May.
Article En | MEDLINE | ID: mdl-30091036

Exercise increases the levels of neurogenic factors and enhances neurogenesis, memory, and learning. However, the molecular link between exercise and neurogenesis is not clear. The purpose of this study was to examine the effects of exercise intensity on cognitive function and protein expression in the hippocampus of old mice. To compare the effects of aerobic exercise intensity on cognition in old mice, we exposed 18-month-old mice to low- and moderate-intensity treadmill exercise for 4 weeks. Moderate-intensity exercise improved cognitive function in the old mice, while low-intensity exercise did not. To investigate the underlying mechanisms, two-dimensional electrophoresis was used to examine protein expression. Using peptide fingerprinting mass spectrometry, we identified 19 proteins that were upregulated in the hippocampus following exercise training, and seven of these proteins were normalized by the control value. Among them, the levels of 14-3-3 zeta and heat shock protein 70, which have been shown to be induced by exercise training and related to neurogenesis, were dramatically increased by moderate exercise. Hippocalcin, α-spectrin, ovarian tumor domain-containing ubiquitin aldehyde-binding protein 1 (otub1), mu-crystallin, serine racemase, and rho GDP dissociation inhibitor 1, which are related to neurogenesis, neuroprotection, and synaptic strength, were upregulated in the hippocampus by moderate exercise. In addition, we confirmed that neurogenic markers, including doublecortin and the neuronal nuclei antigen, and hippocalcin, otub1, and spectrin-α are potential molecular links between hippocampal neurogenesis and exercise in the elderly. Thus, these results showed that steady moderate-intensity exercise delayed the declines in cognitive function in the elderly through the activation of multiple factors.


Cognition , Cysteine Endopeptidases/metabolism , Hippocalcin/metabolism , Hippocampus/metabolism , Neurogenesis , Physical Conditioning, Animal , Spectrin/metabolism , Up-Regulation , Aging/metabolism , Animals , Biomarkers/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Nerve Tissue Proteins/metabolism
9.
Oncogene ; 38(12): 2192-2205, 2019 03.
Article En | MEDLINE | ID: mdl-30467379

Although cell polarity plays an important role in epithelial tumorigenesis, the consequence of polarity protein loss in prostatic tumorigenesis and the underlying mechanisms remain unclear. Using conditional knockout mouse models, we found in the current study that loss of polarity protein Par3 increases prostatic epithelial cell growth, elevates symmetrical cell divisions in basal cells, and randomizes spindle orientation in luminal cells, causing the development of high-grade prostatic intraepithelial neoplasia (PIN). Mechanistically, loss of Par3 dissociates the Par3/merlin/Lats1 complex, consequently inhibiting phosphorylation of Lats1 to attenuate the Hippo pathway. Furthermore, attenuated Hippo pathway enhances nuclear translocation of Yes-associated protein (YAP), which promotes cell proliferation and symmetrical cell divisions through transcriptional activation of Ki-67 and Sox2. In addition, Lats1 dephosphorylation impairs its interaction with G protein signaling modulator 2 (GPSM2, which is also known as LGN) that causes randomization of spindle orientation in luminal cells. Interestingly, co-deletion of Par3 and Lats1 for complete blockade of the Hippo pathway in mice results in prostate tumor initiation, whereas co-deletion of Par3 and YAP for disrupting YAP nuclear translocation reverses the phenotypes to a relatively normal state. Therefore, our findings highlight combination of Par3 loss and blockade of the Hippo pathway as a novel mechanism for prostatic tumorigenesis.


Carcinogenesis/genetics , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Division , Gene Knockout Techniques , Prostatic Neoplasms/pathology , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Epithelial Cells/pathology , Hippo Signaling Pathway , Hippocalcin/metabolism , Male , Mice , Neoplasm Grading , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , YAP-Signaling Proteins
10.
J Cell Physiol ; 234(4): 3775-3789, 2019 04.
Article En | MEDLINE | ID: mdl-30256386

Heatstroke is a devastating condition that is characterized by severe hyperthermia and central nervous system dysfunction. However, the mechanism of thermoregulatory center dysfunction of the hypothalamus in heatstroke is unclear. In this study, we established a heatstroke mouse model and a heat-stressed neuronal cellular model on the pheochromocytoma-12 (PC12) cell line. These models revealed that HS promoted obvious neuronal injury in the hypothalamus, with high pathological scores. In addition, PC12 cell apoptosis was evident by decreased cell viability, increased caspase-3 activity, and high apoptosis rates. Furthermore, 14 differentially expressed proteins in the hypothalamus were analyzed by fluorescence two-dimensional difference gel electrophoresis and identified by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Expression changes in hippocalcin (HPAC), a downregulated neuron-specific calcium-binding protein, were confirmed in the hypothalamus of the heatstroke mice and heat-stressed PC12 cells by immunochemistry and western blot. Moreover, HPAC overexpression and HPAC-targeted small interfering RNA experiments revealed that HPAC functioned as an antiapoptotic protein in heat-stressed PC12 cells and hypothalamic injury. Lastly, ulinastatin (UTI), a cell-protective drug that is clinically used to treat patients with heatstroke, was used in vitro and in vivo to confirm the role of HPAC; UTI inhibited heat stress (HS)-induced downregulation of HPAC expression, protected hypothalamic neurons and PC12 cells from HS-induced apoptosis and increased heat tolerance in the heatstroke animals. In summary, our study has uncovered and demonstrated the protective role of HPAC in heatstroke-induced hypothalamic injury in mice.


Apoptosis , Brain Diseases/metabolism , Heat Stroke/metabolism , Hippocalcin/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Proteomics , Animals , Apoptosis/drug effects , Brain Diseases/etiology , Brain Diseases/pathology , Brain Diseases/prevention & control , Disease Models, Animal , Glycoproteins/pharmacology , Heat Stroke/complications , Heat Stroke/drug therapy , Hippocalcin/genetics , Hypothalamus/drug effects , Hypothalamus/pathology , Male , Mice, Inbred BALB C , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , PC12 Cells , Proteomics/methods , Rats , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Two-Dimensional Difference Gel Electrophoresis
11.
PLoS One ; 13(4): e0194031, 2018.
Article En | MEDLINE | ID: mdl-29694385

Estimations of intracellular concentrations of fluorescently-labeled molecules within living cells are very important for guidance of biological experiments and interpretation of their results. Here we propose a simple and universal approach for such estimations. The approach is based upon common knowledge that the dye fluorescence is directly proportional to its quantum yield and the number of its molecules and that a coefficient of proportionality is determined by spectral properties of the dye and optical equipment used to record fluorescent signals. If two fluorescent dyes are present in the same volume, then a ratio of their concentrations is equal to a ratio of their fluorescence multiplied by some dye- and equipment-dependent coefficient. Thus, if the coefficient and concentration of one dye is known then the concentration of another dye can be determined. Here we have demonstrated how to calculate this coefficient (called a ratio factor) and how to use it for concentration measurements of fluorescently tagged molecules. As an example of how this approach can be used, we estimated a concentration of exogenously expressed neuronal Ca2+ sensor protein, hippocalcin, tagged by a fluorescent protein in a dendritic tree of rat hippocampal neurons loaded via a patch pipette with Alexa Fluor dye of known concentration. The new approach should allow performing a fast, inexpensive and reliable quantitative analysis of fluorescently-labeled targets in different parts of living cells.


Fluorescent Dyes/metabolism , Hippocalcin/metabolism , Microscopy, Fluorescence/methods , Neurons/metabolism , Spectrometry, Fluorescence/methods , Animals , Cytoplasm/metabolism , Hippocampus/metabolism , Rats
12.
Neurosci Lett ; 662: 189-194, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-29061397

Hyperglycemia is a major risk factor for stroke and increases brain damage during ischemic stroke. Hyperglycemia increases the intracellular calcium concentration after ischemic injury, thereby triggering neuronal cell death. Calcium binding proteins, including hippocalcin and parvalbumin, are critical regulators of intracellular calcium levels. This study aimed to investigate whether hyperglycemic conditions affect hippocalcin and parvalbumin expression during ischemic brain injury. Male adult rats were treated intraperitoneally with streptozotocin (40mg/kg) to induce hyperglycemia. Four weeks later, cerebral ischemic injury was induced via surgical middle cerebral artery occlusion (MCAO). Cerebral cortex samples were collected 24h after MCAO. A proteomic approach showed that the protein levels of hippocalcin and parvalbumin were significantly decreased in streptozotocin-treated animals with MCAO injury compared to streptozotocin-treated animals and animals that underwent MCAO alone. Reverse transcription-PCR and Western blot analyses clearly confirmed the decreased levels of these proteins. These decreases indicate dysregulation of the intracellular calcium balance and induction of cell death. Thus, these results suggest that significantly decreased levels of hippocalcin and parvalbumin exacerbate neuronal cell death in diabetic animals with ischemic brain injury.


Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Diabetes Complications/metabolism , Hippocalcin/metabolism , Parvalbumins/metabolism , Animals , Brain Ischemia/complications , Brain Ischemia/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Male , Rats, Sprague-Dawley , Streptozocin
13.
Hum Mol Genet ; 26(13): 2426-2435, 2017 07 01.
Article En | MEDLINE | ID: mdl-28398555

Dystonia is a neurological movement disorder that forces the body into twisting, repetitive movements or sometimes painful abnormal postures. With the advent of next-generation sequencing technologies, the homozygous mutations T71N and A190T in the neuronal calcium sensor (NCS) hippocalcin were identified as the genetic cause of primary isolated dystonia (DYT2 dystonia). However, the effect of these mutations on the physiological role of hippocalcin has not yet been elucidated. Using a multidisciplinary approach, we demonstrated that hippocalcin oligomerises in a calcium-dependent manner and binds to voltage-gated calcium channels. Mutations T71N and A190T in hippocalcin did not affect stability, calcium-binding affinity or translocation to cellular membranes (Ca2+/myristoyl switch). We obtained the first crystal structure of hippocalcin and alignment with other NCS proteins showed significant variability in the orientation of the C-terminal part of the molecule, the region expected to be important for target binding. We demonstrated that the disease-causing mutations did not affect the structure of the protein, however both mutants showed a defect in oligomerisation. In addition, we observed an increased calcium influx in KCl-depolarised cells expressing mutated hippocalcin, mostly driven by N-type voltage-gated calcium channels. Our data demonstrate that the dystonia-causing mutations strongly affect hippocalcin cellular functions which suggest a central role for perturbed calcium signalling in DYT2 dystonia.


Dystonia/genetics , Hippocalcin/genetics , Hippocalcin/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Calcium-Binding Proteins/genetics , Cell Culture Techniques , Cell Membrane/metabolism , Dystonic Disorders , Hippocalcin/physiology , Humans , Mutation , Myristic Acid/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism
14.
Stem Cell Reports ; 8(1): 95-111, 2017 01 10.
Article En | MEDLINE | ID: mdl-28017654

Hippocalcin (HPCA) is a calcium-binding protein that is restricted to nervous tissue and contributes to neuronal activity. Here we report that, in addition to inducing neurogenesis, HPCA inhibits astrocytic differentiation of neural stem cells. It promotes neurogenesis by regulating protein kinase Cα (PKCα) activation by translocating to the membrane and binding to phosphoinositide-dependent protein kinase 1 (PDK1), which induces PKCα phosphorylation. We also found that phospholipase D1 (PLD1) is implicated in the HPCA-mediated neurogenesis pathway; this enzyme promotes dephosphorylation of signal transducer and activator of transcription 3 (STAT3[Y705]), which is necessary for astrocytic differentiation. Moreover, we found that the SH2-domain-containing tyrosine phosphatase 1 (SHP-1) acts upstream of STAT3. Importantly, this SHP-1-dependent STAT3-inhibitory mechanism is closely involved in neurogenesis and suppression of gliogenesis by HPCA. Taken together, these observations suggest that HPCA promotes neuronal differentiation through activation of the PKCα/PLD1 cascade followed by activation of SHP-1, which dephosphorylates STAT3(Y705), leading to inhibition of astrocytic differentiation.


Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation/genetics , Hippocalcin/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Calcium/metabolism , Gene Expression , Hippocalcin/metabolism , Models, Biological , Neurogenesis , Neurons/cytology , Neurons/metabolism , Phospholipase D/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Tubulin/genetics
15.
Aquat Toxicol ; 181: 67-75, 2016 Dec.
Article En | MEDLINE | ID: mdl-27816759

Benzotriazole (BT) is a high-production volume chemical which has been ubiquitously detected in aquatic environments. Although adverse effects from acute and chronic exposure to BT have been reported, the neurotoxic effect of BT and the mechanisms of toxicity are not well documented. In this study, adult female Chinese rare minnow (Gobiocypris rarus) were exposed to 0.05, 0.5, and 5mg/L BT for 28days. The brain proteome showed that BT exposure mainly involved in metabolic process, signal transduction, stress response, cytoskeleton, and transport. Pathway analysis revealed that cellular processes affected by BT included cellular respiration, G-protein signal cascades, Ca2+-dependent signaling, cell cycle and apoptosis. Moreover, data on relative mRNA levels demonstrated that genes related to these toxic pathways were also significantly affected by BT. Furthermore, proteins affected by BT such as CKBB, GS, HPCA, VDAC1, and FLOT1A are associated with neurological disorders. Therefore, our finding suggested that BT induced molecular responses in the brain and could provide new insight into BT neurotoxicity in Chinese rare minnow.


Brain/metabolism , Cyprinidae/metabolism , Proteome/drug effects , Triazoles/toxicity , Water Pollutants, Chemical/toxicity , Animals , China , Cluster Analysis , Cyprinidae/growth & development , Electrophoresis, Gel, Two-Dimensional , Female , Fish Proteins/genetics , Fish Proteins/metabolism , Hippocalcin/genetics , Hippocalcin/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteome/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism , Water Pollutants, Chemical/chemistry
16.
Cell Biochem Biophys ; 74(4): 449-457, 2016 Dec.
Article En | MEDLINE | ID: mdl-27372904

Visinin-like proteins (VILIPs) belong to the calcium sensor protein family. VILIP-1 has been examined as a cerebrospinal fluid biomarker and as a potential indicator for cognitive decline in Alzheimer's disease (AD). However, little is known about VILIP-3 protein biochemistry. We performed co-immunoprecipitation experiments to examine whether VILIP-3 can interact with reduced nicotine adenine dinucleotide (NADH)-cytochrome b 5 reductase. We also evaluated the specificity of cytochrome b 5 within the visinin-like protein subfamily and identified cytochrome P450 isoforms in the brain. In this study, we show that cytochrome b 5 has an affinity for hippocalcin, neurocalcin-δ, and VILIP-3, but not visinin-like protein-1. VILIP-3 was also shown to interact with NADH-cytochrome b 5 reductase in a Ca2+-dependent manner. These results suggest that VILIP-3, hippocalcin, and neurocalcin-δ provide a Ca2+-dependent modulation to the NADH-dependent microsomal electron transport. The results also suggest that future therapeutic strategies that target calcium-signaling pathways and VILIPs may be of value.


Calcium-Binding Proteins/metabolism , Cytochrome-B(5) Reductase/metabolism , Cytochromes b5/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Calcium/chemistry , Calcium/metabolism , Calcium-Binding Proteins/genetics , Cytochrome P-450 CYP4A/metabolism , Cytochrome-B(5) Reductase/chemistry , Cytochromes b5/chemistry , HEK293 Cells , Hippocalcin/chemistry , Hippocalcin/metabolism , Humans , Immunoprecipitation , Ions/chemistry , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurocalcin/chemistry , Neurocalcin/metabolism , Plasmids/genetics , Plasmids/metabolism , Protein Binding , Sequence Alignment
17.
Biophys J ; 110(5): 1089-98, 2016 Mar 08.
Article En | MEDLINE | ID: mdl-26958886

The slow afterhyperpolarization (sAHP) is a calcium-activated potassium conductance with critical roles in multiple physiological processes. Pharmacological and genetic data suggest that KCNQ channels partly mediate the sAHP. However, these channels are not typically open within the observed voltage range of the sAHP. Recent work has shown that the sAHP is gated by increased PIP2 levels, which are generated downstream of calcium binding by neuronal calcium sensors such as hippocalcin. Here, we examined whether changes in PIP2 levels could shift the voltage-activation range of KCNQ channels. In HEK293T cells, expression of the PIP5 kinase PIPKIγ90, which increases global PIP2 levels, shifted the KCNQ voltage activation to within the operating range of the sAHP. Further, the sensitivity of this effect on KCNQ3 channels appeared to be higher than that on KCNQ2. Therefore, we predict that KCNQ3 plays an essential role in maintaining the sAHP under low PIP2 conditions. In support of this notion, we find that sAHP inhibition by muscarinic receptors that increase phosphoinositide turnover in neurons is enhanced in Kcnq3-knockout mice. Likewise, the presence of KCNQ3 is essential for maintaining the sAHP when hippocalcin is ablated, a condition that likely impairs PIP2 generation. Together, our results establish the relationship between PIP2 and the voltage dependence of cortical KCNQ channels (KCNQ2/3, KCNQ3/5, and KCNQ5), and suggest a possible mechanism for the involvement of KCNQ channels in the sAHP.


Cerebral Cortex/metabolism , Ion Channel Gating , KCNQ3 Potassium Channel/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Carbamates/pharmacology , Female , HEK293 Cells , Hippocalcin/metabolism , Humans , KCNQ3 Potassium Channel/deficiency , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phenylenediamines/pharmacology , Pyramidal Cells/metabolism
18.
Protein Expr Purif ; 123: 35-41, 2016 07.
Article En | MEDLINE | ID: mdl-27001424

Hippocalcin is a 193 aa protein that is a member of the neuronal calcium sensor protein family, whose functions are regulated by calcium. Mice that lack the function of this protein are compromised in the long term potentiation aspect of memory generation. Recently, mutations in the gene have been linked with dystonia in human. The protein has no intrinsic enzyme activity but is known to bind to variety of target proteins. Very little information is available on how the protein executes its critical role in signaling pathways, except that it is regulated by binding of calcium. Further delineation of its function requires large amounts of pure protein. In this report, we present a single-step purification procedure that yields high quantities of the bacterially expressed, recombinant protein. The procedure may be adapted to purify the protein from inclusion bodies or cytosol in its myristoylated or non-myristoylated forms. MALDI-MS (in source decay) analyses demonstrates that the myristoylation occurs at the glycine residue. The protein is also biologically active as measured through tryptophan fluorescence, mobility shift and guanylate cyclase activity assays. Thus, further analyses of hippocalcin, both structural and functional, need no longer be limited by protein availability.


Escherichia coli/genetics , Hippocalcin/genetics , Hippocalcin/isolation & purification , Animals , Chromatography, Liquid , Gene Expression , Genetic Vectors/genetics , Hippocalcin/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Rats , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Vet Sci ; 17(2): 137-44, 2016 Jun 30.
Article En | MEDLINE | ID: mdl-26435544

Hippocalcin participates in the maintenance of neuronal calcium homeostasis. In the present study, we examined the time-course changes of neuronal degeneration and hippocalcin protein level in the mouse hippocampus following pilocarpine-induced status epilepticus (SE). Marked neuronal degeneration was observed in the hippocampus after SE in a time-dependent manner, although neuronal degeneration differed according to the hippocampal subregions. Almost no hippocalcin immunoreactivity was detected in the pyramidal neurons of the cornu ammonis 1 (CA1) region from 6 h after SE. However, many pyramidal neurons in the CA2 region showed hippocalcin immunoreactivity until 24 h after SE. In the CA3 region, only a few hippocalcin immunoreactive cells were observed at 12 h after SE, and almost no hippocalcin immunoreactivity was observed in the pyramidal neurons from 24 h after SE. Hippocalcin immunoreactivity in the polymorphic cells of the dentate gyrus was markedly decreased from 6 h after SE. In addition, hippocalcin protein level in the hippocampus began to decrease from 6 h after SE, and was significantly decreased at 24 h and 48 h after pilocarpine-induced SE. These results indicate that marked reduction of hippocalcin level may be closely related to neuronal degeneration in the hippocampus following pilocarpine-induced SE.


Gene Expression Regulation , Hippocalcin/genetics , Hippocampus/metabolism , Nerve Degeneration/physiopathology , Status Epilepticus/physiopathology , Animals , Gene Expression Regulation/drug effects , Hippocalcin/metabolism , Male , Mice , Mice, Inbred ICR , Nerve Degeneration/chemically induced , Pilocarpine/pharmacology , Status Epilepticus/chemically induced , Time Factors
20.
Exp Eye Res ; 150: 122-34, 2016 09.
Article En | MEDLINE | ID: mdl-26521765

PURPOSE: To characterize the relationship between fundus autofluorescence (FAF), Optical Coherence Tomography (OCT) and immunohistochemistry (IHC) over the course of chronic retinal degeneration in the P23H rat. METHODS: Homozygous albino P23H rats, Sprague-Dawley (SD) rats as controls and pigmented Long Evans (LE) rats were used. A Spectralis HRA OCT system was used for scanning laser ophthalmoscopy (SLO) imaging OCT and angiography. To determine FAF, fluorescence was excited using diode laser at 488 nm. A fast retina map OCT was performed using the optic nerve as a landmark. IHC was performed to correlate with the findings of OCT and FAF changes. RESULTS: During the course of retinal degeneration, the FAF pattern evolved from some spotting at 2 months old to a mosaic of hyperfluorescent dots in rats 6 months and older. Retinal thicknesses progressively diminished over the course of the disease. At later stages of degeneration, OCT documented changes in the retinal layers, however, IHC better identified the cell loss and remodeling changes. Angiography revealed attenuation of the retinal vascular plexus with time. CONCLUSION: We provide for the first time a detailed long-term analysis of the course of retinal degeneration in P23H rats using a combination of SLO and OCT imaging, angiography, FAF and IHC. Although, the application of noninvasive methods enables longitudinal studies and will decrease the number of animals needed for a study, IHC is still an essential tool to identify retinal changes at the cellular level.


Fluorescein Angiography/methods , Hippocalcin/metabolism , Immunohistochemistry/methods , Retinal Degeneration , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence/methods , Visual Acuity , Animals , Disease Models, Animal , Fundus Oculi , Humans , Rats , Retinal Degeneration/diagnosis , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/metabolism , Time Factors
...