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










Publication year range
1.
Exp Neurobiol ; 33(1): 1-17, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38471800

ABSTRACT

Autoimmune encephalitis (AIE) is a type of immunoreactive encephalitic disorder and is recognized as the most prevalent noninfectious encephalitis. Nevertheless, the rarity of definitive AIE diagnosis through biopsy or autopsy represents a significant hurdle to understanding and managing the disease. In this article, we present the pathological findings of AIE and review the literature based on a distinct case of AIE presenting as CD8+ T-lymphocyte predominant encephalitis. We describe the clinical progression, diagnostic imaging, laboratory data, and autopsy findings of an 80-year-old deceased male patient. The patient was diagnosed with pulmonary tuberculosis 6 months before death and received appropriate medications. A week before admission to the hospital, the patient manifested symptoms such as a tendency to sleep, decreased appetite, and confusion. Although the patient temporally improved with medication including correction of hyponatremia, the patient progressed rapidly and died in 6 weeks. The brain tissue revealed lymphocytic infiltration in the gray and white matter, leptomeninges, and perivascular infiltration with a predominance of CD8+ T lymphocytes, suggesting a case of AIE. There was no detectable evidence of viral infection or underlying neoplasm. The autopsy revealed that this patient also had Alzheimer's disease, atherosclerosis, arteriolosclerosis, and aging-related tau astrogliopathy. This report emphasizes the pivotal role of pathological examination in the diagnosis of AIE, especially when serological autoantibody testing is not available or when a patient is suspected of having multiple diseases.

2.
Sci Rep ; 13(1): 6761, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185778

ABSTRACT

This study aimed to find any ambiguous genetic outlier for "oligodendroglioma, IDH-mutant and 1p/19q-codeleted (O_IDH_mut)" and "astrocytoma, IDH-mutant (A_IDH_mut)" and to redefine the genetic landscape and prognostic factors of IDH-mutant gliomas. Next-generation sequencing (NGS) using a brain tumor-targeted gene panel, methylation profiles, and clinicopathological features were analyzed for O_IDH_mut (n = 74) in 70 patients and for A_IDH_mut (n = 95) in 90 patients. 97.3% of O_IDH_mut and 98.9% of A_IDH_mut displayed a classic genomic landscape. Combined CIC (75.7%) and/or FUBP1 (45.9%) mutations were detected in 93.2% and MGMTp methylation in 95.9% of O_IDH_mut patients. In A_IDH_mut, TP53 mutations were found in 86.3% and combined ATRX (82.1%) and TERTp (6.3%) mutations in 88.4%. Although there were 3 confusing cases, NOS (not otherwise specified) category, based on genetic profiles, but they were clearly classified by combining histopathology and DKFZ methylation classifier algorithms. The patients with MYCN amplification and/or CDKN2A/2B homozygous deletion in the A_IDH_mut category had a worse prognosis than those without these gene alterations and MYCN-amplified A_IDH_mut showed the worst prognosis. However, there was no prognostic genetic marker in O_IDH_mut. In histopathologically or genetically ambiguous cases, methylation profiles can be used as an objective tool to avoid a diagnosis of NOS or NEC (not elsewhere classified), as well as for tumor classification. The authors have not encountered a case of true mixed oligoastrocytoma using an integrated diagnosis of histopathological, genetic and methylation profiles. MYCN amplification, in addition to CDKN2A/2B homozygous deletion, should be included in the genetic criteria for CNS WHO grade 4 A_IDH_mut.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Humans , Astrocytoma/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA-Binding Proteins/genetics , Genetic Profile , Glioma/diagnosis , Glioma/genetics , Glioma/pathology , Homozygote , Isocitrate Dehydrogenase/genetics , Mutation , N-Myc Proto-Oncogene Protein/genetics , Prognosis , RNA-Binding Proteins/genetics , Sequence Deletion
3.
Lab Invest ; 103(8): 100159, 2023 08.
Article in English | MEDLINE | ID: mdl-37088465

ABSTRACT

Central neurocytoma (CN) is a low-grade neuronal tumor that mainly arises from the lateral ventricle (LV). This tumor remains poorly understood in the sense that no driver gene aberrations have been identified thus far. We investigated immunomarkers in fetal and adult brains and 45 supratentorial periventricular tumors to characterize the biomarkers, cell of origin, and tumorigenesis of CN. All CNs occurred in the LV. A minority involved the third ventricle, but none involved the fourth ventricle. As expected, next-generation sequencing performed using a brain-tumor-targeted gene panel in 7 CNs and whole exome sequencing in 5 CNs showed no driver mutations. Immunohistochemically, CNs were robustly positive for FGFR3 (100%), SSTR2 (92%), TTF-1 (Nkx2.1) (88%), GLUT-1 (84%), and L1CAM (76%), in addition to the well-known markers of CN, synaptophysin (100%) and NeuN (96%). TTF-1 was also positive in subependymal giant cell astrocytomas (100%, 5/5) and the pituicyte tumor family, including pituicytoma and spindle cell oncocytoma (100%, 5/5). Interestingly, 1 case of LV subependymoma (20%, 1/5) was positive for TTF-1, but all LV ependymomas were negative (0/5 positive). Because TTF-1-positive cells were detected in the medial ganglionic eminence around the foramen of Monro of the fetal brain and in the subventricular zone of the LV of the adult brain, CN may arise from subventricular TTF-1-positive cells undergoing neuronal differentiation. H3K27me3 loss was observed in all CNs and one case (20%) of LV subependymoma, suggesting that chromatin remodeling complexes or epigenetic alterations may be involved in the tumorigenesis of all CNs and some ST-subependymomas. Further studies are required to determine the exact tumorigenic mechanism of CN.


Subject(s)
Glioma, Subependymal , Neurocytoma , Humans , Neurocytoma/genetics , Neurocytoma/pathology , Histones/genetics , Epigenesis, Genetic , Carcinogenesis
4.
J Neuropathol Exp Neurol ; 82(3): 250-260, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36592415

ABSTRACT

The MYB/MYBL1::QKI fusion induces the protooncogene, MYB, and deletes the tumor suppressor gene, QKI. MYB/MYBL1::QKI rearrangement was previously reported only in angiocentric glioma (AG) and diffuse low-grade glioma. This report compares 2 tumors containing the MYB/MYBL1::QKI fusion: a diffuse pediatric-type high-grade glioma (DPedHGG) in an 11-year-old boy and an AG in a 46-year-old woman. We used immunohistochemistry, next-generation sequencing, and methylation profiling to characterize each tumor and compare our findings to the literature on AG and tumors with the MYB/MYBL1::QKI rearrangement. Both tumors were astrocytic with angiocentric patterns. The MYB::QKI fusion-positive DPedHGG, which recurred once, was accompanied by TP53 mutation and amplification of CDK6 and KRAS, suggesting malignant transformation secondary to additional genetic aberrations. The second case was the adult AG with MYBL1::QKI fusion, which mimicked ependymoma based on histopathology and its dot- and ring-like epithelial membrane antigen positivity. Combined with a literature review, our results suggest that MYB/MYBL1 alterations are not limited to low-grade gliomas, including AG. AG is most common in the cerebra of children and adolescents but exceptional cases occur in adults and the acquisition of additional genetic mutations may contribute to high-grade glioma. These cases further demonstrate that molecular characteristics, morphologic features, and clinical context are essential for diagnosis.


Subject(s)
Brain Neoplasms , Ependymoma , Glioma , Male , Adult , Female , Adolescent , Humans , Child , Middle Aged , Neoplasm Recurrence, Local , Glioma/genetics , Glioma/pathology , Mutation , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , RNA-Binding Proteins/genetics
5.
Lab Invest ; 102(2): 160-171, 2022 02.
Article in English | MEDLINE | ID: mdl-34848827

ABSTRACT

Mismatch repair-deficient (MMRD) brain tumors are rare among primary brain tumors and can be induced by germline or sporadic mutations. Here, we report 13 MMRD-associated (9 sporadic and 4 Lynch syndrome) primary brain tumors to determine clinicopathological and molecular characteristics and biological behavior. Our 13 MMRD brain tumors included glioblastoma (GBM) IDH-wildtype (n = 9) including 1 gliosarcoma, astrocytoma IDH-mutant WHO grade 4 (n = 2), diffuse midline glioma (DMG) H3 K27M-mutant (n = 1), and pleomorphic xanthoastrocytoma (PXA) (n = 1). Next-generation sequencing using a brain tumor-targeted gene panel, microsatellite instability (MSI) testing, Sanger sequencing for germline MMR gene mutation, immunohistochemistry of MMR proteins, and clinicopathological and survival analysis were performed. There were many accompanying mutations, suggesting a high tumor mutational burden (TMB) in 77%, but TMB was absent in one case of GBM, IDH-wildtype, DMG, and PXA, respectively. MSH2, MLH1, MSH6, and PMS2 mutations were found in 31%, 31%, 31% and 7% of patients, respectively. MSI-high and MSI-low were found in 50% and 8% of these gliomas, respectively and 34% was MSI-stable. All Lynch syndrome-associated GBMs had MSI-high. In addition, 77% (10/13) had histopathologically multinucleated giant cells. The progression-free survival tended to be poorer than the patients with no MMRD gliomas, but the number and follow-up duration of our patients were insufficient to get statistical significance. In the present study, we found that the most common MMRD primary brain tumor was GBM IDH-wildtype. The genetic profile of MMRD GBM was different from that of conventional GBM. MMRD gliomas with TMB and MSI-H may be sensitive to immunotherapy but resistant to temozolomide. Our findings can help develop better treatment options.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , Mutation , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Biomarkers, Tumor/metabolism , Brain/diagnostic imaging , Brain/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Child , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Pedigree , Temozolomide/therapeutic use
6.
Brain Tumor Pathol ; 39(1): 1-13, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34812989

ABSTRACT

Although ependymomas (EPNs) have similar histopathology, they are heterogeneous tumors with diverse immunophenotypes, genetics, epigenetics, and different clinical behavior according to anatomical locations. We reclassified 141 primary EPNs from a single institute with immunohistochemistry (IHC) and next-generation sequencing (NGS). Supratentorial (ST), posterior fossa (PF), and spinal (SP) EPNs comprised 12%, 41%, and 47% of our cohort, respectively. Fusion genes were found only in ST-EPNs except for one SP-EPN with ZFTA-YAP1 fusion, NF2 gene alterations were found in SP-EPNs, but no driver gene was present in PF-EPNs. Surrogate IHC markers revealed high concordance rates between L1CAM and ZFTA-fusion and H3K27me3 loss or EZHIP overexpression was used for PFA-EPNs. The 7% cut-off of Ki-67 was sufficient to classify EPNs into two-tiered grades at all anatomical locations. Multivariate analysis also delineated that a Ki-67 index was the only independent prognostic factor in both overall and progression-free survivals. The gain of chromosome 1q and CDKN2A/2B deletion were associated with poor outcomes, such as multiple recurrences or extracranial metastases. In this study, we propose a cost-effective schematic diagnostic flow of EPNs by the anatomical location, three biomarkers (L1CAM, H3K27me3, and EZHIP), and a cut-off of a 7% Ki-67 labeling index.


Subject(s)
Ependymoma , Infratentorial Neoplasms , Ki-67 Antigen , Spinal Neoplasms , Supratentorial Neoplasms , Ependymoma/diagnosis , Ependymoma/genetics , Humans , Ki-67 Antigen/genetics , Neural Cell Adhesion Molecule L1 , Oncogene Proteins , Prognosis
7.
Diagn Pathol ; 16(1): 83, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34465349

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by hamartomas in multiple organs associated with germline mutations in TSC1 and TSC2, including exonic, intronic, or mosaic mutations. Gastrointestinal (GI) tract Lymphangioleiomyomatosis (LAM) is an extremely rare manifestation of TSC, with few reported cases. Herein, we aimed to determine the driver mutation, pathogenesis, and relationship of germline and somatic mutations of LAM through whole-genome sequencing (WGS) of the tumor and blood samples and whole transcriptome sequencing (WTS) analysis. CASE PRESENTATION: A nine-year-old girl with a full-blown TSC presented with abdominal masses detected during a routine check-up. Resected intestinal masses were diagnosed as LAM by thorough pathological examination. Interestingly, the LAM presented a somatic TSC2 gene mutation in exon 24 (p.R905W, c.C2713T), and the patient had intron retention by a novel germline mutation in the intron region of TSC2 (chr16:2126489, C > G). CONCLUSION: Our case suggests that intron retention by a single nucleotide intronic mutation of TSC2 is sufficient to develop severe manifestations of TSC, but the development of LAM requires an additional somatic oncogenic mutation of TSC2.


Subject(s)
Biomarkers, Tumor/genetics , Germ-Line Mutation , Intestinal Neoplasms/genetics , Lymphangioleiomyomatosis/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis/genetics , Biopsy , Child , DNA Mutational Analysis , Exons , Female , Humans , Intestinal Neoplasms/diagnostic imaging , Intestinal Neoplasms/pathology , Intestinal Neoplasms/surgery , Introns , Lymphangioleiomyomatosis/diagnostic imaging , Lymphangioleiomyomatosis/pathology , Lymphangioleiomyomatosis/surgery , Tuberous Sclerosis/diagnostic imaging
8.
Mol Neurobiol ; 56(3): 2007-2021, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29982983

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal, adult-onset, progressive neurodegenerative disorder with no known cure. Cu/Zn-superoxide dismutase (SOD1) was the first identified protein associated with familial ALS (fALS). Recently, TAR DNA-binding protein 43 (TDP-43) has been found to be a principal component of ubiquitinated cytoplasmic inclusions in neurons and glia in ALS. However, it remains unclear whether these ALS-linked proteins partly have a shared pathogenesis. Here, we determine the association between mutant SOD1 and the modification of TDP-43 and the relationship of pathologic TDP-43 to neuronal cytotoxicity in SOD1 ALS. In this work, using animal model, human tissue, and cell models, we provide the evidence that the association between the TDP-43 modification and the pathogenesis of SOD1 fALS. We demonstrated an age-dependent increase in TDP-43 C-terminal fragments and phosphorylation in motor neurons and glia of SOD1 mice and SOD1G85S ALS patient. Cytoplasmic TDP-43 was also observed in iPSC-derived motor neurons from SOD1G17S ALS patient. Moreover, we observed that mutant SOD1 interacts with TDP-43 in co-immunoprecipitation assays with G93A hSOD1-transfected cell lines. Mutant SOD1 overexpression led to an increase in TDP-43 modification in the detergent-insoluble fraction in the spinal cord of SOD1 mice and fALS patient. Additionally, we showed cellular apoptosis in response to the interaction of mutant SOD1 and fragment forms of TDP-43. These findings suggest that mutant SOD1 could affect the solubility/insolubility of TDP-43 through physical interactions and the resulting pathological modifications of TDP-43 may be involved in motor neuron death in SOD1 fALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Motor Neurons/metabolism , Mutation , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , DNA-Binding Proteins/genetics , Disease Models, Animal , Humans , Mice , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroglia/metabolism , Neuroglia/pathology , Phosphorylation
9.
Mol Cancer Ther ; 17(4): 825-837, 2018 04.
Article in English | MEDLINE | ID: mdl-29437878

ABSTRACT

The most common therapy for estrogen receptor-positive breast cancer is antihormone therapy, such as tamoxifen. However, acquisition of resistance to tamoxifen in one third of patients presents a serious clinical problem. Polo-like kinase 1 (Plk1) is a key oncogenic regulator of completion of G2-M phase of the cell cycle. We assessed Plk1 expression in five chemoresistant cancer cell types and found that Plk1 and its downstream phosphatase Cdc25c were selectively overexpressed in tamoxifen-resistant MCF-7 (TAMR-MCF-7) breast cancer cells. Real-time monitoring of cell proliferation also showed that TAMR-MCF-7 cells were more sensitive to inhibition of cell proliferation by the ATP-competitive Plk1 inhibitor BI2536 than were the parent MCF-7 cells. Moreover, BI2536 suppressed expression of epithelial-mesenchymal transition marker proteins and 3D spheroid formation in TAMR-MCF-7 cells. Using TAMR-MCF-7 cell-implanted xenograft and spleen-liver metastasis models, we showed that BI2536 inhibited tumor growth and metastasis in vivo Our results suggest that Plk1 could be a novel target for the treatment of tamoxifen-resistant breast cancer. Mol Cancer Ther; 17(4); 825-37. ©2018 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Drug Resistance, Neoplasm , Liver Neoplasms/secondary , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Splenic Neoplasms/secondary , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pteridines/pharmacology , Splenic Neoplasms/drug therapy , Splenic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
10.
J Biophotonics ; 10(1): 118-127, 2017 01.
Article in English | MEDLINE | ID: mdl-26768511

ABSTRACT

We report a two color monitoring of drug-induced cell deaths using total internal reflection fluorescence (TIRF) as a novel method to determine anticancer activity. Instead of cancer cells, breast cancer stem cells (CSCs) were directly tested in the present assay to determine the effective concentration (EC50 ) values of camptothecin and cisplatin. Phosphatidylserine and HMGB1 protein were concurrently detected to observe apoptotic and necrotic cell death induced by anticancer drugs using quantum dot (Qdot)-antibody conjugates. Only 50-to-100 breast CSCs were consumed at each cell chamber due to the high sensitivity of Qdot-based TIRF. The high sensitivity of Qdot-based TIRF, that enables the consumption of a small number of cells, is advantageous for cost-effective large-scale drug screening. In addition, unlike MTT assay, this approach can provide a more uniform range of EC50 values because the average values of single breast CSCs fluorescence intensities are observed to acquire EC50 values as a function of dose. This research successfully demonstrated the possibility that Qdot-based TIRF can be widely used as an improved alternative to MTT assay for the determination of anticancer drug efficacies.


Subject(s)
Cell Death , Microscopy, Fluorescence , Neoplastic Stem Cells/cytology , Quantum Dots , Breast Neoplasms/drug therapy , Camptothecin/pharmacology , Cisplatin/pharmacology , Humans , Tumor Cells, Cultured
11.
Toxicol Lett ; 243: 31-9, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26739636

ABSTRACT

Cancer stem cells (CSCs), known as tumor initiating cells, have become a critically important issue for cancer therapy. Although much research has demonstrated the induction of hepato cellular carcinoma by aflatoxin B1, the formation of hepatocellular CSCs and their quantitative determination is hardly reported. In this work, it was found that hepatocellular CSCs were produced from HepG2 cells by aflatoxin B1-induced mutation, and their amount was quantitatively determined using crosstalk-eliminated multicolor cellular imaging based on quantum dot (Qdot) nanoprobes and an acousto-optical tunable filter (AOTF). Hepatocellular CSCs were acquired via magnetic bead-based sorting and observed using concurrent detection of three different markers: CD133, CD44, and aldehyde dehydrogenase1 (ALDH1). The DNA mutation of HepG2 cells caused by aflatoxin B1 was quantitatively observed via absorbance spectra of aflatoxin B1-8, 9-epoxide-DNA adducts. The percentages of hepatocellular CSCs formed in the entire HepG2 cells were determined to be 9.77±0.65%, 10.9±1.39%, 11.4±1.32%, and 12.8±0.7%, respectively, at 0 µM, 5 µM, 10 µM, and 20 µM of aflatoxin B1. The results matched well with those obtained utilizing flow cytometry. This study demonstrates that aflatoxin mediated mutation induced the conversion of hepatic cancer cell to hepatic CSCs by using a Qdot based constructed multicolor cellular imaging system.


Subject(s)
Aflatoxin B1/toxicity , Aldehyde Dehydrogenase/metabolism , Antigens, CD/metabolism , DNA Adducts/drug effects , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/drug effects , Peptides/metabolism , AC133 Antigen , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Mutation , Neoplastic Stem Cells/pathology
12.
Neurochem Res ; 41(4): 913-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26646002

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating human neurodegenerative disease. The precise pathogenic mechanisms of the disease remain uncertain, and as of yet, there is no effective cure. Human adipose stem cells (hASC) can be easily obtained during operative procedures. hASC have a clinically feasible potential to treat neurodegenerative disorders, since cytosolic extract of hASC contain a number of essential neurotrophic factors. In this study, we investigated effects of hASC extract on the SOD1 G93A mouse model of ALS and in vitro test. Administration of hASC extract improved motor function and prolonged the time until symptom onset, rotarod failure, and death in ALS mice. In the hASC extracts group, choline acetyltransferase immunostaining in the ventral horn of the lumbar spinal cord showed a large number of motor neurons, suggesting normal morphology. The neuroprotective effect of hASC extract in ALS mice was also suggested by western blot analysis of spinal cord extract from ALS mice and in vitro test. hASC extract treatment significantly increased expression of p-Akt, p-CREB, and PGC-1α in SOD1 G93A mouse model and in vitro test. Our results indicated that hASC extract reduced apoptotic cell death and recovered mutant SOD1-induced mitochondrial dysfunction. Moreover, hASC extract reduced mitochondrial membrane potential. In conclusion, we have demonstrated, for the first time, that hASC extract exert a potential therapeutic action in the SOD1 G93A mouse model of ALS and in vitro test. These findings suggest that hASC hold promise as a novel therapeutic strategy for treating ALS.


Subject(s)
Adipose Tissue/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Cell Extracts/pharmacology , Neuroprotective Agents/pharmacology , Stem Cells/metabolism , Adipose Tissue/cytology , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Extracts/therapeutic use , Cell Survival , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Humans , Male , Mice, Transgenic , Mitochondria/metabolism , Motor Neurons/drug effects , Motor Neurons/pathology , Neuroprotective Agents/therapeutic use , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Transcription Factors/metabolism
13.
Biotechnol J ; 11(1): 31-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26709963

ABSTRACT

Semiconductor quantum dots (QD) possess unique optical and electric properties like size-tunable light emission, narrow emission range, high brightness and photostability. Recent research advances have minimized the toxicity of QDs and they are successfully used in in vitro and in vivo imaging. Encapsulation of QDs into polymeric nanoparticles and linking them with targeting ligands enabled the detection of tumors and cancer cells in vivo. QD-antibody conjugates were successfully used in monitoring and diagnosis of HIV and myocardial infarction. Application of near infrared (NIR) QDs was found to minimize the absorption and scattering of light by native tissues thus rendering them suitable in deep tissue analysis. Aggregation and endosomal sequestration of QDs pose major challenges for the effective delivery of QDs to the cell cytosol. Toxicity minimization and effective delivery strategies may further increase their suitability for utilization in disease diagnosis. New synthesis of QDs may provide new types of bioconjugates of QDs to biomolecules, which leads to a variety of applications to many challenged research areas. QDs with narrow emission wavelength ranges are very suitable for monitoring multiple cellular targets simultaneously, and still remain the best known probes for imaging as an alternative to traditional fluorophores in disease diagnosis.


Subject(s)
Neoplasms/diagnosis , Quantum Dots/metabolism , Biocompatible Materials/adverse effects , Biocompatible Materials/metabolism , Diagnostic Imaging/methods , Drug Delivery Systems , Humans , Quantum Dots/adverse effects , Tissue Distribution
14.
Mol Cell Probes ; 29(6): 376-381, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26384954

ABSTRACT

Breast cancer is the major cause of cancer death for women worldwide. Breast cancer patients are treated with chemotherapy and radiotherapy. Although chemotherapy and radiotherapy are applied, some cancer cells still survive. These cells, called cancer stem cell (CSC), exhibit special capabilities, such as drug and radio resistance. The remaining CSC can trigger cancer recurrence. Thus, it is critical to find an effective way to target CSC. Capsaicin has been reported to affect anticancer activity in many cancers. It also has been shown that capsaicin induces apoptosis in the MCF-7 breast cancer cell line. In this study, we demonstrate that capsaicin causes dose-dependent growth disruption in breast CSC and inhibits translocation of notch intracellular membrane domain (NICD) into the nucleus. MCF-7 cells were treated with capsaicin at various concentrations (5 µM, 10 µM, and 20 µM) for 24 h. After capsaicin treatment, it was found that the number of breast CSC (%) decreased as the treatment concentration of capsaicin increased. This result was also confirmed with FACS. NICD translocation to the nucleus and apoptotic cell death of breast CSC were concurrently observed at the single breast CSC level using highly sensitive quantum dot (Qdot)-antibody nanoprobes. The control breast CSCs without the capsaicin treatment were able to translocate NICD into the nucleus. On the other hand, translocation of NICD into the nucleus was not observed in capsaicin-treated cells. In addition, apoptotic cell death was caused when the breast CSC were treated with capsaicin at more than 10 µM. Although many studies have shown that capsaicin produces anticancer activity in cancer cell lines, the present result is the first report to demonstrate that capsaicin is capable of causing breast CSC apoptotic cell death via inhibiting its notch signaling pathway.


Subject(s)
Breast Neoplasms/metabolism , Capsaicin/pharmacology , Neoplastic Stem Cells/drug effects , Quantum Dots/metabolism , Receptors, Notch/metabolism , Single-Cell Analysis/methods , Apoptosis , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Nanoparticles , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects
15.
Exp Neurobiol ; 24(3): 226-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26412972

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by selective degeneration of motor neurons. Mutant superoxide dismutase 1 (SOD1) is often found as aggregates in the cytoplasm in motor neurons of various mouse models and familial ALS patients. The interplay between motor neurons and astrocytes is crucial for disease outcome, but the mechanisms underlying this phenomenon remain unknown. In this study, we investigated whether transient transfection with wild-type and mutant-type SOD1 may lead to amplification of mutant SOD1-mediated toxicity in cortical neurons and astrocytes derived from wild-type and mutant-type (human G93A-SOD1) mice. In transgenic mice expressing either wild- or mutant-type SOD1, we found that green fluorescent protein (GFP)-wtSOD1 was present in the cytoplasm and nuclei of wild-type cortical neurons and astrocytes, whereas GFP-mutant SOD1 was mainly cytoplasmic in wild- and mutant-type cortical neurons and astrocytes. These findings indicate that intracellular propagation of misfolding of GFP-wt or mtSOD1 are possible mediators of toxic processes involved in initiating mislocalization and aggregation. Here, we provide evidence that cytoplasmic aggregates induce apoptosis in G93A-SOD1 mouse cortical neurons and astrocytes and that the toxicity of mutant SOD1 in astrocytes is similar to the pathological effects of ALS on neurons in vitro. Collectively, our results indicate that mtSOD1 probably interacts with wtSOD1 via an unknown mechanism to produce augmented toxicity and may influence aggregate formation and apoptosis.

16.
Biochem Biophys Res Commun ; 464(1): 236-43, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26102026

ABSTRACT

Two DNA/RNA binding proteins, TDP-43 and FUS/TLSU, are involved in RNA processing, and their aberrant mutations induce inherited amyotrophic lateral sclerosis and frontotemporal lobar degeneration with ubiquitinated inclusions. Wild type TDP-43 and FUS (wtTDP-43 and wtFUS) are mainly localized in the nucleus and biochemically interact with the microRNA processing enzyme Drosha. In this study, we investigated Drosha stability in Neuro 2A cells by gain and loss of function studies of wtTDP-43 and wtFUS and cycloheximide mediated protein degradation assay. We also generated three different phosphomimetic mutants of TDP-43 (S379E, S403/404E and S409/410E) by using a site-directed mutagenesis method and examined Drosha stability to elucidate a correlation between the phosphorylated TDP-43 mutants and Drosha stability. Overexpression of wtTDP-43 and/or wtFUS increased Drosha stability in Neuro 2A cells and double knockdown of wtTDP-43 and wtFUS reduced its stability. However, knockdown of wtTDP-43 or wtFUS did not affect Drosha stability in Neuro 2A cells. Interestingly, a phosphomimetic mutant TDP-43 (S409/410E) significantly reduced Drosha stability via prevention of protein-protein interactions between wtFUS and Drosha, and induced cytotoxicity in Neuro 2A cells. Our findings suggest that TDP-43 and FUS controls Drosha stability in Neuro 2A cells and that a phosphomimetic mutant TDP-43 (S409/410E) which is associated with Drosha instability can induce neuronal toxicity.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs/genetics , Neurons/metabolism , Phosphoproteins/genetics , RNA-Binding Protein FUS/genetics , Ribonuclease III/genetics , Animals , Cell Death/genetics , Cell Line, Tumor , Cycloheximide/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Enzyme Stability/genetics , Mice , MicroRNAs/metabolism , Molecular Mimicry , Mutagenesis, Site-Directed , Neurons/drug effects , Neurons/pathology , Phosphoproteins/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Protein FUS/antagonists & inhibitors , RNA-Binding Protein FUS/metabolism , Ribonuclease III/metabolism
17.
Nanoscale Res Lett ; 10: 140, 2015.
Article in English | MEDLINE | ID: mdl-25852430

ABSTRACT

With advancements in nanotechnology, silver has been engineered into a nanometre size and has attracted great research interest for use in the treatment of wounds. Silver nanoparticles (AgNPs) have emerged as a potential alternative to conventional antibiotics because of their potential antimicrobial property. However, AgNPs also induce cytotoxicity, generate reactive oxygen species (ROS), and cause mitochondrial damage to human cells. Pyridoxine possesses antioxidant and cell proliferation activity. Therefore, in the present investigation, a nanosilver-pyridoxine complex (AgPyNP) was synthesized, and its cytotoxicity and immune response was compared with AgNPs in macrophage RAW264.7 cells. Results revealed that AgPyNPs showed less cytotoxicity compared with AgNPs by producing a smaller amount of ROS in RAW264.7 cells. Surprisingly, however, AgPyNPs caused macrophage RAW264.7 cells to secrete a larger amount of interleukin-8 (IL-8) and generate a more active inflammatory response compared to AgNPs. It activated TNF-α, NF-κB p65, and NF-κB p50 to generate a more vigorous immune protection that produces a greater amount of IL-8 compared to AgNPs. Overall findings indicate that AgPyNPs exhibited less cytotoxicity and evoked a greater immune response in macrophage RAW264.7 cells. Thus, it can be used as a better wound-healing agent than AgNPs. Graphical AbstractFigurative representation of the comparison of AgNPs and AgPyNPs in macrophage RAW264.7 cells in terms of cytotoxicity and immune response.

18.
J Nanobiotechnology ; 13: 4, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25623542

ABSTRACT

BACKGROUND: TNF-α is an inflammatory cytokine that plays an important role in insulin resistance observed in obesity and chronic inflammation. Many cellular components involved in insulin signaling cascade are known to be inhibited by TNF-α. Insulin receptor substrate (IRS)-1 is one of the major targets in TNF-α-induced insulin resistance. The serine phosphorylation of IRS-1 enables the inhibition of insulin signaling. Until now, many studies have been conducted to investigate the mechanism of TNF-α-induced insulin resistance based on Western blot. Intracellular protein kinase crosstalk is commonly encountered in inflammation-associated insulin resistance. The crosstalk among the signaling molecules obscures the precise role of kinases in insulin resistance. We have developed a cell lysis-free quantum dots (QDots) multicolor cellular imaging to identify the biochemical role of multiple kinases (p38, JNK, IKKß, IRS1ser, IRS1tyr, GSK3ß, and FOXO1) in inflammation-associated insulin resistance pathway with a single assay in one run. QDot-antibody conjugates were used as nanoprobes to simultaneously monitor the activation/deactivation of the above seven intracellular kinases in HepG2 cells. The effect of the test compounds on the suppression of TNF-α-induced insulin resistance was validated through kinase monitoring. Aspirin, indomethacin, cinnamic acid, and amygdalin were tested. RESULTS: Through the measurement of the glycogen level in HepG2 cell treated with TNF-α, it was found that aspirin and indomethacin increased glycogen levels by almost two-fold compared to amygdalin and cinnamic acid. The glucose production assay proved that cinnamic acid was much more efficient in suppressing glucose production, compared with MAP kinase inhibitors and non-steroidal anti-inflammatory drugs. QDot multicolor cellular imaging demonstrated that amygdalin and cinnamic acid selectively acted via the JNK1-dependent pathway to suppress the inflammation-induced insulin resistance and improve insulin sensitivity. CONCLUSION: The regulatory function of multiple kinases could be monitored concurrently at the cellular level. The developed cellular imaging assay provides a unique platform for the understanding of inflammation and insulin resistance signaling pathways in type II diabetes mellitus and how they regulate each other. The results showed that amygdalin and cinnamic acid inhibit serine phosphorylation of IRS-1 through targeting JNK serine kinase and enhance insulin sensitivity.


Subject(s)
Insulin Resistance , Molecular Imaging/methods , Protein Kinases/analysis , Quantum Dots , Tumor Necrosis Factor-alpha/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies/chemistry , Aspirin/pharmacology , Cinnamates/pharmacology , Forkhead Box Protein O1 , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/metabolism , Glucose/analysis , Glycogen/analysis , Hep G2 Cells/drug effects , Hep G2 Cells/metabolism , Humans , Indomethacin/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Insulin Receptor Substrate Proteins/metabolism , Molecular Targeted Therapy , Protein Kinases/immunology , Protein Kinases/metabolism , Quantum Dots/chemistry , Reproducibility of Results , Serine/metabolism
19.
Anal Chim Acta ; 853: 501-507, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25467496

ABSTRACT

The circulating endothelial progenitor cells (EPCs) in blood of acute myocardial infarction (AMI) patient have been monitored in many previous studies. The number of circulating EPC increases in the blood of patients at onset of the AMI. EPC is originated from bone marrow. It performs vessel regeneration. There are many markers used for detecting EPC. Four of these markers, CD31, CD34, CD45, and CD146, were concurrently detected at the single cell level for the identification of EPC in the present preliminary study. The CD45 negative cell sorting was performed to peripheral blood mononuclear cells (PBMCs) acquired from four AMI patients with a magnetic bead sorter, since, EPCs expressed CD45 negative or dim. The resultant PBMC eluents were treated with quantum-antibody conjugates for the probing four different markers of EPCs and then applied to a high-content single cell imaging cytometer using acousto-optical tunable filter (AOTF). The use of quantum dot, with narrow emission wavelength range and AOTF enabling cellular image at a particular single wavelength, is very advantageous for accurate high-content AMI diagnosis based on simultaneous monitoring of many markers. The number of EPC increased as compared with control in three of four AMI patients. In this approach, two EPC subtypes were found, CD31(+), CD34(+), CD45(-/dim), CD146(-) as early outgrowth EPCs and CD31(+), CD34(+), CD45(-/dim), CD146(+) as late outgrowth EPCs. Patient 1 had CD31(+), CD34(+), CD45(-/dim), CD146(+) cells whose percentage was 4.21% of cells. Patient 2 had 2.38% of CD31(+), CD34(+), CD45(-/dim), CD146(-) cells and patient 3 had 4.28% of CD31(+), CD34(+), CD45(-/dim), CD146(+) cells.


Subject(s)
Antigens, CD/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Myocardial Infarction/pathology , Single-Cell Analysis/methods , Aged , Antibodies/chemistry , Antibodies/immunology , Antigens, CD/blood , Antigens, CD/immunology , Antigens, CD34/immunology , Antigens, CD34/metabolism , Biomarkers/metabolism , CD146 Antigen/immunology , CD146 Antigen/metabolism , Color , Flow Cytometry , Humans , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Quantum Dots/chemistry
20.
Chem Commun (Camb) ; 51(11): 2118-21, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25536409

ABSTRACT

In this study, it was found that breast cancer stem cells (CSCs) are formed from MCF-7 cells by benzo[a]pyrene (BP)-induced mutation. The breast CSCs were detected through simultaneous monitoring of CD44, CD24 and aldehyde dehydrogenase 1 (ALDH1) by hypermulticolor cellular imaging using an acousto-optical tunable filter (AOTF) and quantum dots (Q-dots).


Subject(s)
Benzo(a)pyrene/chemistry , Benzo(a)pyrene/toxicity , Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Quantum Dots/chemistry , Spectrum Analysis , Aldehyde Dehydrogenase 1 Family , CD24 Antigen/metabolism , Carcinogens/chemistry , Carcinogens/toxicity , Cell Separation , Humans , Hyaluronan Receptors/metabolism , Isoenzymes/metabolism , MCF-7 Cells , Molecular Imaging , Mutation/drug effects , Neoplastic Stem Cells/metabolism , Retinal Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...