Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Histochem Cell Biol ; 157(3): 359-369, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35024954

ABSTRACT

High-mobility group box 2 (HMGB2) is a chromatin-associated protein that is an important regulator of gene transcription, recombination, and repair processes. The functional importance of HMGB2 has been reported in various organs, including the testis, heart, and cartilage. However, its role in the ovary is largely unknown. In this study, ovary tissues from wild-type (WT) and HMGB2-knock-out (KO) mice were examined by histopathological staining and immunohistochemistry. The ovary size and weight were significantly lower in HMGB2-KO mice than in age-matched WT littermates. Histopathological analysis revealed ovarian atrophy and progressive fibrosis in 10-month-old HMGB2-KO mouse ovaries. Compared to age-matched WT mice, the numbers of oocytes and developing follicles were significantly decreased at 2 months of age and were completely depleted at 10 months of age in HMGB2-KO mice. Immunohistochemistry revealed the expression of HMGB2 in the granulosa cells of developing follicles, oocytes, some corpora lutea, and stromal cells. Importantly, HMGB2-positive cells were co-localized with estrogen receptor beta (ERß), but not ERα. Estrogen response element-binding activity was demonstrated by southwestern histochemistry, and it was decreased in HMGB2-KO mouse ovaries. Cell proliferation activity was also decreased in HMGB2-KO mouse ovaries in parallel with the decreased folliculogenesis. These results indicated that the depletion of HMGB2 induced ovarian atrophy that was characterized by a decreased ovarian size and weight, progressive fibrosis, as well as decreased oocytes and folliculogenesis. In conclusion, we demonstrated the crucial role of HMGB2 in mouse ovarian folliculogenesis through ERß expression.


Subject(s)
Estrogen Receptor beta , HMGB2 Protein , Animals , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/analysis , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Granulosa Cells , HMGB2 Protein/analysis , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , Mice , Mice, Knockout , Ovary/metabolism
2.
Anal Chem ; 91(9): 6035-6042, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30990031

ABSTRACT

A new proteomic strategy combining functionalized magnetic nanoparticle affinity probes with mass spectrometry was developed to capture and identify proteins specifically responding to 1,2-d(GpG) intrastrand cisplatin-cross-linked DNA, the major DNA lesion caused by cisplatin and thought to induce apoptosis. A 16-mer oligodeoxynucleotide (ODN) duplex and its cisplatin-cross-linked adduct were immobilized on magnetic nanoparticles via click reaction, respectively, to fabricate negative and positive affinity probes which were very stable in cellular protein extracts due to the excellent bio-orthogonality of click chemistry and the inertness of covalent triazole linker. Quantitative mass spectrometry results unambiguously revealed the predominant binding of HMGB1 and HMGB2, the well-established specific binders of 1,2-cisplatin-cross-linked DNA, to the cisplatin-cross-linked ODN, thus validating the accuracy and reliability of our strategy. Furthermore, 5 RNA or single-stranded DNA binding proteins, namely, hnRNP A/B, RRP44, RL30, RL13, and NCL, were demonstrated to recognize specifically the cisplatinated ODN, indicating the significantly unwound ODN duplex by cisplatin cross-linking. In contrast, the binding of a transcription factor TFIIFa to DNA was retarded due to cisplatin damage, implying that the cisplatin lesion stalls DNA transcription. These findings promote understanding in the cellular responses to cisplatin-damaged DNA and inspire further precise elucidation of the action mechanism of cisplatin.


Subject(s)
Cisplatin/pharmacology , DNA/drug effects , HMGB1 Protein/analysis , HMGB2 Protein/analysis , Proteomics , DNA Damage , Humans , MCF-7 Cells , Magnetite Nanoparticles/chemistry , Mass Spectrometry , Molecular Structure , Tumor Cells, Cultured
3.
Circ J ; 83(2): 368-378, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30487376

ABSTRACT

BACKGROUND: The rapid increase in the number of heart failure (HF) patients in parallel with the increase in the number of older people is receiving attention worldwide. HF not only increases mortality but decreases quality of life, creating medical and social problems. Thus, it is necessary to define molecular mechanisms underlying HF development and progression. HMGB2 is a member of the high-mobility group superfamily characterized as nuclear proteins that bind DNA to stabilize nucleosomes and promote transcription. A recent in vitro study revealed that HMGB2 loss in cardiomyocytes causes hypertrophy and increases HF-associated gene expression. However, it's in vivo function in the heart has not been assessed. Methods and Results: Western blotting analysis revealed increased HMGB2 expression in heart tissues undergoing pressure overload by transverse aorta constriction (TAC) in mice. Hmgb2 homozygous knockout (Hmgb2-/-) mice showed cardiac dysfunction due to AKT inactivation and decreased sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a activity. Compared to wild-type mice, Hmgb2-/- mice had worsened cardiac dysfunction after TAC surgery, predisposing mice to HF development and progression. CONCLUSIONS: This study demonstrates that upregulation of cardiac HMGB2 is an adaptive response to cardiac stress, and that loss of this response could accelerate cardiac dysfunction, suggesting that HMGB2 plays a cardioprotective role.


Subject(s)
HMGB2 Protein/analysis , Heart Failure/etiology , Animals , Blotting, Western , Cardiotonic Agents/analysis , Cardiotonic Agents/pharmacology , Constriction, Pathologic/complications , HMGB2 Protein/genetics , HMGB2 Protein/pharmacology , Heart Failure/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
4.
J Toxicol Sci ; 43(6): 359-367, 2018.
Article in English | MEDLINE | ID: mdl-29877212

ABSTRACT

Screening prostatic carcinogens is time-consuming due to the time needed to induce preneoplastic and neoplastic lesions. To overcome this, we investigated alternative molecular markers for detection of prostatic carcinogens in a short period in rats. After treatment with 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), expression of high-mobility group protein B2 (HMGB2) was up-regulated in rat ventral prostate. To evaluate the applicability of HMGB2 in the early detection of carcinogenicity of chemicals using animal models, we examined HMGB2 expression in prostate of rats. Six-week-old male F344 rats were gavaged for four weeks with a total of eight individual chemicals, divided into two categories based on prostate carcinogenicity. Animals were sacrificed at the end of the study and HMGB2 immunohistochemistry was performed. HMGB2 expression in least one prostate lobe was significantly increased by all four prostate carcinogens compared with the controls. In contrast, the four chemicals that were not carcinogenic in the prostate did not cause HMGB2 up-regulation. Additionally, high HMGB2 expression in neoplastic lesions in both rat and human was detected. Therefore HMGB2 expression may be a good screening tool for the identification of potential of prostate carcinogens.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinogens/analysis , Early Detection of Cancer , Gene Expression , HMGB2 Protein/analysis , HMGB2 Protein/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Animals , Humans , Immunohistochemistry , Male , Rats, Inbred F344 , Up-Regulation
5.
Proteomics ; 15(2-3): 383-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25327479

ABSTRACT

Dysregulation of protein expression is associated with most diseases including cancer. MS-based proteomic analysis is widely employed as a tool to study protein dysregulation in cancers. Proteins that are differentially expressed in head and neck squamous cell carcinoma (HNSCC) cell lines compared to the normal oral cell line could serve as biomarkers for patient stratification. To understand the proteomic complexity in HNSCC, we carried out iTRAQ-based MS analysis on a panel of HNSCC cell lines in addition to a normal oral keratinocyte cell line. LC-MS/MS analysis of total proteome of the HNSCC cell lines led to the identification of 3263 proteins, of which 185 proteins were overexpressed and 190 proteins were downregulated more than twofold in at least two of the three HNSCC cell lines studied. Among the overexpressed proteins, 23 proteins were related to DNA replication and repair. These included high-mobility group box 2 (HMGB2) protein, which was overexpressed in all three HNSCC lines studied. Overexpression of HMGB2 has been reported in various cancers, yet its role in HNSCC remains unclear. Immunohistochemical labeling of HMGB2 in a panel of HNSCC tumors using tissue microarrays revealed overexpression in 77% (54 of 70) of tumors. The HMGB proteins are known to bind to DNA structure resulting from cisplatin-DNA adducts and affect the chemosensitivity of cells. We observed that siRNA-mediated silencing of HMGB2 increased the sensitivity of the HNSCC cell lines to cisplatin and 5-FU. We hypothesize that targeting HMGB2 could enhance the efficacy of existing chemotherapeutic regimens for treatment of HNSCC. All MS data have been deposited in the ProteomeXchange with identifier PXD000737 (http://proteomecentral.proteomexchange.org/dataset/PXD000737).


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , HMGB2 Protein/genetics , Head and Neck Neoplasms/drug therapy , RNA Interference , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line , Cell Line, Tumor , HMGB2 Protein/analysis , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Proteomics , RNA, Small Interfering/genetics , Squamous Cell Carcinoma of Head and Neck , Tandem Mass Spectrometry
6.
Tumour Biol ; 35(10): 10555-69, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25060178

ABSTRACT

High-mobility group box (HMGB) proteins are ubiquitous, abundant nuclear non-histone chromosomal proteins that play a critical role in binding to distorted DNA structures and subsequently regulating DNA transcription, replication, repair, and recombination. Both HMGB1 and HMGB2 exhibit a high expression in several human cancers and are closely associated with tumor progression and a poor prognosis. However, the expression patterns of these molecules in pancreatic ductal adenocarcinoma (PDAC) remain to be elucidated. As most cases of postoperative relapse of PDAC occur within the first 2 years, the clinical significance of accurate biomarkers is needed. Therefore, we investigated the correlation between the immunohistochemical HMGB1 and HMGB2 expression and the clinicopathological characteristics and prognosis using 62 paraffin-embedded tumor samples obtained from patients with surgically resected PDAC. The HMGB1/2 expression was considered to be positive when 10 % or more of the cancer cells showed positive nuclear, not merely cytoplasmic, staining. Consequently, the expression of HMGB1/2 was observed in 54 (87.1 %) and 31 (50.0 %) patients, respectively. Unexpectedly, a positive HMGB1 expression was found to have a significantly close relationship with a negative HMGB2 expression. The univariate and multivariate analyses demonstrated that the patients with a HMGB1+ and HMGB2- status had markedly lower disease-specific survival rates, especially within the first 2 years postoperatively, whereas those with a HMGB1+ status alone did not. Therefore, the combination of a HMGB1+ and HMGB2- expression potentially predicts a poor prognosis in patients with PDAC, and these new biomarkers may be useful parameters for clinical management in the early postoperative phase.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/pathology , HMGB1 Protein/biosynthesis , HMGB2 Protein/biosynthesis , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Female , Fluorescent Antibody Technique , HMGB1 Protein/analysis , HMGB2 Protein/analysis , Humans , Immunohistochemistry , Immunoprecipitation , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Prognosis , Proportional Hazards Models , RNA, Small Interfering , Transfection
7.
Cancer Invest ; 26(8): 843-51, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18798064

ABSTRACT

High mobility group B (HMGB) chromosomal proteins, which plays important role in cancer and inflammation, were followed at various stages of the squamous cell carcinoma of skin. Present results were analyzed by histopathology, BrdU assay, immunohistochemistry, western blot and RT-PCR, which indicate that at early stages of tumorigenesis, expression of HMGB (B1, B2), raised only by about 20%. However, the advanced (> or =12 weeks) tumors showed significant (> or =80%) increase in HMG levels. Using skin cancer model, we demonstrated that high levels of HMGB directly correlate with the extent of neoplastic changes, and it appears that HMGB is an effective stimulus for cell differentiation, tumor progression, and metastatic invasion.


Subject(s)
Carcinoma, Squamous Cell/chemistry , HMGB1 Protein/analysis , HMGB2 Protein/analysis , Neoplasm Proteins/analysis , Skin Neoplasms/chemistry , Animals , Benzo(a)pyrene , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cell Differentiation , DNA Replication , Disease Progression , Female , HMGB1 Protein/physiology , HMGB2 Protein/physiology , Mice , Neoplasm Invasiveness , Neoplasm Proteins/physiology , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology
8.
Nat Cell Biol ; 9(4): 402-14, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17384639

ABSTRACT

Nuclear dysfunction is a key feature of the pathology of polyglutamine (polyQ) diseases. It has been suggested that mutant polyQ proteins impair functions of nuclear factors by interacting with them directly in the nucleus. However, a systematic analysis of quantitative changes in soluble nuclear proteins in neurons expressing mutant polyQ proteins has not been performed. Here, we perform a proteome analysis of soluble nuclear proteins prepared from neurons expressing huntingtin (Htt) or ataxin-1 (AT1) protein, and show that mutant AT1 and Htt similarly reduce the concentration of soluble high mobility group B1/2 (HMGB1/2) proteins. Immunoprecipitation and pulldown assays indicate that HMGBs interact with mutant AT1 and Htt. Immunohistochemistry showed that these proteins were reduced in the nuclear region outside of inclusion bodies in affected neurons. Compensatory expression of HMGBs ameliorated polyQ-induced pathology in primary neurons and in Drosophila polyQ models. Furthermore, HMGBs repressed genotoxic stress signals induced by mutant Htt or transcriptional repression. Thus, HMGBs may be critical regulators of polyQ disease pathology and could be targets for therapy development.


Subject(s)
HMGB1 Protein/physiology , HMGB2 Protein/physiology , Neurodegenerative Diseases/metabolism , Nuclear Proteins/physiology , Proteomics/methods , Animals , Blotting, Western , Cell Death , Cells, Cultured , Drosophila , Electrophoresis, Gel, Two-Dimensional , HMGB1 Protein/analysis , HMGB1 Protein/metabolism , HMGB2 Protein/analysis , HMGB2 Protein/metabolism , Immunohistochemistry , Immunoprecipitation , Models, Biological , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Protein Binding , Purkinje Cells/cytology , Purkinje Cells/metabolism , RNA, Small Interfering , Rats , Rats, Wistar , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...