Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cancer Genomics Proteomics ; 21(4): 414-420, 2024.
Article in English | MEDLINE | ID: mdl-38944423

ABSTRACT

BACKGROUND/AIM: Clear cell carcinoma is a prevalent histological type of ovarian cancer in East Asia, particularly in Japan, known for its resistance to chemotherapeutic agents and poor prognosis. ARID1A gene mutations, commonly found in ovarian clear cell carcinoma (OCCC), contribute to its pathogenesis. Recent data revealed that the ARID1A mutation is related to better outcomes of cancer immunotherapy. Thus, this study aimed to investigate the immunotherapy treatment susceptibility of OCCC bearing ARID1A mutations. MATERIALS AND METHODS: Expression of ARID1A was analyzed using western blotting in ovarian cancer cell lines. OCCC cell lines JHOC-9 and RMG-V were engineered to overexpress NY-ESO-1, HLA-A*02:01, and ARID1A. Sensitivity to chemotherapy and T cell receptor-transduced T (TCR-T) cells specific for NY-ESO-1 was assessed in ARID1A-restored cells compared to ARID1A-deficient wild-type cells. RESULTS: JHOC-9 cells and RMG-V cells showed no expression of ARID1A protein. Overexpression of ARID1A in JHOC-9 and RMG-V cells did not impact sensitivity to gemcitabine. While ARID1A overexpression decreased sensitivity to cisplatin in RMG-V cells, it had no such effect in JHOC-9 cells. ARID1A overexpression reduced the reactivity of NY-ESO-1-specific TCR-T cells, as observed by the IFNγ ESLIPOT assay. CONCLUSION: Cancer immunotherapy is an effective approach to target ARID1A-deficient clear cell carcinoma of the ovary.


Subject(s)
Adenocarcinoma, Clear Cell , DNA-Binding Proteins , Ovarian Neoplasms , T-Lymphocytes, Cytotoxic , Transcription Factors , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Clear Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Cell Line, Tumor , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Membrane Proteins
2.
Cancer Sci ; 115(1): 24-35, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37879364

ABSTRACT

We previously identified papillomavirus binding factor (PBF) as an osteosarcoma antigen recognized by an autologous cytotoxic T lymphocyte clone. Vaccination with PBF-derived peptide presented by HLA-A24 (PBF peptide) elicited strong immune responses. In the present study, we generated T cell receptor-engineered T cells (TCR-T cells) directed against the PBF peptide (PBF TCR-T cells). PBF TCR was successfully transduced into T cells and detected using HLA-A*24:02/PBF peptide tetramer. PBF TCR-T cells generated from a healthy donor were highly expanded and recognized T2-A24 cells pulsed with PBF peptide, HLA-A24+ 293T cells transfected with PBF cDNA, and sarcoma cell lines. To establish an adoptive cell therapy model, we modified the PBF TCR by replacing both α and ß constant regions with those of mice (hybrid PBF TCR). Hybrid PBF TCR-T cells also showed reactivity against T2-A24 cells pulsed with PBF peptide and to HLA-A24+ 293T cells transfected with various lengths of PBF cDNA including the PBF peptide sequence. Subsequently, we generated target cell lines highly expressing PBF (MFH03-PBF [short] epitope [+]) containing PBF peptide with in vivo tumorigenicity. Hybrid PBF TCR-T cells exhibited antitumor effects compared with mock T cells in NSG mice xenografted with MFH03-PBF (short) epitope (+) cells. CD45+ T cells significantly infiltrated xenografted tumors only in the hybrid PBF TCR T cell group and most of these cells were CD8-positive. CD8+ T cells also showed Ki-67 expression and surrounded the CD8-negative tumor cells expressing Ki-67. These findings suggest that PBF TCR-T cell therapy might be a candidate immunotherapy for sarcoma highly expressing PBF.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Mice , CD8-Positive T-Lymphocytes , HLA-A24 Antigen , DNA, Complementary/metabolism , Ki-67 Antigen/metabolism , T-Lymphocytes, Cytotoxic , Peptides , Osteosarcoma/genetics , Epitopes/metabolism , Bone Neoplasms/metabolism , Receptors, Antigen, T-Cell
3.
Cancer Sci ; 114(12): 4511-4520, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37991442

ABSTRACT

Eribulin inhibits microtubule polymerization and improves the overall survival of patients with recurrent metastatic breast cancer. A subgroup analysis revealed a low neutrophil to lymphocyte ratio (NLR) (<3) to be a prognostic factor of eribulin treatment. We thus hypothesized that eribulin might be related to the immune response for breast cancer cells and we analyzed the effects of eribulin on the immune system. Immunohistochemical staining revealed that human leukocyte antigen (HLA) class I expression was increased in clinical samples after eribulin treatment. In vitro assays revealed that eribulin treatment increased HLA class I expression in breast cancer line cells. RNA-sequencing demonstrated that eribulin treatment increased the expression of the NOD-like family CARD domain-containing 5 (NLRC5), a master regulator of HLA class I expression. Eribulin treatment increased the NY-ESO-1-specific T-cell receptor (TCR) transduced T (TCR-T) cell response for New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) overexpressed breast cancer cells. The eribulin and TCR-T combined therapy model revealed that eribulin and immunotherapy using TCR-T cells has a synergistic effect. In summary, eribulin increases the expression of HLA class 1 via HLA class 1 transactivatior NLRC5 and eribulin combination with immunotherapy can be effective for the treatment of breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , NLR Proteins , Caspase Activation and Recruitment Domain , Neoplasm Recurrence, Local , Receptors, Antigen, T-Cell/metabolism , Antigens, Neoplasm , HLA Antigens , Intracellular Signaling Peptides and Proteins/metabolism
4.
J Pathol ; 260(3): 304-316, 2023 07.
Article in English | MEDLINE | ID: mdl-37138382

ABSTRACT

Evasion from immunity is a major obstacle to the achievement of successful cancer immunotherapy. Hybrids derived from cell-cell fusion are theoretically associated with tumor heterogeneity and progression by conferring novel properties on tumor cells, including drug resistance and metastatic capacity; however, their impact on immune evasion remains unknown. Here, we investigated the potency of tumor-macrophage hybrids in immune evasion. Hybrids were established by co-culture of a melanoma cell line (A375 cells) and type 2 macrophages. The hybrids showed greater migration ability and greater tumorigenicity than the parental melanoma cells. The hybrids showed heterogeneous sensitivity to New York esophageal squamous cell carcinoma-1 (NY-ESO-1)-specific T-cell receptor-transduced T (TCR-T) cells and two out of four hybrid clones showed less sensitivity to TCR-T compared with the parental cells. An in vitro tumor heterogeneity model revealed that the TCR-T cells preferentially killed the parental cells compared with the hybrids and the survival rate of the hybrids was higher than that of the parental cells, indicating that the hybrids evade killing by TCR-T cells efficiently. Analysis of a single-cell RNA sequencing dataset of patients with melanoma revealed that a few macrophages expressed RNA encoding melanoma differentiation antigens including melan A, tyrosinase, and premelanosome protein, which indicated the presence of hybrids in primary melanoma. In addition, the number of potential hybrids was correlated with a poorer response to immune checkpoint blockade. These results provide evidence that melanoma-macrophage fusion has a role in tumor heterogeneity and immune evasion. © 2023 The Pathological Society of Great Britain and Ireland.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Melanoma , Humans , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Melanoma/metabolism , Macrophages/pathology , Receptors, Antigen, T-Cell/metabolism , Antigens, Neoplasm
5.
Anticancer Res ; 43(4): 1477-1484, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36974807

ABSTRACT

BACKGROUND/AIM: Malignant melanoma is a fatal skin cancer and is among the most immunogenic malignancies expressing melanoma-differentiation antigens and neoantigens. SRY-related HMG-box 10 (SOX10) is a transcription factor and a neural-crest differentiation marker that is used as a diagnostic marker for melanoma whilst playing a role in melanoma initiation through activation of the SOX10-MITF axis. SOX10 was shown to play a role in melanoma initiation by inducing expression of immune checkpoint molecules (e.g., HVEM and CEACAM1). In this study, we aimed to investigate the relationship between SOX10 and the expression an immune checkpoint molecule, programmed death-1 ligand 1 (PD-L1). MATERIALS AND METHODS: SOX10 overexpression and knockdown was performed using SOX10 gene transfection and SOX10 siRNA transfection into A375 melanoma cells. PD-L1 expression was assessed by flow cytometry and western blotting. T cell response was evaluated using NY-ESO-1 specific TCR-transduced T (TCR-T) cells by IFNγ ELISPOT assay. RESULTS: SOX10 overexpression increased the expression of PD-L1, whereas SOX10 knockdown, using siRNA, decreased its expression. IFNγ ELISPOT assay revealed that overexpression of SOX10 decreased the susceptibility of cells to NY-ESO-1-specific TCR-T cells. CONCLUSION: SOX10 has a role in the intrinsic immune suppressive mechanisms of melanoma through expression of PD-L1.


Subject(s)
Immune Checkpoint Proteins , Melanoma , Humans , T-Lymphocytes/metabolism , B7-H1 Antigen/genetics , Ligands , Programmed Cell Death 1 Receptor/metabolism , Melanoma/metabolism , Receptors, Antigen, T-Cell , SOXE Transcription Factors/genetics
6.
Cancer Immunol Immunother ; 72(7): 2057-2065, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36795123

ABSTRACT

Bladder cancer is a major and fatal urological disease. Cisplatin is a key drug for the treatment of bladder cancer, especially in muscle-invasive cases. In most cases of bladder cancer, cisplatin is effective; however, resistance to cisplatin has a significant negative impact on prognosis. Thus, a treatment strategy for cisplatin-resistant bladder cancer is essential to improve the prognosis. In this study, we established a cisplatin-resistant (CR) bladder cancer cell line using an urothelial carcinoma cell lines (UM-UC-3 and J82). We screened for potential targets in CR cells and found that claspin (CLSPN) was overexpressed. CLSPN mRNA knockdown revealed that CLSPN had a role in cisplatin resistance in CR cells. In our previous study, we identified human leukocyte antigen (HLA)-A*02:01-restricted CLSPN peptide by HLA ligandome analysis. Thus, we generated a CLSPN peptide-specific cytotoxic T lymphocyte clone that recognized CR cells at a higher level than wild-type UM-UC-3 cells. These findings indicate that CLSPN is a driver of cisplatin resistance and CLSPN peptide-specific immunotherapy may be effective for cisplatin-resistant cases.


Subject(s)
Adaptor Proteins, Signal Transducing , Drug Resistance, Neoplasm , Urinary Bladder Neoplasms , Humans , Cell Line, Tumor , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/therapy , Cisplatin/therapeutic use , Immunotherapy , Adaptor Proteins, Signal Transducing/metabolism , Up-Regulation , T-Lymphocytes, Cytotoxic/cytology , Neoplastic Stem Cells/drug effects
7.
Cancer Med ; 12(4): 4605-4615, 2023 02.
Article in English | MEDLINE | ID: mdl-36200687

ABSTRACT

Immune checkpoint inhibitor-based cancer immunotherapy has provided an additional therapeutic option for oral squamous cell carcinoma (OSCC) with recurrence or distant metastases. However, further improvement of OSCC treatment is required to develop the optimal combination or order for chemoradiotherapy and immunotherapy. Along with the accumulation of clinical knowledge and evidence, it is also essential to clarify the biological impact of chemo-radiotherapeutic agents on the cancer immune microenvironment. In this study, we investigated the effects of cisplatin (CDDP), a key therapeutic agent for OSCC, on programmed death-ligand 1 (PD-L1) expression in OSCC lines. Although CDDP treatment increased the surface levels of PD-L1 on OSCC cell lines, the gene and total protein expression levels of PD-L1 were not altered. We also demonstrated that the phosphorylation of heat shock factor 1 and heat shock protein 90 was involved in this process. In addition, CDDP-induced PD-L1 attenuated the target-specific cytotoxic T lymphocyte reaction to OSCC. These results provide an immunobiological basis for the response of OSCC to CDDP and will contribute to our biological understanding of the action of novel combination therapy including immunotherapy together with platinum-based chemotherapy for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Squamous Cell Carcinoma of Head and Neck , B7-H1 Antigen/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Tumor Microenvironment
8.
Biochem Biophys Res Commun ; 590: 117-124, 2022 01 29.
Article in English | MEDLINE | ID: mdl-34974299

ABSTRACT

The association between type 2 diabetes mellitus and prostate cancer is still under investigation, and the relationship between hyperinsulinemia and prostate cancer stem-like cells (CSCs) is elusive. Here, we investigated the function of insulin/AKT signaling in prostate CSCs. We isolated prostate CSCs as aldehyde dehydrogenase 1-high (ALDH1high) cells from the human prostate cancer 22Rv1 cell line using an ALDEFLUOR assay and established several ALDH1high and ALDH1low clones. ALDH1high clones showed high ALDH1 expression which is a putative CSC marker; however, they showed heterogeneity regarding tumorigenicity and resistance to radiation and chemotherapy. Interestingly, all ALDH1high clones showed lower phosphorylated AKT (Ser473) (pAKT) levels than the ALDH1low clones. PI3K/AKT signaling is a key cell survival pathway and we analyzed radiation resistance under AKT signaling activation by insulin. Insulin increased pAKT levels in ALDH1high and ALDH1low cells; the fold increase rate of pAKT was higher in ALDH1high cells than in ALDH1low cells. Insulin induced resistance to radiation and chemotherapy in ALDH1high cells, and the increased levels of pAKT induced by insulin were significantly related to radiation resistance. These results suggest that ALDH1 suppresses baseline pAKT levels, but AKT can be activated by insulin, leading to treatment resistance.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Insulin/pharmacology , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance , Signal Transduction , Animals , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Male , Mice , Phosphorylation/drug effects , Prostatic Neoplasms/pathology
10.
EBioMedicine ; 44: 86-97, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31129096

ABSTRACT

BACKGROUND: The therapeutic benefits of mesenchymal stromal cells (MSCs) include treatment of chronic inflammation. However, given the short-lived engraftment of these cells in vivo, their therapeutic efficacy remains mysterious. Transient induction of cellular senescence contributes to activation of immune cells, which promotes clearance of damaged cells during tissue remodelling. This may occur in tissue-resident mesenchymal progenitor cells during regeneration. Elucidation of the role of senescence in tissue-resident mesenchymal progenitor cells during regeneration would provide insight into the profile of therapeutic MSCs for treatment of chronic inflammatory disease. METHODS: We evaluated multipotent mesenchymal progenitor cells, termed fibro/adipogenic progenitors (FAPs), and immune cells in acute muscle injury (AMI) model mice and mice with myosin-induced experimental autoimmune myositis, a model of chronic inflammatory myopathy (CIM). Human bone marrow MSCs were optimised for the treatment of CIM using placental extract. FINDING: FAPs in AMI transiently expressed p16INK4A on days 1 and 2 after injury and recruited phagocytic immune cells, whereas in CIM, p16INK4A expression in FAPs was low. Cellular senescence occurs during the natural maturation of the placenta. Therefore, we used human placental extract to induce p16INK4A expression in therapeutic human bone marrow MSCs in culture. Treatment of CIM with p16INK4A-expressing MSCs promoted tissue remodelling by transiently increasing the abundance of engrafted MSCs, inducing cellular senescence in innate FAPs, and recruiting phagocytic immune cells. INTERPRETATION: MSCs may exert their effect by remodelling the chronic inflammatory environment via senescence-related regenerative processes.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cytophagocytosis/genetics , Mesenchymal Stem Cells/metabolism , Muscle Development/genetics , Myositis/etiology , Animals , Biomarkers , Cell Proliferation , Cellular Senescence/immunology , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytokines/metabolism , Cytophagocytosis/immunology , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mice , Myositis/metabolism , Myositis/pathology , Regeneration , Regenerative Medicine
11.
PLoS One ; 13(9): e0204252, 2018.
Article in English | MEDLINE | ID: mdl-30240403

ABSTRACT

Increasing evidence suggests that an enriched environment (EE) ameliorates cognitive impairment by promoting repair of brain damage. However, the mechanisms by which this occurs have not been determined. To address this issue, we investigated whether an EE enhanced the capability of endogenous bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) to prevent hippocampal damage due to diabetes by focusing on miRNA carried in BM-MSC-derived exosomes. In diabetic streptozotocin (STZ) rats housed in an EE (STZ/EE), cognitive impairment was significantly reduced, and both neuronal and astroglial damage in the hippocampus was alleviated compared with STZ rats housed in conventional cages (STZ/CC). BM-MSCs isolated from STZ/CC rats had functional and morphological abnormalities that were not detected in STZ/EE BM-MSCs. The miR-146a levels in exosomes in conditioned medium of cultured BM-MSCs and serum from STZ/CC rats were decreased compared with non-diabetic rats, and the level was restored in STZ/EE rats. Thus, the data suggest that increased levels of miR-146a in sera were derived from endogenous BM-MSCs in STZ/EE rats. To examine the possibility that increased miR-146a in serum may exert anti-inflammatory effects on astrocytes in diabetic rats, astrocytes transfected with miR-146a were stimulated with advanced glycation end products (AGEs) to mimic diabetic conditions. The expression of IRAK1, NF-κB, and tumor necrosis factor-α was significantly higher in AGE-stimulated astrocytes, and these factors were decreased in miR-146a-transfected astrocytes. These results suggested that EEs stimulate up-regulation of exosomal miR-146a secretion by endogenous BM-MSCs, which exerts anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment.


Subject(s)
Cognitive Dysfunction/prevention & control , Diabetes Mellitus, Experimental/pathology , MicroRNAs/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Bone Marrow Cells/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cells, Cultured , Cognitive Dysfunction/etiology , Diabetes Mellitus, Experimental/complications , Exosomes/metabolism , Glycation End Products, Advanced/pharmacology , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Maze Learning , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/blood , Oxidative Stress , Rats , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
12.
Sci Rep ; 8(1): 1161, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348535

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) are the most valuable source of autologous cells for transplantation and tissue regeneration to treat osteoporosis. Although BM-MSCs are the primary cells responsible for maintaining bone metabolism and homeostasis, their regenerative ability may be attenuated in postmenopausal osteoporosis patients. Therefore, we first examined potential abnormalities of BM-MSCs in an oestrogen-deficient rat model constructed by ovariectomy (OVX-MSCs). Cell proliferation, mobilisation, and regulation of osteoclasts were downregulated in OVX-MSCs. Moreover, therapeutic effects of OVX-MSCs were decreased in OVX rats. Accordingly, we developed a new activator for BM-MSCs using human umbilical cord extracts, Wharton's jelly extract supernatant (WJS), which improved cell proliferation, mobilisation and suppressive effects on activated osteoclasts in OVX-MSCs. Bone volume, RANK and TRACP expression of osteoclasts, as well as proinflammatory cytokine expression in bone tissues, were ameliorated by OVX-MSCs activated with WJS (OVX-MSCs-WJ) in OVX rats. Fusion and bone resorption activity of osteoclasts were suppressed in macrophage-induced and primary mouse bone marrow cell-induced osteoclasts via suppression of osteoclast-specific genes, such as Nfatc1, Clcn7, Atp6i and Dc-stamp, by co-culture with OVX-MSCs-WJ in vitro. In this study, we developed a new activator, WJS, which improved the functional abnormalities and therapeutic effects of BM-MSCs on postmenopausal osteoporosis.


Subject(s)
Bone Density/drug effects , Complex Mixtures/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Osteoclasts/metabolism , Osteoporosis/therapy , Umbilical Cord/chemistry , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Coculture Techniques , Complex Mixtures/isolation & purification , Disease Models, Animal , Female , Gene Expression , Mesenchymal Stem Cells/metabolism , Mice , Osteoclasts/cytology , Osteoporosis/etiology , Osteoporosis/genetics , Osteoporosis/pathology , Ovariectomy/adverse effects , RAW 264.7 Cells , Rats , Rats, Wistar , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tartrate-Resistant Acid Phosphatase/genetics , Tartrate-Resistant Acid Phosphatase/metabolism , Tomography, X-Ray Computed , Transcription Factors/genetics , Transcription Factors/metabolism , Wharton Jelly/chemistry
13.
Sci Rep ; 8(1): 1712, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374250

ABSTRACT

Although the cognitive impairment in Alzheimer's disease (AD) is believed to be caused by amyloid-ß (Aß) plaques and neurofibrillary tangles (NFTs), several postmortem studies have reported cognitive normal subjects with AD brain pathology. As the mechanism underlying these discrepancies has not been clarified, we focused the neuroprotective role of astrocytes. After examining 47 donated brains, we classified brains into 3 groups, no AD pathology with no dementia (N-N), AD pathology with no dementia (AD-N), and AD pathology with dementia (AD-D), which represented 41%, 21%, and 38% of brains, respectively. No differences were found in the accumulation of Aß plaques or NFTs in the entorhinal cortex (EC) between AD-N and AD-D. Number of neurons and synaptic density were increased in AD-N compared to those in AD-D. The astrocytes in AD-N possessed longer or thicker processes, while those in AD-D possessed shorter or thinner processes in layer I/II of the EC. Astrocytes in all layers of the EC in AD-N showed enhanced GLT-1 expression in comparison to those in AD-D. Therefore these activated forms of astrocytes with increased GLT-1 expression may exert beneficial roles in preserving cognitive function, even in the presence of Aß and NFTs.


Subject(s)
Alzheimer Disease/pathology , Astrocytes/enzymology , Astrocytes/pathology , Brain/pathology , Cognition Disorders/pathology , Glutamate Plasma Membrane Transport Proteins/analysis , Aged , Aged, 80 and over , Amyloid beta-Peptides/analysis , Excitatory Amino Acid Transporter 2 , Female , Humans , Male , Neurofibrillary Tangles/pathology
14.
Sci Rep ; 7(1): 8484, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814814

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSC) has been applied as the most valuable source of autologous cell transplantation for various diseases including diabetic complications. However, hyperglycemia may cause abnormalities in intrinsic BM-MSC which might lose sufficient therapeutic effects in diabetic patients. We demonstrated the functional abnormalities in BM-MSC derived from both type 1 and type 2 diabetes models in vitro, which resulted in loss of therapeutic effects in vivo in diabetic nephropathy (DN). Then, we developed a novel method to improve abnormalities in BM-MSC using human umbilical cord extracts, namely Wharton's jelly extract supernatant (WJs). WJs is a cocktail of growth factors, extracellular matrixes and exosomes, which ameliorates proliferative capacity, motility, mitochondrial degeneration, endoplasmic reticular functions and exosome secretions in both type 1 and type 2 diabetes-derived BM-MSC (DM-MSC). Exosomes contained in WJs were a key factor for this activation, which exerted similar effects to complete WJs. DM-MSC activated by WJs ameliorated renal injury in both type 1 and type 2 DN. In this study, we developed a novel activating method using WJs to significantly increase the therapeutic effect of BM-MSC, which may allow effective autologous cell transplantation.


Subject(s)
Diabetic Nephropathies/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetic Nephropathies/pathology , Disease Models, Animal , Exosomes , Humans , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Rats, Inbred OLETF , Rats, Sprague-Dawley , Wharton Jelly/chemistry
15.
Sci Rep ; 6: 34842, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721418

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs) have contributed to the improvement of diabetic nephropathy (DN); however, the actual mediator of this effect and its role has not been characterized thoroughly. We investigated the effects of MSC therapy on DN, focusing on the paracrine effect of renal trophic factors, including exosomes secreted by MSCs. MSCs and MSC-conditioned medium (MSC-CM) as renal trophic factors were administered in parallel to high-fat diet (HFD)-induced type 2 diabetic mice and streptozotocin (STZ)-induced insulin-deficient diabetic mice. Both therapies showed approximately equivalent curative effects, as each inhibited the exacerbation of albuminuria. They also suppressed the excessive infiltration of BMDCs into the kidney by regulating the expression of the adhesion molecule ICAM-1. Proinflammatory cytokine expression (e.g., TNF-α) and fibrosis in tubular interstitium were inhibited. TGF-ß1 expression was down-regulated and tight junction protein expression (e.g., ZO-1) was maintained, which sequentially suppressed the epithelial-to-mesenchymal transition of tubular epithelial cells (TECs). Exosomes purified from MSC-CM exerted an anti-apoptotic effect and protected tight junction structure in TECs. The increase of glomerular mesangium substrate was inhibited in HFD-diabetic mice. MSC therapy is a promising tool to prevent DN via the paracrine effect of renal trophic factors including exosomes due to its multifactorial action.


Subject(s)
Culture Media, Conditioned/pharmacology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/therapy , Kidney Tubules/metabolism , Mesenchymal Stem Cell Transplantation/methods , Albuminuria , Animals , Bone Marrow Cells/pathology , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/metabolism , Diet, High-Fat/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Exosomes/metabolism , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Rats, Inbred Lew
16.
Sci Rep ; 6: 24805, 2016 04 22.
Article in English | MEDLINE | ID: mdl-27102354

ABSTRACT

The incidence of dementia is higher in diabetic patients, but no effective treatment has been developed. This study showed that rat bone marrow mesenchymal stem cells (BM-MSCs) can improve the cognitive impairments of STZ-diabetic mice by repairing damaged neurons and astrocytes. The Morris water maze test demonstrated that cognitive impairments induced by diabetes were significantly improved by intravenous injection of BM-MSCs. In the CA1 region of the hippocampus, degeneration of neurons and astrocytes, as well as synaptic loss, were prominent in diabetes, and BM-MSC treatment successfully normalized them. Since a limited number of donor BM-MSCs was observed in the brain parenchyma, we hypothesized that humoral factors, especially exosomes released from BM-MSCs, act on damaged neurons and astrocytes. To investigate the effectiveness of exosomes for treatment of diabetes-induced cognitive impairment, exosomes were purified from the culture media and injected intracerebroventricularly into diabetic mice. Recovery of cognitive impairment and histological abnormalities similar to that seen with BM-MSC injection was found following exosome treatment. Use of fluorescence-labeled exosomes demonstrated that injected exosomes were internalized into astrocytes and neurons; these subsequently reversed the dysfunction. The present results indicate that exosomes derived from BM-MSCs might be a promising therapeutic tool for diabetes-induced cognitive impairment.


Subject(s)
Astrocytes/physiology , Cognitive Dysfunction/therapy , Diabetes Complications/therapy , Exosomes/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Neurons/physiology , Administration, Intravenous , Animals , CA1 Region, Hippocampal/pathology , Disease Models, Animal , Mice , Rats , Treatment Outcome
17.
Hepatology ; 59(2): 580-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23913513

ABSTRACT

UNLABELLED: The role of the cytokine, macrophage migration inhibitory factor (MIF), and its receptor, CD74, was assessed in autoimmune hepatitis (AIH) and primary biliary cirrhosis (PBC). Two MIF promoter polymorphisms, a functional -794 CATT5-8 microsatellite repeat (rs5844572) and a -173 G/C single-nucleotide polymorphism (rs755622), were analyzed in DNA samples from over 500 patients with AIH, PBC, and controls. We found a higher frequency of the proinflammatory and high-expression -794 CATT7 allele in AIH, compared to PBC, whereas lower frequency was found in PBC, compared to both AIH and healthy controls. MIF and soluble MIF receptor (CD74) were measured by enzyme-linked immunosorbent assay in 165 serum samples of AIH, PBC, and controls. Circulating serum and hepatic MIF expression was elevated in patients with AIH and PBC versus healthy controls. We also identified a truncated circulating form of the MIF receptor, CD74, that is released from hepatic stellate cells and that binds MIF, neutralizing its signal transduction activity. Significantly higher levels of CD74 were found in patients with PBC versus AIH and controls. CONCLUSIONS: These data suggest a distinct genetic and immunopathogenic basis for AIH and PBC at the MIF locus. Circulating MIF and MIF receptor profiles distinguish PBC from the more inflammatory phenotype of AIH and may play a role in pathogenesis and as biomarkers of these diseases.


Subject(s)
Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/physiopathology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/physiology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/physiopathology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/physiology , Adult , Aged , Aged, 80 and over , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/physiology , Biomarkers/metabolism , Biopsy , Case-Control Studies , Cohort Studies , Female , Gene Frequency/genetics , Hepatitis, Autoimmune/pathology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/physiology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/pathology , Male , Microsatellite Repeats/genetics , Middle Aged , Phenotype , Polymorphism, Single Nucleotide/genetics
18.
Arthritis Rheum ; 63(12): 3942-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22127710

ABSTRACT

OBJECTIVE: To study the effect of the innate cytokine macrophage migration inhibitory factor (MIF) on the susceptibility and severity of systemic lupus erythematosus (SLE) in a multinational population of 1,369 Caucasian and African American patients. METHODS: Two functional polymorphisms in the MIF gene, a -794 CATT(5-8) microsatellite repeat (rs5844572) and a -173 G/C single-nucleotide polymorphism (rs755622), were assessed for association with SLE in 3,195 patients and healthy controls. We also measured MIF plasma levels in relation to genotypes and clinical phenotypes, and assessed Toll-like receptor 7 (TLR-7)-stimulated MIF production in vitro. RESULTS: Both Caucasians and African Americans with the high-expression MIF haplotype -794 CATT(7)/-173*C had a lower incidence of SLE (in Caucasians, odds ratio [OR] 0.63, 95% confidence interval [95% CI] 0.53-0.89, P = 0.001; in African Americans, OR 0.46, 95% CI 0.23-0.95, P = 0.012). In contrast, among patients with established SLE, reduced frequencies of low-expression MIF genotypes (-794 CATT(5)) were observed in those with nephritis, those with serositis, and those with central nervous system (CNS) involvement when compared to patients without end-organ involvement (P = 0.023, P = 0.005, and P = 0.04, respectively). Plasma MIF levels and TLR-7-stimulated MIF production in vitro reflected the underlying MIF genotype of the studied groups. CONCLUSION: These findings suggest that MIF, which has both proinflammatory properties and macrophage and B cell survival functions, exerts a dual influence on the immunopathogenesis of SLE. High-expression MIF genotypes are associated with a reduced susceptibility to SLE and may contribute to an enhanced clearance of infectious pathogens. Once SLE develops, however, low-expression MIF genotypes may protect from ensuing inflammatory end-organ damage.


Subject(s)
Genetic Predisposition to Disease/genetics , Lupus Erythematosus, Systemic/genetics , Macrophage Migration-Inhibitory Factors/genetics , Polymorphism, Single Nucleotide/genetics , Severity of Illness Index , Adult , Black or African American/ethnology , Case-Control Studies , Cross-Sectional Studies , Female , Genotype , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/ethnology , Macrophage Migration-Inhibitory Factors/blood , Male , Middle Aged , Phenotype , Tumor Necrosis Factor-alpha/blood , White People/ethnology
19.
Anticancer Res ; 31(7): 2511-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21873168

ABSTRACT

We generated an evolutionary computer program that generates complementary peptide (C-pep) sequences, with the potential to interact with a target peptide, by comparing several physico-chemical parameters of each pair of the complementary peptides being analyzed. We generated C-peps to target several molecules. About 30% of synthesized C-peps interfered with the function of their targets. C5a stimulates generation of TNFα and other inflammatory cytokines. Inhibition of C5a should be effective against sepsis, which impairs the status of cancer-bearing patients. One of the inhibitory C-peps of C5a, termed AcPepA, was effective in Cynomolgus monkeys intravenously infused with a lethal dose of bacterial LPS (4 mg/kg) destined to die. The monkeys were rescued by intravenous administration of 2 mg/kg/h of AcPepA. The excellent therapeutic effect of AcPepA is likely to be due to restriction of high mobility group box 1 (HMGB1) surge induced by the effect of C5a on C5L2, which is the second C5a receptor, since the released HMGB1 has the capacity to stimulate TLR4 as an endogeneous ligand resulting in further activation of inflammatory cells to release inflammatory cytokines forming a positive feedback circuit of inflammation.


Subject(s)
Molecular Targeted Therapy , Peptide Library , Peptides/therapeutic use , Amino Acid Sequence , Animals , Complement C5a/antagonists & inhibitors , Cytokines/metabolism , Directed Molecular Evolution , Drug Evaluation, Preclinical , Endotoxemia/drug therapy , Endotoxemia/pathology , Endotoxemia/physiopathology , Feedback, Physiological , HMGB1 Protein/physiology , Inflammation/drug therapy , Inflammation/physiopathology , Lipopolysaccharides/toxicity , Lung/pathology , Macaca fascicularis , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Receptor, Anaphylatoxin C5a/physiology , Software , Structure-Activity Relationship , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/analysis
20.
J Clin Microbiol ; 49(7): 2552-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543572

ABSTRACT

Acute infection with Toxoplasma gondii during pregnancy can cause congenital toxoplasmosis. The aim of this study was to evaluate whether screening with the use of IgG avidity and multiplex nested PCR methods was effective to detect a high-risk pregnancy. In a prospective study, serum T. gondii IgG avidity was measured in consecutive 146 pregnant women testing positive for T. gondii antibody and either positive or equivocal for IgM. Multiplex nested PCR for T. gondii DNA on amniotic fluid, maternal blood, and umbilical cord blood were performed with informed consent. A total of 51 (34.9%) women presented with low IgG avidity (<30%), 15 (10.3%) presented with borderline avidity (30 to 35%), and 80 (54.8%) presented with high avidity (>35%) indices. Amniotic fluid obtained at amniocentesis or birth yielded positive PCR results in nine women with low IgG avidity indices. Of these nine women, three had congenital toxoplasmosis. None of women with high or border line IgG avidity indices had a positive PCR result in the amniotic fluid or congenital toxoplasmosis. No congenital toxoplasmosis was detected in women whose amniotic fluids yielded negative PCR results. Ingestion of raw or undercooked meat was found to be the main risk factor for acute T. gondii infection. Congenital toxoplasmosis screening with a combination of IgG avidity in the maternal blood and multiplex nested PCR in the amniotic fluid was useful for detecting a high risk pregnancy and diagnosing congenital toxoplasmosis.


Subject(s)
Antibodies, Protozoan/blood , Antibody Affinity , Immunoglobulin G/blood , Mass Screening/methods , Polymerase Chain Reaction/methods , Pregnancy Complications, Infectious/diagnosis , Toxoplasmosis/diagnosis , Amniotic Fluid/parasitology , Child, Preschool , Female , Humans , Immunoassay/methods , Infant , Infant, Newborn , Male , Pregnancy , Prognosis , Prospective Studies , Toxoplasma/genetics , Toxoplasma/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...