Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
J Clin Oncol ; : JCO2302376, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776485

ABSTRACT

PURPOSE: The humanized antivascular endothelial growth factor (VEGF) antibody bevacizumab (Bev) is efficacious for the treatment of NF2-related schwannomatosis (NF2), previously known as neurofibromatosis type 2. This study evaluated the safety and efficacy of a VEGF receptor (VEGFR) vaccine containing VEGFR1 and VEGFR2 peptides in patients with NF2 with progressive schwannomas (jRCTs031180184). MATERIALS AND METHODS: VEGFR1 and VEGFR2 peptides were injected subcutaneously into infra-axillary and inguinal regions, once a week for 4 weeks and then once a month for 4 months. The primary end point was safety. Secondary end points included tolerability, hearing response, imaging response, and immunologic response. RESULTS: Sixteen patients with NF2 with progressive schwannomas completed treatment and were assessed. No severe vaccine-related adverse events occurred. Among the 13 patients with assessable hearing, word recognition score improved in five patients at 6 months and two at 12 months. Progression of average hearing level of pure tone was 0.168 dB/mo during the year of treatment period, whereas long-term progression was 0.364 dB/mo. Among all 16 patients, a partial response was observed in more than one schwannoma in four (including one in which Bev had not been effective), minor response in 5, and stable disease in 4. Both VEGFR1-specific and VEGFR2-specific cytotoxic T lymphocytes (CTLs) were induced in 11 patients. Two years after vaccination, a radiologic response was achieved in nine of 20 assessable schwannomas. CONCLUSION: This study demonstrated the safety and preliminary efficacy of VEGFR peptide vaccination in patients with NF2. Memory-induced CTLs after VEGFR vaccination may persistently suppress tumor progression.

2.
Cancer Sci ; 115(1): 48-58, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37879607

ABSTRACT

We previously reported that the inhibition of stearoyl-CoA desaturase 1 (SCD1) enhances the antitumor function of CD8+ T cells indirectly via restoring production of DC recruiting chemokines by cancer cells and subsequent induction of antitumor CD8+ T cells. In this study, we investigated the molecular mechanism of direct enhancing effects of SCD1 inhibitors on CD8+ T cells. In vitro treatment of CD8+ T cells with SCD1 inhibitors enhanced IFN-γ production and cytotoxic activity of T cells along with decreased oleic acid and esterified cholesterol, which is generated by cholesterol esterase, acetyl-CoA acetyltransferase 1 (ACAT1), in CD8+ T cells. The addition of oleic acid or cholesteryl oleate reversed the enhanced functions of CD8+ T cells treated with SCD1 inhibitors. Systemic administration of SCD1 inhibitor to MCA205 tumor-bearing mice enhanced IFN-γ production of tumor-infiltrating CD8+ T cells, in which oleic acid and esterified cholesterol, but not cholesterol, were decreased. These results indicated that SCD1 suppressed effector functions of CD8+ T cells through the increased esterified cholesterol in an ACAT1-dependent manner, and SCD1 inhibition enhanced T cell activity directly through decreased esterified cholesterol. Finally, SCD1 inhibitors or ACAT1 inhibitors synergistically enhanced the antitumor effects of anti-PD-1 antibody therapy or CAR-T cell therapy in mouse tumor models. Therefore, the SCD1-ACAT1 axis is regulating effector functions of CD8+ T cells, and SCD1 inhibitors, and ACAT1 inhibitors are attractive drugs for cancer immunotherapy.


Subject(s)
Neoplasms , Oleic Acid , Mice , Animals , Oleic Acid/pharmacology , CD8-Positive T-Lymphocytes , Acetyltransferases , Cholesterol , Stearoyl-CoA Desaturase
4.
Ann Surg Oncol ; 30(8): 5267-5277, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37222942

ABSTRACT

BACKGROUND: The prognosis for patients with colorectal cancer (CRC) is determined by tumor characteristics as well as the host immune response. This study investigated the relationship between an immunosuppressive state and patient prognosis by evaluating the systemic and tumor microenvironment (TME) interleukin (IL)-6 levels. METHODS: Preoperative serum IL-6 levels were measured using an electrochemiluminescence assay. Expression of IL-6 in tumor and stromal cells was evaluated immunohistochemically in 209 patients with resected CRC. Single-cell analysis of tumor-infiltrating immune cells was performed using mass cytometry in 10 additional cases. RESULTS: Elevated serum IL-6 levels were associated with elevated stromal IL-6 levels and a poor prognosis for patients with CRC. High IL-6 expression in stromal cells was associated with low-density subsets of CD3+ and CD4+ T cells as well as FOXP3+ cells. Mass cytometry analysis showed that IL-6+ cells among tumor-infiltrating immune cells were composed primarily of myeloid cells and rarely of lymphoid cells. In the high-IL-6-expression group, the percentages of myeloid-derived suppressor cells (MDSCs) and CD4+FOXP3highCD45RA- effector regulatory T cells (eTreg) were significantly higher than in the low-IL-6-expression group. Furthermore, the proportion of IL-10+ cells in MDSCs and that of IL-10+ or CTLA-4+ cells in eTregs correlated with IL-6 levels. CONCLUSION: Elevated serum IL-6 levels were associated with stromal IL-6 levels in CRC. High IL-6 expression in tumor-infiltrating immune cells also was associated with accumulation of immunosuppressive cells in the TME.


Subject(s)
Colorectal Neoplasms , Interleukin-10 , Humans , Colorectal Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Interleukin-6 , Lymphocytes, Tumor-Infiltrating , Prognosis , Tumor Microenvironment
5.
J Gastroenterol ; 58(6): 540-553, 2023 06.
Article in English | MEDLINE | ID: mdl-36859628

ABSTRACT

BACKGROUND: Recent advances in immune checkpoint blockade (ICB) have improved patient prognosis in mismatch repair-deficient and microsatellite instability-high colorectal cancer (dMMR/MSI-H CRC); however, PD-1 blockade has faced a challenge in early progressive disease. We aimed to understand the early event in ICB resistance using an in vivo model. METHODS: We subcutaneously transplanted the MC38 colon cancer cells into C57BL/6 mice, intraperitoneally injected anti-PD-1 antibody and then isolated ICB-resistant subclones from the recurrent tumors. RESULTS: Comparative gene expression analysis discovered seven genes significantly downregulated in the ICB-resistant cells. Tumorigenicity assay of the MC38 cells knocked out each of the seven candidate genes into C57BL/6 mice treated with anti-PD-1 antibody and bioinformatics analysis of the relationship between the expression of the seven candidate genes and the outcome of cancer patients receiving immunotherapy identified Rtp4, an interferon-stimulated gene and a chaperon protein of G protein-coupled receptors, as a gene involved in ICB resistance. Immunohistochemical analysis of transplanted tumor tissues demonstrated that anti-PD-1 antibody failed to recruit T lymphocytes in the Rtp4-KO MC38 cells. Mouse and human RTP4 expression could be silenced via histone H3 lysine 9 (H3K9) trimethylation, and public transcriptome data indicated the high expression level of RTP4 in most but not all of dMMR/MSI-H CRC. CONCLUSIONS: We clarified that RTP4 could be silenced by histone H3K9 methylation as the early event of ICB resistance. RTP4 expression could be a promising biomarker for predicting ICB response, and the combination of epigenetic drugs and immune checkpoint inhibitors might exhibit synergistic effects on dMMR/MSI-H CRC.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, Inbred C57BL , Neoplasm Recurrence, Local , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Microsatellite Instability , Molecular Chaperones/genetics , Molecular Chaperones/therapeutic use
6.
Exp Dermatol ; 32(3): 297-305, 2023 03.
Article in English | MEDLINE | ID: mdl-36607252

ABSTRACT

Melanoma has been a prototype for cancer immunology research, and the mechanisms of anti-tumor T-cell responses have been extensively investigated in patients treated with various immunotherapies. Individual differences in cancer-immune status are defined mainly by cancer cell characteristics such as DNA mutations generating immunogenic neo-antigens, and oncogene activation causing immunosuppression, but also by patients' genetic backgrounds such as HLA types and genetic polymorphisms of immune related molecules, and environmental and lifestyle factors such as UV rays, smoking, gut microbiota and concomitant medications; these factors have an influence on the efficacy of immunotherapy. Recent comparative studies on responders and non-responders in immune-checkpoint inhibitor therapy using various new technologies including multi-omics analyses on genomic DNA, mRNA, metabolites and microbiota and single cell analyses of various immune cells have led to the advance of human tumor immunology and the development of new immunotherapy. Based on the new findings from these investigations, personalized cancer immunotherapies along with appropriate biomarkers and therapeutic targets are being developed for patients with melanoma. Here, we will discuss one of the essential subjects in tumor immunology: identification of immunogenic tumor antigens and their effective use in various immunotherapies including cancer vaccines and adoptive T-cell therapy.


Subject(s)
Cancer Vaccines , Melanoma , Humans , T-Lymphocytes , Melanoma-Specific Antigens , Melanoma/drug therapy , Immunotherapy , Antigens, Neoplasm , Cancer Vaccines/therapeutic use , Immunotherapy, Adoptive
7.
J Neurochem ; 164(1): 29-43, 2023 01.
Article in English | MEDLINE | ID: mdl-36448220

ABSTRACT

Neurons in the central nervous system (CNS) have limited capacity for axonal regeneration after trauma and neurological disorders due to an endogenous nonpermissive environment for axon regrowth in the CNS. Lateral olfactory tract usher substance (LOTUS) contributes to axonal tract formation in the developing brain and axonal regeneration in the adult brain as an endogenous Nogo receptor-1 (NgR1) antagonist. However, how LOTUS expression is regulated remains unclarified. This study examined molecular mechanism of regulation in LOTUS expression and found that brain-derived neurotrophic factor (BDNF) increased LOTUS expression in cultured hippocampal neurons. Exogenous application of BDNF increased LOTUS expression at both mRNA and protein levels in a dose-dependent manner. We also found that pharmacological inhibition with K252a and gene knockdown by siRNA of tropomyosin-related kinase B (TrkB), BDNF receptor suppressed BDNF-induced increase in LOTUS expression. Further pharmacological analysis of the TrkB signaling pathway revealed that BDNF increased LOTUS expression through mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) cascades, but not phospholipase C-γ (PLCγ) cascade. Additionally, treatment with c-AMP response element binding protein (CREB) inhibitor partially suppressed BDNF-induced LOTUS expression. Finally, neurite outgrowth assay in cultured hippocampal neurons revealed that BDNF treatment-induced antagonism for NgR1 by up-regulating LOTUS expression. These findings suggest that BDNF may acts as a positive regulator of LOTUS expression through the TrkB signaling, thereby inducing an antagonistic action for NgR1 function by up-regulating LOTUS expression. Also, BDNF may synergistically affect axon regrowth through the upregulation of LOTUS expression.


Subject(s)
Brain-Derived Neurotrophic Factor , Olfactory Bulb , Cells, Cultured , Phosphatidylinositol 3-Kinases , Receptor, trkB , Signal Transduction , Up-Regulation , Animals
8.
Article in English | MEDLINE | ID: mdl-36347468

ABSTRACT

Metamorphosis of teleosts including Anguilliformes is well known to be induced by thyroid hormone (TH), although the underlying mechanism is not fully understood. In this study, we investigated the experimental conditions needed to induce normal metamorphosis in artificially spawned Japanese eel (Anguilla japonica), including initial larval size, TH concentration, and timing of TH immersion. Around 37 mm TL was found to be the minimum size of larvae that underwent successful metamorphosis induced by l-thyroxine (T4); notably, smaller larvae did not show increased expression of TH receptors in response to T4, suggesting that small leptocephali are not sufficiently responsive to TH. Furthermore, successful completion of metamorphosis depended on sensitivity to TH, which changed with metamorphic stage; for example, prolonged exposure to higher TH concentrations led to morphological defects. Collectively, these results reveal that the induction of metamorphosis by TH is dependent on larval size, and that the concentration of TH must be adjusted in line with metamorphic stage to achieve successful progression of metamorphosis. Our findings will contribute to improving production technology in the aquaculture of Japanese eels by facilitating the earlier induction of metamorphosis in artificial leptocephali.


Subject(s)
Anguilla , Thyroid Hormones , Animals , Larva/metabolism , Thyroid Hormones/pharmacology , Thyroxine/pharmacology , Thyroxine/metabolism , Anguilla/metabolism , Metamorphosis, Biological/physiology
9.
Mol Med ; 28(1): 154, 2022 12 12.
Article in English | MEDLINE | ID: mdl-36510132

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide but has no effective treatment. Amyloid beta (Aß) protein, a primary risk factor for AD, accumulates and aggregates in the brain of patients with AD. Paired immunoglobulin-like receptor B (PirB) has been identified as a receptor of Aß and Aß-PirB molecular interactions that cause synapse elimination and synaptic dysfunction. PirB deletion has been shown to suppress Aß-induced synaptic dysfunction and behavioral deficits in AD model mice, implying that PirB mediates Aß-induced AD pathology. Therefore, inhibiting the Aß-PirB molecular interaction could be a successful approach for combating AD pathology. We previously showed that lateral olfactory tract usher substance (LOTUS) is an endogenous antagonist of type1 Nogo receptor and PirB and that LOTUS overexpression promotes neuronal regeneration following damage to the central nervous system, including spinal cord injury and ischemic stroke. Therefore, in this study, we investigated whether LOTUS inhibits Aß-PirB interaction and Aß-induced dendritic spine elimination. METHODS: The inhibitory role of LOTUS against Aß-PirB (or leukocyte immunoglobulin-like receptor subfamily B member 2: LilrB2) binding was assessed using a ligand-receptor binding assay in Cos7 cells overexpressing PirB and/or LOTUS. We assessed whether LOTUS inhibits Aß-induced intracellular alterations and synaptotoxicity using immunoblots and spine imaging in a primary cultured hippocampal neuron. RESULTS: We found that LOTUS inhibits the binding of Aß to PirB overexpressed in Cos7 cells. In addition, we found that Aß-induced dephosphorylation of cofilin and Aß-induced decrease in post-synaptic density-95 expression were suppressed in cultured hippocampal neurons from LOTUS-overexpressing transgenic (LOTUS-tg) mice compared with that in wild-type mice. Moreover, primary cultured hippocampal neurons from LOTUS-tg mice improved the Aß-induced decrease in dendritic spine density. Finally, we studied whether human LOTUS protein inhibits Aß binding to LilrB2, a human homolog of PirB, and found that human LOTUS inhibited the binding of Aß to LilrB2 in a similar manner. CONCLUSIONS: This study implied that LOTUS improved Aß-induced synapse elimination by suppressing Aß-PirB interaction in rodents and inhibited Aß-LilrB2 interaction in humans. Our findings revealed that LOTUS may be a promising therapeutic agent in counteracting Aß-induced AD pathologies.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Calcium-Binding Proteins , Receptors, Immunologic , Animals , Humans , Mice , Alzheimer Disease/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Immunoglobulins/metabolism , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Receptors, Immunologic/metabolism , Calcium-Binding Proteins/metabolism
10.
BMC Cancer ; 22(1): 1071, 2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36253752

ABSTRACT

BACKGROUND: We recently reported the relapse-free survival (RFS) significance of the combination of CD4+ and forkhead box P3+ (FOXP3) T-cell densities identified by immunohistochemistry in patients with stage I, II, and III colorectal cancer (CRC) who underwent curative resections. This study was designed to determine the optimal combination of markers that predict recurrence in patients with T factors of T3/T4a stage II CRC by applying a novel Bayes decision rule. METHODS: Using 137 cancer tissue specimens from T3/T4a stage II patients, 12 clinicopathologic and immune factors were analysed as predictive candidates for recurrence. RESULTS: Our study showed that the combination of low CD4+ and low FOXP3+ T-cell densities resulted in extremely poor RFS. CONCLUSIONS: Adjuvant chemotherapy may be considered for patients with a combination of low CD4+ and low FOXP3+ T-cell densities. The discovery of this new prognostic indicator is important for the appropriate management of patients undergoing curative resection for T3/T4a stage II CRC.


Subject(s)
Colorectal Neoplasms , Forkhead Transcription Factors , Bayes Theorem , Biomarkers , Colorectal Neoplasms/pathology , Humans , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis
11.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35793868

ABSTRACT

BACKGROUND: Understanding the mechanisms of non-T cell inflamed tumor microenvironment (TME) and their modulation are important to improve cancer immunotherapies such as immune checkpoint inhibitors. The involvement of various immunometabolisms has recently been indicated in the formation of immunosuppressive TME. In this study, we investigated the immunological roles of stearoyl-CoA desaturase 1 (SCD1), which is essential for fatty acid metabolism, in the cancer immune response. METHODS: We investigated the roles of SCD1 by inhibition with the chemical inhibitor or genetic manipulation in antitumor T cell responses and the therapeutic effect of anti-programmed cell death protein 1 (anti-PD-1) antibody using various mouse tumor models, and their cellular and molecular mechanisms. The roles of SCD1 in human cancers were also investigated by gene expression analyses of colon cancer tissues and by evaluating the related free fatty acids in sera obtained from patients with non-small cell lung cancer who were treated with anti-PD-1 antibody. RESULTS: Systemic administration of a SCD1 inhibitor in mouse tumor models enhanced production of CCL4 by cancer cells through reduction of Wnt/ß-catenin signaling and by CD8+ effector T cells through reduction of endoplasmic reticulum stress. It in turn promoted recruitment of dendritic cells (DCs) into the tumors and enhanced the subsequent induction and tumor accumulation of antitumor CD8+ T cells. SCD1 inhibitor was also found to directly stimulate DCs and CD8+ T cells. Administration of SCD1 inhibitor or SCD1 knockout in mice synergized with an anti-PD-1 antibody for its antitumor effects in mouse tumor models. High SCD1 expression was observed in one of the non-T cell-inflamed subtypes in human colon cancer, and serum SCD1 related fatty acids were correlated with response rates and prognosis of patients with non-small lung cancer following anti-PD-1 antibody treatment. CONCLUSIONS: SCD1 expressed in cancer cells and immune cells causes immunoresistant conditions, and its inhibition augments antitumor T cells and therapeutic effects of anti-PD-1 antibody. Therefore, SCD1 is an attractive target for the development of new diagnostic and therapeutic strategies to improve current cancer immunotherapies including immune checkpoint inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung , Colonic Neoplasms , Lung Neoplasms , Stearoyl-CoA Desaturase , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mice , Mice, Knockout , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/immunology , Tumor Microenvironment , Wnt Signaling Pathway/immunology , beta Catenin/immunology
12.
Neurochem Res ; 47(9): 2741-2756, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35622214

ABSTRACT

One of the key areas in stem cell research is the identification of factors capable of promoting the expansion of Neural Stem Cell/Progenitor Cells (NSPCs) and understanding their molecular mechanisms for future use in clinical settings. We previously identified Macrophage Migration Inhibitory Factor (MIF) as a novel factor that can support the proliferation and/or survival of NSPCs based on in vitro functional cloning strategy and revealed that MIF can support the proliferation of human brain tumor-initiating cells (BTICs). However, the detailed downstream signaling for the functions has largely remained unknown. Thus, in the present study, we newly identified translationally-controlled tumor protein-1 (TPT1), which is expressed in the ventricular zone of mouse embryonic brain, as a downstream target of MIF signaling in mouse and human NSPCs and human BTICs. Using gene manipulation (over or downregulation of TPT1) techniques including CRISPR/Cas9-mediated heterozygous gene disruption showed that TPT1 contributed to the regulation of cell proliferation/survival in mouse NSPCs, human embryonic stem cell (hESC) derived-NSPCs, human-induced pluripotent stem cells (hiPSCs) derived-NSPCs and BTICs. Furthermore, gene silencing of TPT1 caused defects in neuronal differentiation in the NSPCs in vitro. We also identified the MIF-CHD7-TPT1-SMO signaling axis in regulating hESC-NSPCs and BTICs proliferation. Intriguingly, TPT1suppressed the miR-338 gene, which targets SMO in hESC-NSPCs and BTICs. Finally, mice with implanted BTICs infected with lentivirus-TPT1 shRNA showed a longer overall survival than control. These results also open up new avenues for the development of glioma therapies based on the TPT1 signaling pathway.


Subject(s)
Macrophage Migration-Inhibitory Factors , Neoplastic Stem Cells , Neural Stem Cells , Tumor Protein, Translationally-Controlled 1 , Animals , Brain/metabolism , Cell Proliferation/physiology , Humans , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Mice , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Tumor Protein, Translationally-Controlled 1/genetics
13.
Article in English | MEDLINE | ID: mdl-35339681

ABSTRACT

Hyaluronic acids (hyaluronans, HAs) are glycosaminoglycans produced in the bodies of Anguilliforme and Elopiforme leptocephali, and HA is thought to serve as a metabolic energy source during planktonic life-stages. To examine this hypothesis, we investigated the dynamics of HA during early growth of the Japanese eel (Anguilla japonica), including during metamorphosis. From histochemical observations in the fully grown leptocephalus, HA occupied approximately 40-50% of the cross-sectional area and muscle tissue occupied less than 20%. However, the HA and water content are at a maximum during leptocephalus, decreasing during metamorphosis. We found that during leptocephalus, HA is actively accumulated in the body and plays a role in specific density adjustment, facilitating planktonic life. It was thought that after metamorphosis the role of HA in facilitating floating would end, and it would be metabolized to glucose and/or triglyceride.


Subject(s)
Anguilla , Anguilla/physiology , Animals , Hyaluronic Acid , Metamorphosis, Biological/physiology , Muscles
14.
Gen Comp Endocrinol ; 317: 113977, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35065055

ABSTRACT

Growth hormone (Gh) regulates somatic growth in fishes, particularly through the Gh - insulin-like growth factor-I (Igf-I) axis. In this study, recombinant Japanese eel Ghs with or without C-terminal peptides of human chorionic gonadotropin (CTP), which are known to prolong the half-life, were produced using the HEK 293 and CHO expression system. The effect of recombinant Gh administration to eel larvae on their somatic growth was investigated in short-term feeding experiments, and it was found that three types of recombinant Ghs with CTP (CTP-reGh, reGh-CTP and reGh-CTP × 2) were more effective in promoting somatic growth in eel larvae than recombinant Ghs without CTP. Among the three recombinant Ghs with CTP, reGh-CTP × 2 had the highest growth-promoting effects, however only when provided in the short term. After long-term administration of reGh-CTP × 2, there was no difference in growth between the Gh administrated group and the control group. The survival rate of eel larvae were not affected by recombinant Ghs. In addition, the mRNA expression of gh, Gh receptors, Igf-I and IGF-II were measured by quantitative real-time PCR, and significant reductions in the expression of gh, Gh receptors and Igf-I were observed. These findings provide useful tools to study the mechanisms of somatic growth and increase understanding of Gh regulation in anguillid eel larvae.


Subject(s)
Anguilla , Anguilla/genetics , Animals , Growth Hormone/genetics , Growth Hormone/metabolism , Growth Hormone/pharmacology , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Larva/metabolism , Receptors, Somatotropin/metabolism
15.
Article in English | MEDLINE | ID: mdl-34906629

ABSTRACT

Japanese eels store lipids in the peritoneal cavity at the glass eel stage. These lipids are presumed to be consumed as an energy source during migration from oceanic metamorphosing sites to coastal Japan. In this study we investigated the distribution and amount of triglyceride in vivo using artificially bred Japanese eel larvae. Triglycerides accumulated in vivo from the early larval stage onward, until they reached a peak volume in fully grown leptocephali, and subsequently gradually decreased during metamorphosis. Furthermore, during the late metamorphic stages of the artificially bred glass eel, triglycerides were stored mainly in the peritoneal cavity, as in wild glass eels. These observations point to a strategy that the Japanese eels use in consuming the triglycerides derived from leptocephali during metamorphosis (when they do not feed) as a source of energy.


Subject(s)
Anguilla , Anguilla/physiology , Animals , Larva/physiology , Metamorphosis, Biological/physiology , Oceans and Seas , Triglycerides
16.
Sci Rep ; 11(1): 15421, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326367

ABSTRACT

Clinicians often perform pumping of infusions with a syringe (PIS) to quickly deliver fluid or blood transfusion to patients, especially during an emergency. Despite the efforts of the clinicians, critically ill patients are prone to acquire catheter-related bloodstream infections. Although clinicians have reported the possibility of PIS contamination, no group of researchers has studied nor confirmed this possibility. Here, we examined whether PIS can cause bacterial contamination of the fluid inside the syringes, using microbiological tests, including the analysis Escherichia coli DH-5 alpha growth by measuring the absorbance at OD600. We confirmed that contamination of fluid in the barrel was almost proportional to the applied volume of bacterial fluid. Aliquots of DH-5 alpha artificially applied on the surface of the gloved hand of an examiner, the plunger or the inner side of the barrel of a syringe could permeate inside the syringe. Furthermore, disinfection with ethanol before PIS almost successfully prevented bacterial multiplication. Our findings suggest that PIS can cause intraluminal contamination when performed with unsterilized hands, and that previous disinfection with ethanol can effectively prevent PIS-induced contamination. These results highlight the risk of PIS-induced contamination and the importance of disinfection in the daily clinical practice.


Subject(s)
Catheter-Related Infections/prevention & control , Disposable Equipment/microbiology , Drug Delivery Systems/methods , Equipment Contamination/prevention & control , Escherichia coli Infections/prevention & control , Escherichia coli/growth & development , Infusion Pumps , Syringes/microbiology , Anti-Infective Agents, Local/pharmacology , Bacteriological Techniques , Catheter-Related Infections/microbiology , Disinfection/methods , Escherichia coli/drug effects , Escherichia coli Infections/microbiology , Ethanol/pharmacology , Gloves, Surgical/microbiology , Hand/microbiology , Hand Disinfection/methods , Humans
17.
Int J Mol Sci ; 22(11)2021 Jun 06.
Article in English | MEDLINE | ID: mdl-34204098

ABSTRACT

Ocular graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation. Ocular GVHD affects recipients' visual function and quality of life. Recent advanced research in this area has gradually attracted attention from a wide range of physicians and ophthalmologists. This review highlights the mechanism of immune processes and the molecular mechanism, including several inflammation cascades, pathogenic fibrosis, and stress-induced senescence related to ocular GVHD, in basic spectrum topics in this area. How the disease develops and what kinds of cells participate in ocular GVHD are discussed. Although the classical immune process is a main pathological pathway in this disease, senescence-associated changes in immune cells and stem cells may also drive this disease. The DNA damage response, p16/p21, and the expression of markers associated with the senescence-associated secretory phenotype (SASP) are seen in ocular tissue in GVHD. Macrophages, T cells, and mesenchymal cells from donors or recipients that increasingly infiltrate the ocular surface serve as the source of increased secretion of IL-6, which is a major SASP driver. Agents capable of reversing the changes, including senolytic reagents or those that can suppress the SASP seen in GVHD, provide new potential targets for the treatment of GVHD. Creating innovative therapies for ocular GVHD is necessary to treat this intractable ocular disease.


Subject(s)
Aging/pathology , Dry Eye Syndromes/etiology , Graft vs Host Disease/complications , Inflammation/complications , Stress, Physiological , Animals , Chronic Disease , Fibrosis , Graft vs Host Disease/immunology , Humans , Inflammation/immunology
18.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34325468

ABSTRACT

The STING and absent in melanoma 2 (AIM2) pathways are activated by the presence of cytosolic DNA, and STING agonists enhance immunotherapeutic responses. Here, we show that dendritic cell (DC) expression of AIM2 within human melanoma correlates with poor prognosis and, in contrast to STING, AIM2 exerts an immunosuppressive effect within the melanoma microenvironment. Vaccination with AIM2-deficient DCs improves the efficacy of both adoptive T cell therapy and anti-PD-1 immunotherapy for "cold tumors," which exhibit poor therapeutic responses. This effect did not depend on prolonged survival of vaccinated DCs, but on tumor-derived DNA that activates STING-dependent type I IFN secretion and subsequent production of CXCL10 to recruit CD8+ T cells. Additionally, loss of AIM2-dependent IL-1ß and IL-18 processing enhanced the treatment response further by limiting the recruitment of regulatory T cells. Finally, AIM2 siRNA-treated mouse DCs in vivo and human DCs in vitro enhanced similar anti-tumor immune responses. Thus, targeting AIM2 in tumor-infiltrating DCs is a promising new treatment strategy for melanoma.


Subject(s)
Cancer Vaccines/immunology , DNA-Binding Proteins/immunology , Melanoma, Experimental/immunology , Melanoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Tumor Microenvironment/immunology , Young Adult
19.
Cancer Sci ; 112(8): 3163-3172, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34101300

ABSTRACT

To evaluate the feasibility of adoptive cell therapy (ACT) using ex vivo-expanded tumor-infiltrating lymphocytes (TILs) in Japanese patients with melanoma who failed immune-checkpoint inhibitor therapy, an open-label, single-arm, pilot study was conducted. We investigated the immunological and genetic factors of the pretreatment tumor and expanded TILs that may be associated with the clinical response. The treatment protocol comprised preparation of TIL culture, lympho-depleting non-myeloablative preconditioning with cyclophosphamide and fludarabine, TIL infusion, and intravenous administration of low-dose IL-2. Three patients of clinical subtypes mucosal, superficial spreading, and acral melanoma underwent TIL-ACT. Most severe adverse events, including fever and leukopenia, were manageable with the supportive regimen specified in the protocol, suggesting that the TIL-ACT regimen is suitable for Japanese patients with melanoma. One patient showed a short-term partial response, one relatively long-stable disease, and one experienced disease progression. Whole-exome and transcriptional sequencing of isolated tumor cells and immunohistochemical analyses before TIL-ACT revealed various immunostimulatory factors, including a high tumor mutation burden and immune cell-recruiting chemokines, as well as various immunosuppressive factors including TGF-ß, VEGF, Wnt/ß-catenin, and MAPK signaling and epithelial-to-mesenchymal transition, which might influence the efficacy of TIL-ACT. Our results imply mechanisms for the antitumor effect of and resistance to TIL-ACT. Further studies of immune-resistant mechanisms of TIL-ACT are warranted. This study is registered with the UMIN Clinical Trial Registry (UMIN 000011431).


Subject(s)
Cyclophosphamide/administration & dosage , Interleukin-2/administration & dosage , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/therapy , Vidarabine/analogs & derivatives , Administration, Intravenous , Cell Culture Techniques , Cyclophosphamide/therapeutic use , Feasibility Studies , Gene Regulatory Networks , Humans , Immune Checkpoint Inhibitors , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/cytology , Male , Melanoma/genetics , Melanoma/immunology , Middle Aged , Pilot Projects , Transplantation Conditioning , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/therapeutic use
20.
Jpn J Clin Oncol ; 51(8): 1232-1241, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34115870

ABSTRACT

BACKGROUND: Nivolumab, the anti-programmed cell death protein 1 antibody, has been approved for advanced melanoma, mainly based on evidence from Western countries. The profile of melanoma differs between Caucasian and Asian patients. This study was performed to obtain post-marketing data of nivolumab in Japanese patients with advanced melanoma. METHODS: This prospective, observational study involved patients with unresectable or metastatic melanoma treated with nivolumab at dosages of 2 mg/kg every 3 weeks or 3 mg/kg every 2 weeks. The primary endpoints were objective response rate and overall survival. The secondary endpoints were progression-free survival and the objective response rate according to immune-related Response Evaluation Criteria in Solid Tumours. RESULT: Among 124 patients analysed, mucosal melanoma was the most common subtype, followed by acral lentiginous, nodular, superficial spreading and lentigo maligna melanoma. Response Evaluation Criteria in Solid Tumours evaluation showed an objective response rate of 17.7%. The median survival time was 15.93 months, and the 1-year overall survival rate was 66%. Outcomes were not significantly different among melanoma subtypes. Better overall survival and/or progression-free survival but not objective response rate were associated with performance status 0, lower levels of lactate dehydrogenase, C-reactive protein and neutrophil-to-lymphocyte ratio. Patients with immune-related adverse events showed a better objective response rate, 3-month landmark overall survival and progression-free survival than patients without immune-related adverse events. CONCLUSION: The objective response rate and median survival time in Japanese patients treated with nivolumab were lower in daily practice than the >30% and >30 months, respectively, seen in global phase III trials. The occurrence of immune-related adverse events may be a predictor for survival and response to treatment with nivolumab.


Subject(s)
Melanoma , Nivolumab , Skin Neoplasms , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Female , Humans , Japan/epidemiology , Male , Melanoma/drug therapy , Middle Aged , Nivolumab/therapeutic use , Prospective Studies , Skin Neoplasms/drug therapy , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...