Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38731953

Cardiac disorders in cancer patients pose significant challenges to disease prognosis. While it has been established that these disorders are linked to cancer cells, the precise underlying mechanisms remain elusive. In this study, we investigated the impact of cancerous ascites from the rat colonic carcinoma cell line RCN9 on H9c2 cardiomyoblast cells. We found that the ascites reduced mitochondrial volume, increased oxidative stress, and decreased membrane potential in the cardiomyoblast cells, leading to apoptosis and autophagy. Although the ascites fluid contained a substantial amount of high-mobility group box-1 (HMGB1), we observed that neutralizing HMGB1 with a specific antibody mitigated the damage inflicted on myocardial cells. Our mechanistic investigations revealed that HMGB1 activated both nuclear factor κB and phosphoinositide 3-kinases-AKT signals through HMGB1 receptors, namely the receptor for advanced glycation end products and toll-like receptor-4, thereby promoting apoptosis and autophagy. In contrast, treatment with berberine (BBR) induced the expression of miR-181c-5p and miR-340-5p while suppressing HMGB1 expression in RCN9 cells. Furthermore, BBR reduced HMGB1 receptor expression in cardiomyocytes, consequently mitigating HMGB1-induced damage. We validated the myocardial protective effects of BBR in a cachectic rat model. These findings underscore the strong association between HMGB1 and cancer cachexia, highlighting BBR as a promising therapeutic agent for myocardial protection through HMGB1 suppression and modulation of the signaling system.


Apoptosis , Berberine , Cachexia , HMGB1 Protein , Animals , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Berberine/pharmacology , Rats , Cachexia/metabolism , Cachexia/drug therapy , Cachexia/etiology , Cachexia/pathology , Apoptosis/drug effects , Cell Line, Tumor , Autophagy/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Male , Disease Models, Animal , Signal Transduction/drug effects , Oxidative Stress/drug effects , Toll-Like Receptor 4/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Receptor for Advanced Glycation End Products/metabolism , Rats, Sprague-Dawley , Neoplasms/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism
2.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article En | MEDLINE | ID: mdl-38474261

Patients with cancer die from cardiac dysfunction second only to the disease itself. Cardiotoxicity caused by anticancer drugs has been emphasized as a possible cause; however, the details remain unclear. To investigate this mechanism, we treated rat cardiomyoblast H9c2 cells with sunitinib, lapatinib, 5-fluorouracil, and cisplatin to examine their effects. All anticancer drugs increased ROS, lipid peroxide, and iron (II) levels in the mitochondria and decreased glutathione peroxidase-4 levels and the GSH/GSSG ratio. Against this background, mitochondrial iron (II) accumulates through the unregulated expression of haem oxygenase-1 and ferrochelatase. Anticancer-drug-induced cell death was suppressed by N-acetylcysteine, deferoxamine, and ferrostatin, indicating ferroptosis. Anticancer drug treatment impairs mitochondrial DNA and inhibits oxidative phosphorylation in H9c2 cells. Similar results were observed in the hearts of cancer-free rats treated with anticancer drugs in vitro. In contrast, treatment with pterostilbene inhibited the induction of ferroptosis and rescued the energy restriction induced by anticancer drugs both in vitro and in vivo. These findings suggest that induction of ferroptosis and inhibition of oxidative phosphorylation are mechanisms by which anticancer drugs cause myocardial damage. As pterostilbene ameliorates these mechanisms, it is expected to have significant clinical applications.


Antineoplastic Agents , Ferroptosis , Humans , Rats , Animals , Oxidative Phosphorylation , Antineoplastic Agents/pharmacology , Cell Death , Iron/metabolism
3.
Biochem Biophys Res Commun ; 696: 149542, 2024 Feb 12.
Article En | MEDLINE | ID: mdl-38244315

PDZ domain-containing RING finger family protein 3 (PDZRN3) is expressed in various tissues, including the skeletal muscle. Although PDZRN3 plays a crucial role in the terminal differentiation of myoblasts and synaptic growth/maturation in myogenesis, the role of this molecule in postnatal muscles is completely unknown despite its lifelong expression in myofibers. In this study, we aimed to elucidate the function of PDZRN3 in mature myofibers using myofiber-specific conditional knockout mice. After tamoxifen injection, PDZRN3 deficiency was confirmed in both fast and slow myofibers of Myf6-CreERT2; Pdzrn3flox/flox (Pdzrn3mcKO) mice. Transcriptome analysis of the skeletal muscles of Pdzrn3mcKO mice identified differentially expressed genes, including muscle atrophy-related genes such as Smox, Amd1/2, and Mt1/2, suggesting that PDZRN3 is involved in the homeostatic maintenance of postnatal muscles. PDZRN3 deficiency caused muscle atrophy, predominantly in fast-twitch (type II) myofibers, and reduced muscle strength. While myofiber-specific PDZRN3 deficiency did not influence endplate morphology or expression of neuromuscular synaptic formation-related genes in postnatal muscles, indicating that the relationship between PDZRN3 and neuromuscular junctions might be limited during muscle development. Considering that the expression of Pdzrn3 in skeletal muscles was significantly lower in aged mice than in mature adult mice, we speculated that the PDZRN3-mediated muscle maintenance system might be associated with the pathophysiology of age-related muscle decline, such as sarcopenia.


Muscle, Skeletal , Sarcopenia , Mice , Animals , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Neuromuscular Junction/pathology , Sarcopenia/pathology , Myoblasts/metabolism , Mice, Knockout , Ubiquitin-Protein Ligases/metabolism
4.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article En | MEDLINE | ID: mdl-36142607

Long non-coding RNAs (lncRNAs) play critical roles in human cancers. HOXA11 anti-sense RNA (HOXA11-AS) is an lncRNA belonging to the homeobox (HOX) gene cluster that promotes liver metastasis in human colon cancer. However, its role and mechanism of action in human oral squamous cell carcinoma (OSCC) are unclear. In this study, we investigated HOXA11-AS expression and function in human OSCC tissues and cell lines, as well as a mouse model of OSCC. Our analyses showed that HOXA11-AS expression in human OSCC cases correlates with lymph node metastasis, nicotinamide adenine dinucleotide (NAD)(P)H: quinone oxidoreductase 1 (NQO1) upregulation, and dihydronicotinamide riboside (NRH): quinone oxidoreductase 2 (NQO2) downregulation. Using the human OSCC cell lines HSC3 and HSC4, we demonstrate that HOXA11-AS promotes NQO1 expression by sponging microRNA-494. In contrast, HOXA11-AS recruits zeste homolog 2 (EZH2) to the NQO2 promoter to suppress its expression via the trimethylation of H3K27. The upregulation of NQO1 enzymatic activity by HOXA11-AS results in the consumption of flavin adenine dinucleotide (FAD), which reduces FAD-requiring glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity and suppresses glycolysis. However, our analyses show that lactic acid fermentation levels are preserved by glutaminolysis due to increased malic enzyme-1 expression, promoting enhanced proliferation, invasion, survival, and drug resistance. In contrast, suppression of NQO2 expression reduces the consumption of NRH via NQO2 enzymatic activity and increases NAD levels, which promotes enhanced stemness and metastatic potential. In mouse tumor models, knockdown of HOXA11-AS markedly suppressed tumor growth and lung metastasis. From these findings, targeting HOXA11-AS may strongly suppress high-grade OSCC by regulating both NQO1 and NQO2.


Carcinoma, Squamous Cell , Homeodomain Proteins/metabolism , MicroRNAs , Mouth Neoplasms , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinone Reductases/metabolism , RNA, Long Noncoding , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Flavin-Adenine Dinucleotide/genetics , Genes, Homeobox , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Lactic Acid , Mice , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , NAD/genetics , Quinones , RNA, Antisense , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics
5.
Cancer Sci ; 113(8): 2904-2915, 2022 Aug.
Article En | MEDLINE | ID: mdl-35570394

High mobility group box-1 (HMGB1) is known to be a chemotactic factor for mesenchymal stem/stromal cells (MSCs), but the effect of post-translational modification on its function is not clear. In this study, we hypothesized that differences in the oxidation state of HMGB1 would lead to differences in the function of MSCs in cancer. In human colorectal cancer, MSCs infiltrating into the stroma were correlated with liver metastasis and serum HMGB1. In animal models, oxidized HMGB1 mobilized three-fold fewer MSCs to subcutaneous tumors compared with reduced HMGB1. Reduced HMGB1 inhibited the proliferation of mouse bone marrow MSCs (BM-MSCs) and induced differentiation into osteoblasts and vascular pericytes, whereas oxidized HMGB1 promoted proliferation and increased stemness, and no differentiation was observed. When BM-MSCs pretreated with oxidized HMGB1 were co-cultured with syngeneic cancer cells, cell proliferation and stemness of cancer cells were increased, and tumorigenesis and drug resistance were promoted. In contrast, co-culture with reduced HMGB1-pretreated BM-MSCs did not enhance stemness. In an animal orthotopic transplantation colorectal cancer model, oxidized HMGB1, but not reduced HMGB1, promoted liver metastasis with intratumoral MSC chemotaxis. Therefore, oxidized HMGB1 reprograms MSCs and promotes cancer malignancy. The oxidized HMGB1-MSC axis may be an important target for cancer therapy.


Colorectal Neoplasms , HMGB1 Protein , Liver Neoplasms , Mesenchymal Stem Cells , Animals , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Colorectal Neoplasms/pathology , HMGB1 Protein/metabolism , Humans , Liver Neoplasms/secondary , Mice
6.
J Infect Chemother ; 27(9): 1306-1310, 2021 Sep.
Article En | MEDLINE | ID: mdl-33952418

INTRODUCTION: Tuberculin skin test (TST) has been used to diagnose tuberculosis (TB) and latent tuberculosis infection (LTBI). However, in Bacillus Calmette-Guérin (BCG) vaccinated patients, TST tends to produce false-positive results. According to the previous vaccination schedule, Japanese people were mandated to receive up to three doses of BCG-vaccine. The vaccination schedule was changed in 2003 and as per the new schedule, only infants are administered a dose of BCG vaccine. Our hypothesis is that this change can lead to a reduction in the cross-reaction to TST. METHODS: We evaluated the TST results obtained from 1097 recruits from six defense camps and 667 recruits from an air base. These TST data were divided into two groups according to the date of birth: a new group and an old group according to the BCG immunization schedule. We then analyzed positive and negative reaction of TST and erythema sizes. RESULTS: We confirmed that the change in BCG-vaccination schedule significantly decreased TST false-positive reaction (Pmeta = 1.4 × 10-18; risk ratio = 0.83; 95% confidence interval: 0.80-0.87) and erythema size (Pmeta = 1.1 × 10-4; mean difference = 6.6 mm; 95% confidence interval: 3.2 mm-9.9 mm). CONCLUSIONS: We showed the reduction in BCG cross-reaction to TST, in the new BCG vaccination schedule group, compared to the old group, we also have extracted information on the improvement in the specificity of TST for LTBI and TB diagnosis, which resulted from BCG schedule change.


BCG Vaccine , Tuberculosis , Humans , Immunization Schedule , Infant , Japan , Tuberculin Test , Tuberculosis/diagnosis , Tuberculosis/prevention & control
7.
Oncotarget ; 11(41): 3712-3722, 2020 Oct 13.
Article En | MEDLINE | ID: mdl-33110478

Myocardial damage in cancer patients is emphasized as a cause of death; however, there are not many murine cachexia models to evaluate cancer-derived heart disorder. Using the mouse cachexia model that we established previously, we investigated myocardial damage in tumor-bearing mice. In cachexic mice, decreased heart weight and myocardial volume, and dilated left ventricular lumen, and atrophied cardiomyocytes were noted. The cardiomyocytes also showed accumulated 8-hydroxydeoxyguanosine, decreased leucine zipper and EF-hand-containing transmembrane protein-1, and increased microtubule-associated protein light chain3-II. Levels of tumor necrosis factor-α and high-mobility group box-1 proteins in the myocardium were increased, and nuclear factor κB, a signaling molecule associated with these proteins, was activated. When rat cardiomyoblasts (H9c2 cells) were treated with mouse cachexia model ascites and subjected to flux analysis, both oxidative phosphorylation and glycolysis were suppressed, and the cells were in a quiescent state. These results are in good agreement with those previously reported on cancerous myocardial damage. The established mouse cachexia model can therefore be considered useful for analyzing cancer-derived myocardial damage.

8.
Cancer Sci ; 111(12): 4605-4615, 2020 Dec.
Article En | MEDLINE | ID: mdl-32969559

Cancer-derived myocardial damage is an important cause of death in cancer patients. However, the development of dietary interventions for treating such damage has not been advanced. Here, we investigated the effect of dietary intervention with lauric acid (LAA) and glucose, which was effective against skeletal muscle sarcopenia in a mouse cachexia model, on myocardial damage. Treatment of H9c2 rat cardiomyoblasts with lauric acid promoted mitochondrial respiration and increased ATP production by Seahorse flux analysis, but did not increase oxidative stress. Glycolysis was also promoted by LAA. In contrast, mitochondrial respiration and ATP production were suppressed, and oxidative stress was increased in an in vitro cachexia model in which cardiomyoblasts were treated with mouse cachexia ascites. Ascites-treated H9c2 cells with concurrent treatment with LAA and high glucose showed that mitochondrial respiration and glycolysis were promoted more than that of the control, and ATP was restored to the level of the control. Oxidative stress was also reduced by the combined treatment. In the mouse cachexia model, myocardiac atrophy and decreased levels of a marker of muscle maturity, SDS-soluble MYL1, were observed. When LAA in CE-2 diet was orally administered alone, no significant rescue was observed in the cancer-derived myocardial disorder. In contrast, combined oral administration of LAA and glucose recovered myocardial atrophy and MYL1 to levels observed in the control without increase in the cancer weight. Therefore, it is suggested that dietary intervention using a combination of LAA and glucose for cancer cachexia might improve cancer-derived myocardial damage.


Cachexia/diet therapy , Glucose/pharmacology , Lauric Acids/pharmacology , Muscular Atrophy/diet therapy , Myocytes, Cardiac/drug effects , Adenosine Triphosphate/biosynthesis , Animals , Cachexia/complications , Cachexia/pathology , Cell Line , Cell Line, Tumor , Energy Metabolism/drug effects , Glucose/administration & dosage , Glycolysis/drug effects , Lauric Acids/administration & dosage , Male , Mice , Mice, Inbred BALB C , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Oxidative Stress/drug effects , Promyelocytic Leukemia Protein/metabolism , Sarcopenia/diet therapy , Sarcopenia/etiology , Sarcopenia/pathology
9.
Radiol Case Rep ; 15(9): 1480-1484, 2020 Sep.
Article En | MEDLINE | ID: mdl-32670445

Four cases (age range, 60-78 years, male:female = 1:3) who had undergone cholecystectomy presented with fever (n = 1), right abdominal pain with fever (n = 1), appetite loss with fever (n = 1), and absence of symptoms (n = 1). Computed tomography (CT) showed an irregular-shaped invasive mass or fluid collection in the right Morrison's pouch, right paracolic gutter, gallbladder fossa, subphrenic space, or abdominal wall. CT and ultrasound revealed gallstones in the granuloma in 3 cases and an abscess in one case. The inflammatory process induced by dropped gallstones may mimic peritoneal malignancies. Awareness of cholecystectomy and the detection of gallstones in the lesion are essential for the diagnosis of dropped gallstones.

10.
Pathobiology ; 86(5-6): 254-262, 2019.
Article En | MEDLINE | ID: mdl-31722347

Necrosis-inducing anticancer drugs enhance high-mobility group box 1 (HMGB1) release during cell necrosis, and HMGB1-induced autophagy in skeletal muscle induces muscle atrophy. We evaluated the efficacy of magnetic hyperthermia therapy (MHT) using a low-energy magnetic field and self-controlled heating elements in tumor treatment. MHT-induced apoptosis by heating mouse subcutaneous tumors at 43°C using a heat-controlling iron-aluminum (Fe-Al) milling alloy. In contrast, MHT using Fe line-induced necrosis by heating to approximately 100°C. Furthermore, MHT with Fe-Al milling alloy reduced stemness. In hyperthermia using age line or Fe-Al milling alloy, both of them provided histological degeneration in skeletal muscle; however, qualitative differences were observed. MHT using Fe-line induced pronounced autophagy, decrease of myosin heavy chain content, and increase in serum HMGB1. In contrast, MHT using Fe-Al milling alloy induced heat shock protein 90 but no autophagy and decreased serum HMGB1. Therefore, MHT using Fe-Al milling alloy might be a good method for local treatment of tumors to reduce skeletal muscle atrophy.


Alloys , Apoptosis , Hyperthermia, Induced/instrumentation , Hyperthermia, Induced/methods , Magnetic Phenomena , Muscle, Skeletal/pathology , Aluminum/chemistry , Animals , Autophagy , Iron/chemistry , Male , Mice , Mice, Inbred BALB C , Muscular Atrophy/pathology , Necrosis
11.
Cancer Sci ; 110(10): 3391-3399, 2019 Oct.
Article En | MEDLINE | ID: mdl-31432554

Skeletal muscle volume is associated with prognosis of cancer patients. Maintenance of skeletal muscle is an essential concern in cancer treatment. In nutritional intervention, it is important to focus on differences in metabolism between tumor and skeletal muscle. We examined the influence of oral intake of glucose (0%, 10%, 50%) and 2% medium-chain fatty acid (lauric acid, LAA, C12:0) on tumor growth and skeletal muscle atrophy in mouse peritoneal metastasis models using CT26 mouse colon cancer cells and HT29 human colon cancer cells. After 2 weeks of experimental breeding, skeletal muscle and tumor were removed and analyzed. Glucose intake contributed to prevention of skeletal muscle atrophy in a sugar concentration-dependent way and also promoted tumor growth. LAA ingestion elevated the level of skeletal muscle protein and suppressed tumor growth by inducing tumor-selective oxidative stress production. When a combination of glucose and LAA was ingested, skeletal muscle mass increased and tumor growth was suppressed. Our results confirmed that although glucose is an important nutrient for the prevention of skeletal muscle atrophy, it may also foster tumor growth. However, the ingestion of LAA inhibited tumor growth, and its combination with glucose promoted skeletal muscle integrity and function, without stimulating tumor growth. These findings suggest novel strategies for the prevention of skeletal muscle atrophy.


Colonic Neoplasms/drug therapy , Glucose/administration & dosage , Lauric Acids/administration & dosage , Muscular Atrophy/prevention & control , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/complications , Disease Models, Animal , Drug Therapy, Combination , Glucose/adverse effects , Glucose/pharmacology , HT29 Cells , Humans , Lauric Acids/pharmacology , Male , Mice , Muscular Atrophy/etiology , Neoplasm Transplantation , Oxidative Stress/drug effects
12.
J Phys Ther Sci ; 31(5): 462-465, 2019 May.
Article En | MEDLINE | ID: mdl-31164786

[Purpose] We investigated whether an increase or decrease in subcutaneous fat mass secondary to cardiac cachexia can be evaluated using diagnostic ultrasonography in patients with heart failure. [Participant and Methods] We report a case of cardiac cachexia in a patient in whom cachexia was confirmed by weight loss, decreased dietary intake, and biochemical indicators measured by blood tests. We measured the subcutaneous fat mass in the patient's thigh using ultrasonic diagnostic equipment during the cachectic state, as well as 1 and 2 months later. [Results] An increase in weight and ultrasonographically documented femoral subcutaneous fat mass was confirmed by improvement in heart failure-induced cachexia. [Conclusion] Clinically convenient ultrasonic diagnostic equipment is useful to assess subcutaneous fat mass, which serves as an indicator of the degree of cachexia.

13.
Mol Cancer Ther ; 18(8): 1430-1438, 2019 08.
Article En | MEDLINE | ID: mdl-31171582

In clinical N0 (cN0) cases with head and neck squamous cell carcinoma (HNSCC), a treatment selection is still controversial: elective neck dissection or watchful waiting. We focused on sentinel lymph node (SLN)-targeted therapy using the urokinase-type plasminogen activator (uPA)-dependent oncolytic Sendai virus "BioKnife." The objectives of this study were to investigate BioKnife migration into SLNs and elucidate its antitumor effect on lymph node metastases (LNM). We established an orthotopic nude mouse model of HNSCC, with LNM being frequently induced. We inoculated HSC-3-M3, human highly metastatic tongue squamous cell carcinoma cells, in the tongue of the nude mice, and after 2 weeks, we injected BioKnife into the primary tumor. We tracked BioKnife migration into the SLNs by immunostaining, RT-PCR, and an in vivo imaging system. We also examined its antitumor effects and mechanisms through serial section analysis of lymph nodes. GFP reporter expression was clearly visible in the lymph nodes of virus groups, which corresponded to SLNs. Relative GFP mRNA was significantly increased in both the tongues and lymph nodes in the virus groups compared with that in the control group (P < 0.05). Serial section analysis showed that BioKnife infected cancer cells and exhibited significant antitumor effect against LNM compared with the control groups (P < 0.05). We detected apoptosis in LNM infected by BioKnife. BioKnife migrated into SLNs after its injection into the primary tumor and effectively suppressed LNM, suggesting that SLN-targeted therapy using BioKnife has great potential to provide a novel and promising alternative to elective neck dissection in cN0 patients with HNSCC.


Oncolytic Virotherapy , Oncolytic Viruses , Sendai virus , Sentinel Lymph Node/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Humans , Lymphatic Metastasis , Mice , Neoplasm Micrometastasis , Oncolytic Viruses/genetics , Sendai virus/genetics , Sentinel Lymph Node/metabolism , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Xenograft Model Antitumor Assays
14.
Pathobiology ; 86(4): 182-189, 2019.
Article En | MEDLINE | ID: mdl-31132784

CD47 activates signal regulatory protein alpha expressed on macrophages and suppresses its phagocytic ability; therefore, CD47 is drawing attention as an immune checkpoint in the innate immune system. Expression of CD47 in cancer is thought to allow cancer cells to escape antitumor immunity of the innate immune system. In this study, expression of CD47 was examined by immunostaining in colorectal cancer (CRC) and compared with the expression of CD44, which is a marker for cancer stem cells. In 95 cases of stage II-IV CRC, CD47 and CD44 showed overexpression in 82 and 80 cases, respectively. Both expression levels correlated with distant metastasis. Moreover, the expression of CD47 and CD44 in each case showed a significant correlation. In stage III cases, disease-free survival of cases showing high expression of CD47 and CD44 was worse than that of the cases with low expression. Furthermore, 3 of the stage IV cases were administered nivolumab, a checkpoint inhibitor of the acquired immune system, and 2 patients showed recurrence thereafter. All recurrent tumors highly expressed CD47 and CD44 and showed the epithelial-mesenchymal transition (EMT) phenotype. Our results suggest that CD47 promotes the malignancy of CRC in association with EMT and enhances the stemness of cancer cells. Moreover, our study suggests that CD47 and CD44 are involved in imparting resistance to programmed cell death (PD)-1/PD-ligand 1 inhibitors.


CD47 Antigen/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Hyaluronan Receptors/genetics , Adult , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplastic Stem Cells/cytology , Nivolumab/therapeutic use
15.
Head Neck ; 41(9): 2873-2882, 2019 09.
Article En | MEDLINE | ID: mdl-30969451

BACKGROUND: This study was designed to assess the therapeutic effect of urokinase-targeted recombinant oncolytic Sendai virus, termed "BioKnife," on anaplastic thyroid carcinoma (ATC). METHODS: Urokinase activity was investigated in human ATC cell lines, and BioKnife cytotoxicity against the cell lines was evaluated in vitro. Orthotopic mouse models of ATC were treated with three intratumoral injections of BioKnife, control virus, or phosphate-buffered saline (PBS) and were observed daily until >20% weight loss occurred. RESULTS: All three ATC cell lines showed a high level of urokinase activity. BioKnife induced urokinase-dependent cell fusion and cytotoxicity in all cell lines. Orthotopic models treated with BioKnife showed significantly prolonged survival compared with models treated with control virus or PBS (BioKnife 41.6 ± 15.0, control virus 17.0 ± 2.9, PBS 17.7 ± 6.3 days). CONCLUSIONS: BioKnife exerted therapeutic effects in orthotopic ATC mouse models. Thus, BioKnife represents a possible treatment option for ATC.


Oncolytic Virotherapy , Sendai virus/physiology , Thyroid Carcinoma, Anaplastic/therapy , Thyroid Neoplasms/therapy , Urokinase-Type Plasminogen Activator/metabolism , Animals , Apoptosis , Cell Fusion , Cell Line, Tumor , Disease Models, Animal , Giant Cells/metabolism , Mice, Inbred BALB C , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Head Neck ; 41(6): 1676-1686, 2019 06.
Article En | MEDLINE | ID: mdl-30620422

BACKGROUND: The objectives of this study were to demonstrate anti-metastatic effect of BioKnife, uPA activity-dependent oncolytic Sendai virus, after BioKnife treatment for primary tumor, and analyze its mechanisms in a simulated metastasis mouse model of head and neck squamous cell carcinoma (HNSCC). METHODS: We established a simulated metastasis mouse model using a murine HNSCC cell line "SCCVII." We assessed a tumor size and an induction of tumor-specific immunoresponses using cytotoxic T-lymphocyte (CTL) assay, flow cytometry (FCM) in spleen and immunohistochemistry (IHC) in secondary tumor. RESULTS: Secondary tumors were significantly smaller in BioKnife-treated group. CTL activities were significantly improved in BioKnife group. FCM revealed that induction of dendritic cells and CD4+ /CD8+ lymphocytes was significantly higher in BioKnife group. IHC showed that CD8+ lymphocytes invaded secondary tumor. CONCLUSION: Tumor-specific immunoresponses induced by BioKnife has great potential to be a novel, safe, and less invasive option for control and prevention of metastasis.


Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Sendai virus/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Dendritic Cells/metabolism , Disease Models, Animal , Immunocompetence , Mice, Inbred C3H , Urokinase-Type Plasminogen Activator , Xenograft Model Antitumor Assays
17.
Lipids Health Dis ; 17(1): 258, 2018 Nov 17.
Article En | MEDLINE | ID: mdl-30447697

BACKGROUND: Oral intake of medium-chain fatty acids (MCFAs) reportedly suppresses the accumulation of visceral fat and has antitumor effects in tumor-bearing animals. MCFAs penetrate the mitochondrial membrane in a carnitine shuttle-independent manner and are metabolized more quickly than long-chain fatty acids. Based on these characteristics, MCFAs may have pronounced effects in mitochondria-rich tissues, such as the myocardium. We examined the effect of oral intake of MCFAs on the heart. METHODS: We fed BALB/c mice with a control diet supplemented with 0%, 2%, 5%, or 10% lauric acid (LAA; a 12-carbon saturated MCFA). After euthanasia, the hearts, both sides of quadriceps femoris muscle (QFM) and epididymal fat pad (EFP) were excised and weighed. Then myocardial tissue morphology, oxidative stress accumulation, and mitochondrial volume were observed by histological analysis. The expression levels of myosin light chain 1 were measured by ELISA. RESULTS: There were no differences among the groups in food and calorie intake, but the intake of LAA increased with the dietary proportion. The 10%-LAA-fed mice experienced significant weight loss and became moribund on day 6. The body, cardiac and EFP weights of the mice fed 5% and 10% LAA were lower than those of the control group. And 10% LAA fed group showed significant decrease of the QFM weights. Protein analysis of the excised hearts revealed higher expression of myosin light chain 1 in the 5% group than in the control group. Histological examination of the hearts revealed myocardial atrophy and accumulation of oxidative stress in the 10% group. Fewer mitochondria were observed with increased LAA intake. CONCLUSIONS: Excessive LAA consumption may damage the myocardium and the damage might result from oxidative stress accumulation and cellular atrophy.


Fatty Acids/adverse effects , Muscular Atrophy/etiology , Myocardium/metabolism , Oxidative Stress/drug effects , Quadriceps Muscle/drug effects , Animals , Body Weight/drug effects , Fatty Acids/chemistry , Heart/drug effects , Lauric Acids/adverse effects , Male , Mice , Mice, Inbred BALB C , Muscular Atrophy/metabolism
18.
Int J Oncol ; 52(2): 413-423, 2018 Feb.
Article En | MEDLINE | ID: mdl-29345287

The effect of intermittent calorie restriction (ICR) on cancer is controversial. In this study, we examined the effects of ICR and food content in syngeneic BALB/c mice injected with CT26 mouse colon cancer cells. Mice were subjected to 24-h fasting once a week for 4 weeks, and then provided with a control, high-calorie, or trans fatty acid-rich diet. While ICR resulted in increases in tumor weights, metastasis and in the number of cancer stem cells (CSCs) in the tumors or blood of mice fed the control and high-fat diets, it had no effect on body weight after 4 weeks. In particular, we detected increases in the numbers of CSCs in the tumor or blood on the day after starvation, when food overconsumption was detected. Conversely, continuous calorie restriction had no effect on tumor weight, metastasis, or the number of CSCs in tumors or blood. In the post-starvation period, energy metabolism in the tumor was altered from oxidative phosphorylation to glycolysis/lactate fermentation, with the acquisition of the epithelial-mesenchymal transition (EMT) phenotype. Hyperglycemia at the post-starvation period induced the expression of insulin-like growth factor-1, hypoxia-induced factor-1α and Nanog, as well as the phosphorylation of Stat3. Taken together, these findings suggest that ICR induces an increase in the number of CSCs and enhances EMT by promoting the Warburg/Crabtree effect following post-fasting food overconsumption.


Caloric Restriction/methods , Energy Metabolism/physiology , Epithelial-Mesenchymal Transition , Neoplasms, Experimental/diet therapy , Neoplasms, Experimental/pathology , Animals , Cell Line, Tumor , Diet, High-Fat/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Male , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oleic Acid/adverse effects , Oleic Acids , Trans Fatty Acids/adverse effects
19.
Acta Otolaryngol ; 137(4): 403-410, 2017 Apr.
Article En | MEDLINE | ID: mdl-27976979

CONCLUSION: The resection of the medial and lateral pyriform sinus was associated with post-operative voice impairment after TOVS. Scar contracture around the cricoarytenoid joint lead to arytenoid fixation toward lateral position, and this wound healing process caused insufficient glottis closure. Although oncological and functional outcomes of TOVS was satisfactory, surgeons should mention the risk of post-operative voice impairment in pre-operative counseling. OBJECTIVES: Transoral surgery is a minimally invasive treatment option for hypopharyngeal and supraglottic cancer. Post-operative vocal function was satisfactory in most cases, but in some cases vocal cord was fixed and occasionally voice impairment persists. METHODS: Vocal function of 55 patients who underwent transoral videolaryngoscopic surgery (TOVS) for hypopharyngeal and supraglottic cancers was evaluated by the GRBAS perceptive scale, aerodynamic tests and acoustic analyses, and the Voice Handicap Index questionnaire. The risk factors for voice impairment were identified. RESULTS: Voice impairment (G score ≧2) was found in 16 cases (29.1%). Univariate analysis revealed that the resection of medial and lateral pyriform sinus (p = .0018) and neck dissection (p = .0421) were associated with post-operative voice impairment. Multivariate analysis revealed that the resection of medial and lateral pyriform sinus (p = .0021) was associated with post-operative voice impairment.


Hypopharyngeal Neoplasms/surgery , Laryngoscopy/adverse effects , Voice Disorders/etiology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Phonation , Postoperative Complications/etiology , Pyriform Sinus/surgery , Retrospective Studies , Speech Acoustics , Voice
20.
Cancer Med ; 5(3): 415-25, 2016 Mar.
Article En | MEDLINE | ID: mdl-26798989

Improved therapies for individuals with head and neck squamous cell carcinoma (HNSCC) may be developed by identification of appropriate biomarkers. The aim of this study was to evaluate the usefulness of serum midkine measurement as a biomarker for HNSCC. Pretreatment serum midkine concentrations were measured in 103 patients with HNSCC and 116 control individuals by enzyme-linked immunosorbent assay. Midkine expression in tumor tissues from 33 patients with HNSCC who underwent definitive surgical resection without preoperative treatment was examined by immunohistochemistry. The cut-off serum midkine concentrations for predicting the presence of head and neck malignancy and chemosensitivity to induction chemotherapy, as determined using receiver operating characteristic curves, were 482 and 626 pg/mL, respectively. Spearman bivariate correlations showed positive correlations between serum midkine levels and immunohistochemistry staining score (r = 0.612, P < 0.001). The sensitivity, specificity, positive predictive value, negative predictive value, and accuracy of serum midkine concentration for detection of HNSCC were 57.3, 85.3, 77.6, 69.2, and 72.1%, respectively. However, for predicting the response to induction chemotherapy, the values were 84.6, 60.9, 71.0, 77.8, and 73.5%, respectively. Serum midkine concentration was identified as an independent prognostic factor by multivariate analysis, using Cox's proportional hazards model (P = 0.027). Overexpression of serum midkine yielded a relative risk of death of 3.77, with 95% confidence limits ranging from 1.15 to 17.0. Serum midkine levels in patients with HNSCC were associated with malignancy, chemosensitivity, and prognosis. Serum midkine may be a useful, minimally invasive biomarker for early detection, therapeutic decision-making, and predicting prognosis.


Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Nerve Growth Factors/blood , Adult , Aged , Aged, 80 and over , Early Detection of Cancer , Female , Humans , Male , Middle Aged , Midkine , Prognosis , ROC Curve , Squamous Cell Carcinoma of Head and Neck , Survival Analysis
...