Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 403
Filter
1.
Cell Rep ; 36(8): 109537, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433039

ABSTRACT

Increased DNA replication and metastasis are hallmarks of cancer progression, while deregulated proliferation often triggers sustained replication stresses in cancer cells. How cancer cells overcome the growth stress and proceed to metastasis remains largely elusive. Proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA replication machinery. Here, we show that phosphorylation of PCNA on tyrosine 211 (pY211-PCNA) regulates DNA metabolism and tumor microenvironment. Abrogation of pY211-PCNA blocks fork processivity, resulting in biogenesis of single-stranded DNA (ssDNA) through a MRE11-dependent mechanism. The cytosolic ssDNA subsequently induces inflammatory cytokines through a cyclic GMP-AMP synthetase (cGAS)-dependent cascade, triggering an anti-tumor immunity by natural killer (NK) cells to suppress distant metastasis. Expression of pY211-PCNA is inversely correlated with cytosolic ssDNA and associated with poor survival in patients with cancer. Our results pave the way to biomarkers and therapies exploiting immune responsiveness to target metastatic cancer.


Subject(s)
Neoplasms, Experimental/immunology , Proliferating Cell Nuclear Antigen/immunology , Tumor Escape , Tumor Microenvironment/immunology , Animals , Female , Humans , MCF-7 Cells , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Tumor Microenvironment/genetics , Tyrosine/genetics , Tyrosine/immunology
2.
Exp Oncol ; 42(2): 120-125, 2020 06.
Article in English | MEDLINE | ID: mdl-32602294

ABSTRACT

AIM: To estimate immunogenicity and antitumor effect of new DNA vaccine against neuroblastoma using tyrosine hydroxylase as an antigen and linear polyethylenimine (PEI) 20 kDa as a synthetic DNA carrier in syngeneic mouse tumor model. MATERIALS AND METHODS: DNA vaccine was made by cloning the tyrosine hydroxylase minigene fused to the potato virus X coat protein gene into the expression vector. The A/J mice were vaccinated by three intramuscular injections. For immunogenicity study, immune response was estimated by target cells cytotoxicity assay, interferon-gamma production in enzyme-linked immunospot assay and antigen-specific antibodies in 14 days after the final vaccination. Antitumor effect was assessed by measurement of tumor volume and event-free survival rate in mice with engrafted NB41A3 murine neuroblastoma cells following three intramuscular injections of the vaccine: 7 days before, 5 and 10 days after tumor engraftment. The immune response was also assessed on the 30th day after tumor engraftment. RESULTS: The immunogenicity and antitumor effect of the vaccine in the form of aqueous solution of DNA and DNA-PEI conjugate were compared. Splenocytes cytotoxicity was the highest in the group of DNA-PEI vaccines (37.3 ± 6.9% lysis of target cells) compared with the unconjugated DNA vaccine (26.2 ± 4.0%) and placebo control (21.9 ± 3.7%). The production of interferon-gamma in the enzyme-linked immunospot assay was about ten times higher in the DNA-PEI group than in the other groups. The vaccine slowed or prevented the growth of the tumor. Mice vaccinated with the DNA-PEI vaccine had significantly better survival compared to control group (p < 0.0003). CONCLUSIONS: DNA vaccine against tyrosine hydroxylase, administered as a DNA-PEI 20 kDa conjugate, slows down the growth of neuroblastoma cells engrafted to mice.


Subject(s)
Cancer Vaccines/pharmacology , Neuroblastoma/therapy , Vaccines, Conjugate/pharmacology , Vaccines, DNA/pharmacology , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Female , Immunity, Cellular/drug effects , Immunotherapy/methods , Interferon-gamma/metabolism , Male , Mice, Inbred Strains , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neuroblastoma/immunology , Neuroblastoma/mortality , Neuroblastoma/pathology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology , Vaccines, DNA/chemistry , Vaccines, DNA/immunology
3.
Int J Mol Sci ; 21(11)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545380

ABSTRACT

Glioblastoma (GBM) present with an abundant and aberrant tumor neo-vasculature. While rapid growth of solid tumors depends on the initiation of tumor angiogenesis, GBM also progress by infiltrative growth and vascular co-option. The angiogenic factor apelin (APLN) and its receptor (APLNR) are upregulated in GBM patient samples as compared to normal brain tissue. Here, we studied the role of apelin/APLNR signaling in GBM angiogenesis and growth. By functional analysis of apelin in orthotopic GBM mouse models, we found that apelin/APLNR signaling is required for in vivo tumor angiogenesis. Knockdown of tumor cell-derived APLN massively reduced the tumor vasculature. Additional loss of the apelin signal in endothelial tip cells using the APLN-knockout (KO) mouse led to a further reduction of GBM angiogenesis. Direct infusion of the bioactive peptide apelin-13 rescued the vascular loss-of-function phenotype specifically. In addition, APLN depletion massively reduced angiogenesis-dependent tumor growth. Consequently, survival of GBM-bearing mice was significantly increased when APLN expression was missing in the brain tumor microenvironment. Thus, we suggest that targeting vascular apelin may serve as an alternative strategy for anti-angiogenesis in GBM.


Subject(s)
Apelin/metabolism , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/pathology , Animals , Apelin/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Magnetic Resonance Imaging , Mice, Knockout , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/mortality , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
4.
Cancer Lett ; 477: 10-18, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32112900

ABSTRACT

Medulloblastoma (MB) is the most frequent malignant brain tumour in children with a poor outcome. Divided into four molecular subgroups, MB of the Sonic hedgehog (SHH) subgroup accounts for approximately 25% of the cases and is driven by mutations within components of the SHH pathway, such as its receptors PTCH1 or SMO. A fraction of these cases additionally harbour PIK3CA mutations, the relevance of which is so far unknown. To unravel the role of Pik3ca mutations alone or in combination with a constitutively activated SHH signalling pathway, transgenic mice were used. These mice show mutated variants within Smo, Ptch1 or Pik3ca genes in cerebellar granule neuron precursors, which represent the cellular origin of SHH MB. Our results show that Pik3ca mutations alone are insufficient to cause developmental alterations or to initiate MB. However, they significantly accelerate the growth of Shh MB, induce tumour spread throughout the cerebrospinal fluid, and result in lower survival rates of mice with a double Pik3caH1047R/SmoM2 or Pik3caH1047R/Ptch1 mutation. Therefore, PIK3CA mutations in SHH MB may represent a therapeutic target for first and second line combination treatments.


Subject(s)
Cerebellar Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Medulloblastoma/genetics , Mutation , Animals , Cerebellar Neoplasms/pathology , Disease Models, Animal , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/pathology , Mice, Transgenic , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Patched-1 Receptor/genetics , Smoothened Receptor/genetics , Spinal Cord Neoplasms/secondary , Survival Rate , Whole Genome Sequencing
5.
J Control Release ; 306: 138-148, 2019 07 28.
Article in English | MEDLINE | ID: mdl-31176656

ABSTRACT

The increasing use in the last decade of PEGylated nanodrugs such as Doxil® has seen a rise in the number of associated occurrences of hypersensitivity reactions (HSRs). These reactions (also called infusion reactions or IR), can range from harmless symptoms to life-threatening reactions. Current means to prevent IR include the prophylactic use of antihistamines and steroids, but they cannot ensure total prevention. We previously showed that an intravenous injection of doxorubicin-free Doxil-like PEGylated nano-liposomes (Doxebo) prior to Doxil treatment suppresses Doxil-induced complement activation-related pseudoallergy (CARPA) in pigs, a model of human hypersensitivity reactions to Doxil. However, in order to use Doxebo to prevent Doxil-induced IR, we have to prove its safety and that it does not affect Doxil's performance. Here we show that Doxebo itself does not have toxic effects on the host or tumor, and it does not interfere with Doxil's antitumor activity in mice. Blood, microscopic and macroscopic organ evaluation of rats after repeated administration confirm the lack of intrinsic adverse effect of Doxebo. Likewise, the repeated injection of Doxebo before Doxil did not impact Doxil's pharmacokinetics in plasma and therefore does not cause accelerated blood clearance (ABC). Taken together with our previous publications, these data suggest that the injection of Doxebo prior to Doxil administration can help protect against Doxil-induced IR without adversely affecting treatment efficacy and safety.


Subject(s)
Doxorubicin/analogs & derivatives , Drug Hypersensitivity/prevention & control , Liposomes/administration & dosage , Animals , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Female , Humans , Injections, Intravenous , Liposomes/adverse effects , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Rats , Rats, Sprague-Dawley
6.
J Orthop Res ; 37(5): 1192-1201, 2019 05.
Article in English | MEDLINE | ID: mdl-30839125

ABSTRACT

Attempts have been made to visualize tumor cells intraoperatively with fluorescence guidance. However, the clear demarcation and complete tumor resection have always been a challenging task. To address this, we have developed a novel fluorescence bioimaging system with vesicular stomatitis virus (VSV) incorporating Katushka, near-infrared fluorescent protein. VSV is tumor-specific owing to the deficiency of antiviral interferon signaling pathways in tumor cells. We aimed to evaluate the tumor specificity of the recombinant VSV-Katushka (rVSV-K) in osteosarcoma cells and to assess the feasibility of complete tumor resection by the rVSV-K fluorescence guidance. In in vitro experiments, mouse and human osteosarcoma cell lines and normal human mesenchymal stem cells were infected with rVSV-K and observed by fluorescence microscopy. Near-infrared fluorescence was observed only in osteosarcoma cells, even at a low-concentration of virus infections. In in vivo experiments, mouse osteosarcoma (LM8) cells were transplanted subcutaneously into the back of immune-competent mice to produce an osteosarcoma, which was then injected with rVSV-K. The areas emitting fluorescence were resected using a bioimaging system. The distance between the surgical and tumor margins of the fluorescence-guided resection with rVSV-K group was significantly larger than that of the non-guided resection groups. The local recurrence rate was significantly lower in the fluorescence-guided resection with rVSV-K group than in the non-guided resection groups. The distant metastasis rate and average survival rate were not significantly different between all groups. These results suggest that the rVSV-K is specific to osteosarcoma cells and enables complete tumor resection of osteosarcomas in mice. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.


Subject(s)
Fluorescent Dyes , Neoplasms, Experimental/surgery , Osteosarcoma/surgery , Vesiculovirus , Animals , Humans , Male , Mice, Inbred C3H , Neoplasm Recurrence, Local , Neoplasms, Experimental/mortality , Osteosarcoma/mortality
7.
Nat Commun ; 10(1): 1224, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874560

ABSTRACT

Poly-(ADP-ribose) polymerase inhibitors (PARPi) selectively kill breast and ovarian cancers with defects in homologous recombination (HR) caused by BRCA1/2 mutations. There is also clinical evidence for the utility of PARPi in breast and ovarian cancers without BRCA mutations, but the underlying mechanism is not clear. Here, we report that the deubiquitylating enzyme USP15 affects cancer cell response to PARPi by regulating HR. Mechanistically, USP15 is recruited to DNA double-strand breaks (DSBs) by MDC1, which requires the FHA domain of MDC1 and phosphorylated Ser678 of USP15. Subsequently, USP15 deubiquitinates BARD1 BRCT domain, and promotes BARD1-HP1γ interaction, resulting in BRCA1/BARD1 retention at DSBs. USP15 knockout mice exhibit genomic instability in vivo. Furthermore, cancer-associated USP15 mutations, with decreased USP15-BARD1 interaction, increases PARP inhibitor sensitivity in cancer cells. Thus, our results identify a novel regulator of HR, which is a potential biomarker for therapeutic treatment using PARP inhibitors in cancers.


Subject(s)
Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recombinational DNA Repair , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , DNA Breaks, Double-Stranded/drug effects , Drug Resistance, Neoplasm/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplasms/genetics , Neoplasms/mortality , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , RNA, Small Interfering/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Treatment Outcome , Ubiquitin-Specific Proteases/genetics , Whole-Body Irradiation
8.
J Clin Invest ; 129(2): 759-773, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30496141

ABSTRACT

Loss of phosphatase and tensin homolog (PTEN) represents one hallmark of prostate cancer (PCa). However, restoration of PTEN or inhibition of the activated PI3K/AKT pathway has shown limited success, prompting us to identify obligate targets for disease intervention. We hypothesized that PTEN loss might expose cells to unique epigenetic vulnerabilities. Here, we identified a synthetic lethal relationship between PTEN and Brahma-related gene 1 (BRG1), an ATPase subunit of the SWI/SNF chromatin remodeling complex. Higher BRG1 expression in tumors with low PTEN expression was associated with a worse clinical outcome. Genetically engineered mice (GEMs) and organoid assays confirmed that ablation of PTEN sensitized the cells to BRG1 depletion. Mechanistically, PTEN loss stabilized BRG1 protein through the inhibition of the AKT/GSK3ß/FBXW7 axis. Increased BRG1 expression in PTEN-deficient PCa cells led to chromatin remodeling into configurations that drove a protumorigenic transcriptome, causing cells to become further addicted to BRG1. Furthermore, we showed in preclinical models that BRG1 antagonist selectively inhibited the progression of PTEN-deficient prostate tumors. Together, our results highlight the synthetic lethal relationship between PTEN and BRG1 and support targeting BRG1 as an effective approach to the treatment of PTEN-deficient PCa.


Subject(s)
DNA Helicases , Neoplasms, Experimental , Nuclear Proteins , PTEN Phosphohydrolase , Prostatic Neoplasms , Signal Transduction/genetics , Transcription Factors , Animals , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Humans , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PC-3 Cells , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Adv Healthc Mater ; 8(2): e1800995, 2019 01.
Article in English | MEDLINE | ID: mdl-30474227

ABSTRACT

Carbon nanodots (CNDs) with high photothermal conversion efficiency are considered as emerging nanomaterials for advanced biomedical applications attributing to their high biocompatibility, low-cost, and unique photophysical properties. In previous work, supra-CNDs are synthesized exhibiting high absorption in the near-infrared (NIR) region and good NIR photothermal conversion performance. In this work, supra-CNDs are explored as a photothermal agent for photothermal therapy (PTT) and a contrast agent for photoacoustic (PA) imaging, respectively. As a result, in vivo tumor PTT is realized under 655 nm laser irradiation via intratumor injecting supra-CNDs. In vivo PA imaging reveals that supra-CNDs can accumulate in the tumor tissue via the blood circulation after intravenous injection. Moreover, in vivo PTT is conducted after intravenous injection and subsequent tumor accumulation of supra-CNDs, and the lives of mice are prolonged due to the tumor growth inhibition after PTT. These attractive properties indicate that the supra-CNDs can be used as biomedical agents for PA imaging and tumor therapy.


Subject(s)
Nanostructures/chemistry , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Photoacoustic Techniques/methods , Phototherapy/methods , Animals , Body Weight , Carbon/chemistry , Contrast Media/chemistry , Humans , Lasers , Mice, Inbred ICR , Microscopy, Electron, Transmission , Nanostructures/administration & dosage , Nanostructures/therapeutic use , Neoplasms, Experimental/mortality , Temperature , Xenograft Model Antitumor Assays
10.
Eur Rev Med Pharmacol Sci ; 22(24): 8698-8711, 2018 12.
Article in English | MEDLINE | ID: mdl-30575910

ABSTRACT

OBJECTIVE: This study was made to investigate and evaluate the safety and carcinogenicity of nitenpyram (NIT) in rats. MATERIALS AND METHODS: A totally 50 male and 50 female SD rats were treated with NIT at 0, 800, 2400, and 7200 ppm, respectively, for 104 w. The growth, clinical signs, and survival rates, as well as the body and organ weights of these animals, were analyzed. Histopathological examination was also performed. RESULTS: Compared with the control group, survival rates at 104 w were significantly decreased in the 7200 ppm dose group, for both the male and female animals. The occurrence of esophageal squamous papilloma (ESP) was significantly increased in the treated animals. The occurrences of ESP for the 0, 800, 2400, and 7200 ppm NIT treatment groups were 0/39, 0/39, 3/35, and 9/27 for the male animals, and 0/43, 0/43, 6/49, and 12/33 for the female animals, respectively. For pre-neoplastic lesion of ESP, the occurrences of esophageal squamous hyperplasia for the 0, 800, 2400 and 7200 ppm NIT treatment groups were 0/39, 1/39, 10/35, and 9/27 for the male animals, and 0/43, 2/43, 15/49, and 17/33 for the female animals, respectively. The basal cell hyperplasia from mild to severe degrees was observed in the treatment groups. CONCLUSIONS: NIT exhibits carcinogenicity of ESP in the male and female rats after the two-year treatment.


Subject(s)
Carcinogenesis/chemically induced , Esophageal Neoplasms/chemically induced , Insecticides/toxicity , Neonicotinoids/toxicity , Animals , Carcinogenesis/pathology , Carcinogenicity Tests , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagus/drug effects , Esophagus/pathology , Female , Humans , Insecticides/administration & dosage , Male , Neonicotinoids/administration & dosage , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Survival Rate , Time Factors , Toxicity Tests, Chronic
11.
PLoS One ; 13(10): e0204909, 2018.
Article in English | MEDLINE | ID: mdl-30307978

ABSTRACT

RAP1, a component of the telomere-protective shelterin complex, has been shown to have both telomeric and non-telomeric roles. In the liver, RAP1 is involved in the regulation of metabolic transcriptional programs. RAP1-deficient mice develop obesity and hepatic steatosis, these phenotypes being more severe in females than in males. As hepatic steatosis and obesity have been related to increased liver cancer in mice and humans, we set out to address whether RAP1 deficiency resulted in increased liver cancer upon chemical liver carcinogenesis. We found that Rap1-/- females were more susceptible to DEN-induced liver damage and hepatocellular carcinoma (HCC). DEN-treated Rap1-/- female livers showed an earlier onset of both premalignant and malignant liver lesions, which were characterized by increased abundance of γH2AX-positive cells, increased proliferation and shorter telomeres. These findings highlight an important role for RAP1 in protection from liver damage and liver cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , Diethylnitrosamine/adverse effects , Histones/metabolism , Liver Neoplasms/genetics , rap1 GTP-Binding Proteins/genetics , Age of Onset , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Proliferation , Female , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/mortality , Sex Factors , Telomere Shortening
12.
Cancer Immunol Res ; 6(11): 1301-1313, 2018 11.
Article in English | MEDLINE | ID: mdl-30131378

ABSTRACT

Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas have remained elusive. We show that conjugation of a well-documented HPV-E7 SLP to ultra-small polymeric nanoparticles (NP) enhances the antitumor efficacy of therapeutic vaccination in different mouse models of HPV+ cancers. Immunization of TC-1 tumor-bearing mice with a single dose of NP-conjugated E7LP (NP-E7LP) generated a larger pool of E7-specific CD8+ T cells with increased effector functions than unconjugated free E7LP. At the tumor site, NP-E7LP prompted a robust infiltration of CD8+ T cells that was not accompanied by concomitant accumulation of regulatory T cells (Tregs), resulting in a higher CD8+ T-cell to Treg ratio. Consequently, the amplified immune response elicited by the NP-E7LP formulation led to increased regression of large, well-established tumors, resulting in a significant percentage of complete responses that were not achievable by immunizing with the non-NP-conjugated long-peptide. The partial responses were characterized by distinct phases of regression, stable disease, and relapse to progressive growth, establishing a platform to investigate adaptive resistance mechanisms. The efficacy of NP-E7LP could be further improved by therapeutic activation of the costimulatory receptor 4-1BB. This NP-E7LP formulation illustrates a "solid-phase" antigen delivery strategy that is more effective than a conventional free-peptide ("liquid") vaccine, further highlighting the potential of using such formulations for therapeutic vaccination against solid tumors. Cancer Immunol Res; 6(11); 1301-13. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Nanoparticles/chemistry , Papillomavirus E7 Proteins/chemistry , Animals , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/chemistry , Female , Lung Neoplasms/secondary , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Recurrence, Local , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/therapy , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Vaginal Neoplasms/immunology , Vaginal Neoplasms/pathology , Vaginal Neoplasms/prevention & control
13.
Oncogene ; 37(21): 2863-2872, 2018 05.
Article in English | MEDLINE | ID: mdl-29520104

ABSTRACT

The RNA-binding protein Rbm38 is a target of p63 tumor suppressor and can in-turn repress p63 expression via mRNA stability. Thus, Rbm38 and p63 form a negative feedback loop. To investigate the biological significance of the Rbm38-p63 loop in vivo, a cohort of WT, Rbm38-/-, TAp63+/-, and Rbm38-/-;TAp63+/- mice were generated and monitored throughout their lifespan. While mice deficient in Rbm38 or TAp63 alone died mostly from spontaneous tumors, compound Rbm38-/-;TAp63+/- mice had an extended lifespan along with reduced tumor incidence. We also found that loss-of-Rbm38 markedly decreased the percentage of liver steatosis in TAp63+/- mice. Moreover, we found that Rbm38 deficiency extends the lifespan of tumor-free TAp63+/- mice along with reduced expression of senescence-associated biomarkers. Consistent with this, Rbm38-/-;TAp63+/- MEFs were resistant, whereas Rbm38-/- or TAp63+/- MEFs were prone, to cellular senescence. Importantly, we showed that the levels of inflammatory cytokines (IL17D and Tnfsf15) were significantly reduced by Rbm38 deficiency in senescence-resistant Rbm38-/-;TAp63+/- mouse livers and MEFs. Together, our data suggest that Rbm38 and p63 function as intergenic suppressors in aging and tumorigenesis and that the Rbm38-p63 loop may be explored for enhancing longevity and cancer management.


Subject(s)
Aging/genetics , Fatty Liver/genetics , Neoplasms, Experimental/genetics , RNA, Messenger/chemistry , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Feedback, Physiological , Female , Humans , Interleukin-17/metabolism , Male , Mice , Mice, Knockout , Neoplasms, Experimental/mortality , RNA Stability , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15 , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
14.
J Chin Med Assoc ; 81(4): 316-323, 2018 04.
Article in English | MEDLINE | ID: mdl-29246507

ABSTRACT

BACKGROUND: Tumor-induced immunosuppression can impede tumor-specific immune responses and limit the effects of cancer immunotherapy. The aim of this study was to investigate the possible effects of sequential chemoimmunotherapeutic strategies to enhance antitumor immune responses. METHODS: Using the E7-expressing tumor TC-1 as the tumor model, the treatment groups were divided into the following groups: (1) inactivated allogeneic leukocyte infusion (ALI), (2) ALI + MMC-inactivated TC-1 cell vaccine, and (3) ALI + MMC-inactivated TC-1 cell vaccine + cyclophosphamide (CTX). RESULTS: In our study, we demonstrated that treatment with immune-modulating doses of CTX results in a beneficial tumor microenvironment with the suppression of Tregs. ALI has a limited therapeutic effect, as does the MMC-inactivated TC-1 cell vaccine. Our results showed that CTX preconditioning and persistent ALI treatment along with the MMC-inactivated TC-1 cell vaccine resulted in significant inhibition of tumor growth and extended survival. CONCLUSION: Our study illustrated the effects of immune-modulating doses of a sequential chemoimmunotherapeutic strategy targeting the tumor and its microenvironment. The results suggest potential clinical effects for the immunotherapy of HPV-associated malignancies.


Subject(s)
Cyclophosphamide/therapeutic use , Immunotherapy/methods , Leukocyte Transfusion , Neoplasms, Experimental/therapy , Animals , Combined Modality Therapy , Female , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Transplantation, Homologous , Tumor Microenvironment
15.
Int J Oncol ; 50(4): 1221-1232, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259945

ABSTRACT

Urinary bladder cancer is known as a common cancer diagnosed across the world and results in significant mortality and morbidity rates among patients. The retinoblastoma (Rb) protein, as a main tumor suppressor, controls cellular responses to potentially oncogenic stimulation. Rb phosphorylation could disrupt E2F complex formation, resulting in diverse transcription factor dysfunction. In our study, we investigated how Rb is involved in controlling urinary bladder cancer progression. The results indicate that Rb expression is reduced in mice with urinary bladder tumor, and its suppression leads to urinary bladder cancer progression in vivo and in vitro. Rb mutation directly results in tumor size with lower survival rate in vivo. Rb knockdown in vitro promoted bladder tumor cell proliferation, migration and invasion. Interestingly, Rb knockout and knockdown result in autophagy and apoptosis inhibition via suppressing p53 and caspase-3 signaling pathways, enhancing bladder cancer development in vitro and in vivo. These findings reveal that Rb deficiency accelerated urinary bladder cancer progression, exposing an important role of Rb in suppressing urinary bladder cancer for treatment in the future.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Carcinoma/genetics , Neoplasms, Experimental/genetics , Retinoblastoma Protein/genetics , Urinary Bladder Neoplasms/genetics , Animals , Carcinoma/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Knockout , Mutation , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Signal Transduction , Survival Rate , Tumor Suppressor Protein p53/metabolism , Urinary Bladder , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
16.
Planta Med ; 83(1-02): 151-157, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27352383

ABSTRACT

Eight ethanolic extracts of indigenous Taiwanese plants of the genus Alpinia were tested for tumor cytotoxicity against AGS, Hep G2, HeLa, KB, and HL-60 cells. Among the 50 % and 95 % EtOH extracts of eight Alpinia species, the cytotoxic effects of Alpinia intermedia leaves were the strongest. When the leaf extract of A. intermedia was partitioned using n-hexane and aqueous solvents, the n-hexane layer showed a greater cytotoxic effect and could prolong the survival time of P-388D1 tumor-bearing CDF1 mice. Two new labdane diterpene derivatives, intermedin A (1) and intermedin B (2), and coronarin E (3) were isolated from the n-hexane layer of A. intermedia. Intermedin A induced apoptosis in HL-60 cells at 30 µg/mL and significantly prolonged the survival time of P-388D1 tumor-bearing CDF1 mice by 48.7 % at 20 mg/kg of body weight. We suggest that intermedin A is a major compound of A. intermedia and has a cytotoxic effect on HL-60 and P-388D1 cells.


Subject(s)
Alpinia/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , Lignans/pharmacology , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Diterpenes/chemistry , Diterpenes/isolation & purification , Ethanol , Humans , Lignans/chemistry , Lignans/isolation & purification , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology
17.
Oncotarget ; 7(35): 56183-56192, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27487143

ABSTRACT

The transcription factor Zinc finger protein 148 (Zfp148, ZBP-89, BFCOL, BERF1, htß) interacts physically with the tumor suppressor p53, but the significance of this interaction is not known. We recently showed that knockout of Zfp148 in mice leads to ectopic activation of p53 in some tissues and cultured fibroblasts, suggesting that Zfp148 represses p53 activity. Here we hypothesize that targeting Zfp148 would unleash p53 activity and protect against cancer development, and test this idea in the APCMin/+ mouse model of intestinal adenomas. Loss of one copy of Zfp148 markedly reduced tumor numbers and tumor-associated intestinal bleedings, and improved survival. Furthermore, after activation of ß-catenin-the initiating event in colorectal cancer-Zfp148 deficiency activated p53 and induced apoptosis in intestinal explants of APCMin/+ mice. The anti-tumor effect of targeting Zfp148 depended on p53, as Zfp148 deficiency did not affect tumor numbers in APCMin/+ mice lacking one or both copies of Trp53. The results suggest that Zfp148 controls the fate of newly transformed intestinal tumor cells by repressing p53 and that targeting Zfp148 might be useful in the treatment of colorectal cancer.


Subject(s)
Adenoma/pathology , Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , Gastrointestinal Hemorrhage/pathology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Adenoma/mortality , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Colorectal Neoplasms/mortality , DNA-Binding Proteins/genetics , Fibroblasts , Gastrointestinal Hemorrhage/mortality , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , beta Catenin/metabolism
18.
Cancer Res ; 76(20): 5983-5993, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27569209

ABSTRACT

Conventional chemotherapy drugs administered at a maximum tolerated dose (MTD) remains the backbone for treating most cancers. Low-dose metronomic (LDM) chemotherapy, which utilizes lower, less toxic, doses given on a close regular basis over prolonged periods, is an alternative and better tolerated potential strategy to improve chemotherapy. LDM chemotherapy has been evaluated preclinically and clinically and has shown therapeutic benefit, in both early and advanced stage metastatic disease, especially when used as a maintenance therapy. However, knowledge about the antitumor mechanisms by which LDM chemotherapy acts remain limited. Here we characterized the effects of LDM and MTD capecitabine therapy on tumor and host cells using high-throughput systems approaches involving mass spectrometry flow cytometry and automated cell imaging followed by in vivo analyses of such therapies. An increase in myeloid and T regulatory cells and a decrease in NK and T cytotoxic cells were found in MTD-capecitabine-treated tumors compared with LDM-capecitbine-treated tumors. Plasma from MTD capecitabine-treated mice induced a more tumorigenic and metastatic profile in both breast and colon carcinoma cells than plasma from mice treated with LDM capecitabine. These results correlated, in part, with in vivo studies using models of human or mouse advanced metastatic disease, where the therapeutic advantage of MTD capecitabine was limited despite a substantial initial antitumor activity found in the primary tumor setting. Overall these results implicate a possible contribution of immunologic host effects in accounting for the therapeutic limitations of MTD compared with LDM capecitabine. Cancer Res; 76(20); 5983-93. ©2016 AACR.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Capecitabine/therapeutic use , Neoplasms, Experimental/drug therapy , Animals , Bone Marrow Cells/pathology , Cell Line, Tumor , Female , Humans , Maximum Tolerated Dose , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology
19.
Oncotarget ; 7(21): 30781-96, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27096953

ABSTRACT

Malignant gliomas are among the most frequent and aggressive cerebral tumors, characterized by high proliferative and invasive indexes. Standard therapy for patients, after surgery and radiotherapy, consists of temozolomide (TMZ), a methylating agent that blocks tumor cell proliferation. Currently, there are no therapies aimed at reducing tumor cell invasion. Ion channels are candidate molecular targets involved in glioma cell migration and infiltration into the brain parenchyma. In this paper we demonstrate that: i) blockade of the calcium-activated potassium channel KCa3.1 with TRAM-34 has co-adjuvant effects with TMZ, reducing GL261 glioma cell migration, invasion and colony forming activity, increasing apoptosis, and forcing cells to pass the G2/M cell cycle phase, likely through cdc2 de-phosphorylation; ii) KCa3.1 silencing potentiates the inhibitory effect of TMZ on glioma cell viability; iii) the combination of TMZ/TRAM-34 attenuates the toxic effects of glioma conditioned medium on neuronal cultures, through a microglia dependent mechanism since the effect is abolished by clodronate-induced microglia killing; iv) TMZ/TRAM-34 co-treatment increases the number of apoptotic tumor cells, and the mean survival time in a syngeneic mouse glioma model (C57BL6 mice implanted with GL261 cells); v) TMZ/TRAM-34 co-treatment reduces cell viability of GBM cells and cancer stem cells (CSC) freshly isolated from patients.Taken together, these data suggest a new therapeutic approach for malignant glioma, targeting both glioma cell proliferating and migration, and demonstrate that TMZ/TRAM-34 co-treatment affects both glioma cells and infiltrating microglia, resulting in an overall reduction of tumor cell progression.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Pyrazoles/pharmacology , Animals , Apoptosis/drug effects , Brain Neoplasms/mortality , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Synergism , Drug Therapy, Combination , G2 Phase Cell Cycle Checkpoints/drug effects , Glioma/mortality , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/mortality , Neoplastic Stem Cells/drug effects , Phosphorylation , Primary Cell Culture , Pyrazoles/therapeutic use , Temozolomide
20.
J Invest Surg ; 29(6): 366-372, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27027695

ABSTRACT

BACKGROUND: Pancreatic cancer continues to have a poor survival rate with an urgent need for improved treatments. Glaucarubinone, a natural product first isolated from the seeds of the tree Simarouba glauca, has recently been recognized as having anti-cancer properties that may be particularly applicable to pancreatic cancer. METHODS: The effect of glaucarubinone on the growth and migration of murine pancreatic cancer cells was assessed by 3H-thymidine incorporation assay. The survival impact of glaucarubinone alone and in combination with gemcitabine chemotherapy was assessed using an immunocompetent orthotopic murine model of pancreatic cancer. RESULTS: Glaucarubinone inhibited the growth of the murine pancreatic cancer cell lines LM-P and PAN02. Treatment with either glaucarubinone or gemcitabine reduced proliferation in vitro and the combination was synergistic. The combination treatment improved survival two-fold compared to gemcitabine treatment alone (p = 0.046) in PAN02 cells. CONCLUSIONS: The synergistic inhibition by glaucarubinone and gemcitabine observed in vitro and the improved survival in vivo suggest that glaucarubinone may be a useful adjunct to current chemotherapy regimens.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Glaucarubin/analogs & derivatives , Neoplasms, Experimental/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/therapeutic use , Drug Screening Assays, Antitumor , Glaucarubin/pharmacology , Glaucarubin/therapeutic use , Mice , Neoplasms, Experimental/mortality , Pancreatic Neoplasms/mortality , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...