Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 930
Filter
1.
BMC Cancer ; 24(1): 664, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822331

ABSTRACT

Recent studies have shown that blue light-emitting diode (LED) light has anti-tumor effects, suggesting the possibility of using visible light in cancer therapy. However, the effects of blue light irradiation on cells in the tumor microenvironment, including tumor-associated macrophages (TAMs), are unknown. Here, THP-1 cells were cultured in the conditioned medium (CM) of HCT-116 cells to prepare TAMs. TAMs were divided into LED-irradiated and control groups. Then, the effects of blue LED irradiation on TAM activation were examined. Expression levels of M2 macrophage markers CD163 and CD206 expression were significantly decreased in LED-irradiated TAMs compared with the control group. While control TAM-CM could induce HCT-116 cell migration, these effects were not observed in cells cultured in TAM-CM with LED irradiation. Vascular endothelial growth factor (VEGF) secretion was significantly suppressed in LED-exposed TAMs. PD-L1 expression was upregulated in HCT-116 cells cultured with TAM-CM but attenuated in cells cultured with LED-irradiated TAM-CM. In an in vivo model, protein expression levels of F4/80 and CD163, which are TAM markers, were reduced in the LED-exposed group. These results indicate that blue LED light may have an inhibitory effect on TAMs, as well as anti-tumor effects on colon cancer cells.


Subject(s)
Colonic Neoplasms , Light , Tumor-Associated Macrophages , Humans , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Tumor-Associated Macrophages/immunology , Light/adverse effects , Animals , HCT116 Cells , Mice , Tumor Microenvironment/radiation effects , Cell Movement/radiation effects , Culture Media, Conditioned/pharmacology , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Vascular Endothelial Growth Factor A/metabolism , Receptors, Cell Surface/metabolism , Macrophages/metabolism , Macrophages/radiation effects , Macrophages/immunology , Phototherapy/methods , Macrophage Activation/radiation effects , Blue Light
2.
Biomed Pharmacother ; 175: 116668, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701565

ABSTRACT

The combination of radiation treatment and chemotherapy is currently the standard for management of cancer patients. However, safe doses do not often provide effective therapy, then pre-treated patients are forced to repeat treatment with often already increased tumor resistance to drugs and irradiation. One of the solutions we suggest is to improve primary course of radiation treatment via enhancing radiosensitivity of tumors by magnetic-guided iron oxide nanoparticles (magnetite). We obtained spherical heparinized iron oxide nanoparticles (hIONPs, ∼20 nm), characterized it by TEM, Infrared spectroscopy and DLS. Then hIONPs cytotoxicity was assessed for colon cancer cells (XTT assay) and cellular uptake of nanoparticles was analyzed with X-ray fluorescence. Combination of ionizing radiation (IR) and hIONPs in vitro caused an increase of G2/M arrest of cell cycle, mitotic errors and decrease in survival (compared with samples exposed to IR and hIONPs separately). The promising results were shown for magnetic-guided hIONPs in CT26-grafted BALB/C mice: the combination of intravenously administrated hIONPs and IR showed 20,8% T/C ratio (related to non-treated mice), while single radiation had no shown significant decrease in tumor growth (72,4%). Non-guided by magnets hIONPs with IR showed 57,9% of T/C. This indicates that ultra-small size and biocompatible molecule are not the key to successful nano-drug design, in each case, delivery technologies need to be improved when transferred to in vivo model.


Subject(s)
Colonic Neoplasms , Heparin , Magnetic Iron Oxide Nanoparticles , Mice, Inbred BALB C , Radiation-Sensitizing Agents , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , Magnetic Iron Oxide Nanoparticles/chemistry , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/administration & dosage , Humans , Mice , Cell Line, Tumor , Heparin/chemistry , Heparin/pharmacology , Magnetite Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Cell Survival/drug effects
3.
Theranostics ; 14(6): 2573-2588, 2024.
Article in English | MEDLINE | ID: mdl-38646638

ABSTRACT

Background: Hypofractionated radiotherapy (hRT) can induce a T cell-mediated abscopal effect on non-irradiated tumor lesions, especially in combination with immune checkpoint blockade (ICB). However, clinically, this effect is still rare, and ICB-mediated adverse events are common. Lenalidomide (lena) is an anti-angiogenic and immunomodulatory drug used in the treatment of hematologic malignancies. We here investigated in solid tumor models whether lena can enhance the abscopal effect in double combination with hRT. Methods: In two syngeneic bilateral tumor models (B16-CD133 melanoma and MC38 colon carcinoma), the primary tumor was treated with hRT. Lena was given daily for 3 weeks. Besides tumor size and survival, the dependence of the antitumor effects on CD8+ cells, type-I IFN signaling, and T cell costimulation was determined with depleting or blocking antibodies. Tumor-specific CD8+ T cells were quantified, and their differentiation and effector status were characterized by multicolor flow cytometry using MHC-I tetramers and various antibodies. In addition, dendritic cell (DC)-mediated tumor antigen cross-presentation in vitro and directly ex vivo and the composition of tumor-associated vascular endothelial cells were investigated. Results: In both tumor models, the hRT/lena double combination induced a significant abscopal effect. Control of the non-irradiated secondary tumor and survival were considerably better than with the respective monotherapies. The abscopal effect was strongly dependent on CD8+ cells and associated with an increase in tumor-specific CD8+ T cells in the non-irradiated tumor and its draining lymph nodes. Additionally, we found more tumor-specific T cells with a stem-like (TCF1+ TIM3- PD1+) and a transitory (TCF1- TIM3+ CD101- PD1+) exhausted phenotype and more expressing effector molecules such as GzmB, IFNγ, and TNFα. Moreover, in the non-irradiated tumor, hRT/lena treatment also increased DCs cross-presenting a tumor model antigen. Blocking type-I IFN signaling, which is essential for cross-presentation, completely abrogated the abscopal effect. A gene expression analysis of bone marrow-derived DCs revealed that lena augmented the expression of IFN response genes and genes associated with differentiation, maturation (including CD70, CD83, and CD86), migration to lymph nodes, and T cell activation. Flow cytometry confirmed an increase in CD70+ CD83+ CD86+ DCs in both irradiated and abscopal tumors. Moreover, the hRT/lena-induced abscopal effect was diminished when these costimulatory molecules were blocked simultaneously using antibodies. In line with the enhanced infiltration by DCs and tumor-specific CD8+ T cells, including more stem-like cells, hRT/lena also increased tumor-associated high endothelial cells (TA-HECs) in the non-irradiated tumor. Conclusions: We demonstrate that lena can augment the hRT-induced abscopal effect in mouse solid tumor models in a CD8 T cell- and IFN-I-dependent manner, correlating with enhanced anti-tumor CD8 T cell immunity, DC cross-presentation, and TA-HEC numbers. Our findings may be helpful for the planning of clinical trials in (oligo)metastatic patients.


Subject(s)
CD8-Positive T-Lymphocytes , Disease Models, Animal , Lenalidomide , Radiation Dose Hypofractionation , Animals , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor , Combined Modality Therapy/methods , Female , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/therapy
4.
ACS Nano ; 17(24): 25147-25156, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38063344

ABSTRACT

X-ray-induced radiodynamic therapy (RDT) that can significantly reduce radiation dose with an improved anticancer effect has emerged as an attractive and promising therapeutic modality for tumors. However, it is highly significant to develop safe and efficient radiosensitizing agents for tumor radiation therapy. Herein, we present a smart nanotheranostic system FA-Au-CH that consists of gold nanoradiosensitizers, photosensitizer chlorin e6 (Ce6), and folic acid (FA) as a folate-receptor-targeting ligand for improved tumor specificity. FA-Au-CH nanoparticles have been demonstrated to be able to simultaneously serve as radiosensitizers and RDT agents for enhanced computed tomography (CT) imaging-guided radiotherapy (RT) of colon carcinoma, owing to the strong X-ray attenuation capability of high-Z elements Au and Hf, as well as the characteristics of Hf that can transfer radiation energy to Ce6 to generate ROS from Ce6 under X-ray irradiation. The integration of RT and RDT in this study demonstrates great efficacy and offers a promising therapeutic modality for the treatment of malignant tumors.


Subject(s)
Carcinoma , Colonic Neoplasms , Photochemotherapy , Porphyrins , Radiation-Sensitizing Agents , Humans , Porphyrins/therapeutic use , Hafnium , Gold , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Cell Line, Tumor
5.
Int J Mol Sci ; 24(22)2023 Nov 09.
Article in English | MEDLINE | ID: mdl-38003335

ABSTRACT

Cathepsins (Caths) are lysosomal proteases that participate in various physiological and pathological processes. Accumulating evidence suggests that caths play a multifaceted role in cancer progression and radiotherapy resistance responses. Their proteolytic activity influences the tumor's response to radiation by affecting oxygenation, nutrient availability, and immune cell infiltration within the tumor microenvironment. Cathepsin-mediated DNA repair mechanisms can promote radioresistance in cancer cells, limiting the efficacy of radiotherapy. Additionally, caths have been associated with the activation of prosurvival signaling pathways, such as PI3K/Akt and NF-κB, which can confer resistance to radiation-induced cell death. However, the effectiveness of radiotherapy can be limited by intrinsic or acquired resistance mechanisms in cancer cells. In this study, the regulation and expression of cathepsin B (cath B) in the colon carcinoma cell line (caco-2) before and after exposure to radiation were investigated. Cells were exposed to escalating ionizing radiation doses (2 Gy, 4 Gy, 6 Gy, 8 Gy, and 10 Gy). Analysis of protein expression, in vitro labeling using activity-based probes DCG04, and cath B pull-down revealed a radiation-induced up-regulation of cathepsin B in a dose-independent manner. Proteolytic inhibition of cathepsin B by cathepsin B specific inhibitor CA074 has increased the cytotoxic effect and cell death due to ionizing irradiation treatment in caco-2 cells. Similar results were also obtained after cathepsin B knockout by CRISPR CAS9. Furthermore, upon exposure to radiation treatment, the inhibition of cath B led to a significant upregulation in the expression of the proapoptotic protein BAX, while it induced a significant reduction in the expression of the antiapoptotic protein BCL-2. These results showed that cathepsin B could contribute to ionizing radiation resistance, and the abolishment of cathepsin B, either by inhibition of its proteolytic activity or expression, has increased the caco-2 cells susceptibility to ionizing irradiation.


Subject(s)
Carcinoma , Colonic Neoplasms , Humans , Apoptosis , Caco-2 Cells , Cathepsin B/metabolism , Cell Line, Tumor , Colonic Neoplasms/radiotherapy , Phosphatidylinositol 3-Kinases , Radiation, Ionizing , Tumor Microenvironment
6.
Biomater Sci ; 11(18): 6177-6192, 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37504889

ABSTRACT

Radiation therapy (RT) is a mainstream clinical approach in cancer treatment. However, the therapeutic efficacy of RT is greatly hindered by the presence of excessive hydrogen peroxide (H2O2) in the hypoxic region of the solid tumor, thus leading to tumor recurrence and metastasis. Herein, a thioketal-linked amphiphilic nano-assembly (MTS) loaded with hydrophobic manganese oxide (HMO) nanoparticles (MTS@HMO) is examined as a promising multi-purpose reactive oxygen species (ROS)-catalytic nanozyme for transforming an RT-resistant hypoxic tumor microenvironment (TME) into an RT-susceptible one by scavenging ROS in the hypoxic core of the solid tumor. After intravenous injection, the MTS@HMO nano-assembly was able to sense and be degraded by the abundant ROS in the hypoxic TME, thereby releasing HMO particles for subsequent scavenging of H2O2. The oxygen generated during peroxide scavenging then relieved the hypoxic TME, thereby resulting in an increased sensitivity of the hypoxic tumor tissue towards RT. Moreover, the in situ hypoxic status was monitored via the T1-enhanced magnetic resonance (MR) imaging of the Mn2+ ions generated by the ROS-mediated degradation of HMO. The in vitro results demonstrated a significant H2O2 elimination and enhanced oxygen generation after the treatment of the MTS@HMO nano-assembly with tumor cells under hypoxic conditions, compared to the control MTS group. In addition, the combination of RT and pre-treatment with MTS@HMO nano-assembly significantly amplified the permanent DNA strand breaks in tumor cells compared to the control RT group. More importantly, the in vivo results proved that the systemic injection of the MTS@HMO nano-assembly prior to RT irradiation enhanced the RT-mediated tumor suppression and down-regulated the hypoxic marker of HIF-1α in the solid tumor compared to the control RT group. Overall, the present work demonstrates the great potential of the versatile ROS-catalytic hypoxia modulating strategy using the MTS@HMO nano-assembly to enhance the RT-induced antitumor efficacy in hypoxic solid tumors.


Subject(s)
Colonic Neoplasms , Photochemotherapy , Humans , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/chemistry , Cell Line, Tumor , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/pathology , Oxygen/metabolism , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/drug therapy , Tumor Microenvironment , Photochemotherapy/methods
7.
Cancer Gene Ther ; 30(10): 1369-1381, 2023 10.
Article in English | MEDLINE | ID: mdl-37495855

ABSTRACT

Radiotherapy is one of the most common cancer treatments, yet, some patients require high doses to respond. Therefore, the development of new strategies leans toward personalizing therapy to avoid unnecessary burden on cancer patients. This approach prevents the administration of ineffective treatments or uses combination strategies to increase the sensitivity of cancer cells. ADAM12 has been shown to be upregulated in many cancers and correlate with poor survival and chemoresistance, thus making it a potential candidate responsible for radioresistance. Here, we show that ADAM12 expression is upregulated in response to irradiation in both mouse and human cancer cells in vitro, as well as in tumor tissues from rectal cancer patients. Interestingly, the expression of ADAM12 following radiotherapy correlates with the initial disease stage and predicts the response of rectal cancer patients to the treatment. While we found no cell-autonomous effects of ADAM12 on the response of colon cancer cells to irradiation in vitro, depletion of ADAM12 expression markedly reduced the tumor growth of irradiated cancer cells when subcutaneously transplanted in syngeneic mice. Interestingly, loss of cancer cell-derived ADAM12 expression increased the number of CD31+FAP- cells in murine tumors. Moreover, conditioned medium from ADAM12-/- colon cancer cells led to increased tube formation when added to endothelial cell cultures. Thus, it is tempting to speculate that altered tumor vascularity may be implicated in the observed effect of ADAM12 on response to radiotherapy in rectal cancer. We conclude that ADAM12 represents a promising prognostic factor for stratification of rectal cancer patients receiving radiotherapy and suggest that targeting ADAM12 in combination with radiotherapy could potentially improve the treatment response.


Subject(s)
Colonic Neoplasms , Rectal Neoplasms , Animals , Humans , Mice , ADAM12 Protein/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/radiotherapy , Gene Expression Regulation, Neoplastic , Prognosis , Rectal Neoplasms/genetics , Rectal Neoplasms/radiotherapy
8.
Int J Radiat Biol ; 99(5): 779-790, 2023.
Article in English | MEDLINE | ID: mdl-36731457

ABSTRACT

BACKGROUND: Probiotics such as Lactobacillus could modulate the intestinal microbiota and have been considered as an effective strategy for ameliorating colon carcinoma. Nevertheless, its efficiency remains the biggest challenge. METHODS: We investigated the therapeutic efficacy of Lactobacillus reuteri JMR-01 adjuvant 12C6+ irradiation on CT-26 syngeneic mouse models. Meanwhile, intestinal flora and innate immunity were examined to outline mechanisms. RESULTS: Anti-proliferation effect of live probiotic combined with inactivated probiotic JMR-01 (LP + IP) on CT-26 reached a maximum of 39.55% among other experiment groups at 24 h when the ratio of cell to CFU was 1:1 in vitro. These activities have been fully validated in vivo, tumor-bearing mice treated by 12C6+ irradiation combining with living and inactivated probiotics JMR-01 (IR + LP + IP) for 50-day held the highest survival rate (71.4%) and complete remission rate (14.3%). We also demonstrated significant fluctuation in gut microbiota, including the decreased abundance of Bacteroides fragilis and Clostridium perfringens related to tumorigenesis and development, and the increased abundance of Lactobacillus and Bifidobacterium closely associated with health restoration in fecal of mice treated with JMR-01 LP + IP adjuvant 12C6+ irradiation (IR + LP + IP). Similarly, the decreasing nitroreductase activities and increasing short chain fatty acids (SCFAs) concentrations were observed in IR + LP + IP group compared with tumor control group, which further confirmed the changes of gut microbiota. Additionally, we found that the strongest stimulation index of splenocyte (2.47) and the phagocytosis index peritoneal macrophage (3.68) were achieved by LP + IP compared with single live JMR-01 (LP) and inactivated JMR-01 (IP). CONCLUSIONS: JMR-01 LP + IP adjuvant 12C6+ irradiation could mitigate cancer progression by modulating innate immunity as well as intestinal flora.


Subject(s)
Carcinoma , Colonic Neoplasms , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Animals , Mice , Lactobacillus , Colonic Neoplasms/radiotherapy
9.
Lasers Med Sci ; 38(1): 37, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36627516

ABSTRACT

Early cancer diagnosis through characterizing light propagation and nanotechnology increases the survival rate. The present research is aimed at evaluating the consequence of using natural nanoparticles in cancer therapy and diagnosis. Colon cancer cells were differentiated from the normal cells via investigating light diffusion combined with the fluorescence effect of the Ashwagandha chitosan nanoparticles (Ash C NPs). Ionic gelation technique synthesized the Ash C NPs. High-resolution transmission electron microscope, dynamic light scattering, and zeta potential characterized Ash C NPs. Fourier transform infrared spectroscopy analyzed Ash C NPs, chitosan, and Ashwagandha root water extract. Moreover, the MTT assay evaluated the cytotoxicity of Ash C NPs under the action of near-infrared light (NIR) irradiation. The MTT assay outcomes were statistically analyzed by Bonferroni post hoc multiple two-group comparisons using one-way variance analysis (ANOVA). Based on the Monte-Carlo simulation technique, the spatially resolved steady-state diffusely reflected light from the cancerous and healthy cells is acquired. The diffuse equation reconstructed the optical fluence rate using the finite element technique. The fluorescent effect of the nanoparticles was observed when the cells were irradiated with NIR. The MTT assay revealed a decrease in the cell viability under the action of Ash C NPs with and without laser irradiation. Colon cancer and normal cells were differentiated based on the optical characterization after laser irradiation. The light diffusion equation was successfully resolved for the fluence rate on cells' surfaces showing different normal and cancer cells values. Ash C NPs appeared its fluorescent effect in the presence of NIR laser.


Subject(s)
Chitosan , Colonic Neoplasms , Nanoparticles , Humans , Plant Extracts , Coloring Agents , Nanoparticles/chemistry , Colonic Neoplasms/radiotherapy , Spectroscopy, Fourier Transform Infrared
10.
BMC Cancer ; 23(1): 62, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36653774

ABSTRACT

BACKGROUND: Indoleamine 2,3-dioxygenase 1 (IDO1) is an enzyme that converts tryptophan to kynurenine. IDO1 expression is found not only in tumor cells but also in immune cells and is associated with tumor proliferation and immune responses. IDO1 inhibitors and radiation may cooperatively suppress tumor proliferation through the alterations in the Wnt/ß-catenin pathway, cell cycle, and immune response. We investigated the antitumor effects of combination therapy of an IDO1 inhibitor, 1-methyl tryptophan (1-MT), and radiation on colorectal cancer. METHODS: In vitro experiments were conducted using human and murine colon cancer cell lines (HCT116, HT-29, and Colon26). Cell growth inhibition was assessed using a MTS assay and Clonogenic assay. Cells were cultured for 48 h with or without 500 µM 1-MT after exposure to radiation (4 Gy). Cell cycle effects and modulation of Wnt/ß-catenin pathway were evaluated using western blot analysis, flow cytometry, RT-PCR. Subcutaneous Colon26 tumors in BALB/c mice were treated by oral 1-MT (6 mg/mL) for 2 weeks and/or local radiation (10 Gy/10 fr). Bromodeoxyuridine (BrdU) incorporation in tumor cells and expression of differentiation markers of immune cells were evaluated using immunohistochemistry. RESULTS: 1-MT and a small interfering RNA against IDO1 suppressed proliferation of all cell lines, which was rescued by kynurenine. Clonogenic assay showed that administration of 1-MT improved radiosensitivity by suppressing the Wnt/ß-catenin pathway activated by radiation and enhancing cell cycle arrest induced by radiation. Combination therapy showed a further reduction in tumor burden compared with monotherapies or untreated control, inducing the highest numbers of intratumoral CD3 + and CD8 + T cells and the lowest numbers of Foxp3 + and BrdU-positive tumor cells. CONCLUSIONS: The combination of 1-MT and radiation suppressed colon cancer cells in vitro and in vivo via multiple mechanisms.


Subject(s)
Colonic Neoplasms , Kynurenine , Humans , Mice , Animals , Kynurenine/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , beta Catenin , Bromodeoxyuridine , Mice, Inbred C57BL , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , HT29 Cells
11.
J Immunol Res ; 2022: 1328542, 2022.
Article in English | MEDLINE | ID: mdl-35935580

ABSTRACT

Colon cancer ranks third worldwide, and it has a growing incidence with urbanization and industrialization. Drug resistance in colon cancer is gradually affecting the treatment. This study focused on the mechanisms by which acriflavine (ACF) enhances the radiosensitivity of colon cancer cells. First, the expression and activation levels of tumor suppressor protein p53 were shown high in normal cells and tissues in its detection, which suggests that p53 is likely to be a key factor in colon cancer. Then, the expression of p53 ended up increasing in ACF group after SW620 cells were cultured with ACF. In addition, ACF group had some other changes. The expression of mitochondrial related antiapoptotic protein Bcl-2 increased, while the expression of proapoptotic protein Bax, Bad, cytopigment C, and apoptotic inducer AIF decreased. At the same time, the ability of apoptosis was enhanced, and the ability of proliferation and invasion was decreased. This suggests that ACF can promote p53 expression and affect mitochondrial function and the radiosensitivity of SW620. The luciferase reporting experiment showed that there was a binding site between ACF and p53. Besides, when IR treatment was applied to SW620 with high p53 expression, there was an increase in the expression of Bcl-2 in SW620 and decrease in Bax, Bad, and cytopigment C in AIF. Meanwhile, the cell apoptosis became stronger, and the proliferation and invasion became weaker. The experimental results were similar to those of SW620 cells cultured with ACF, suggesting that p53 is an intermediate factor in the regulation of SW620 by ACF. Finally, in this study, cells were cultured with ACF, and p53 was knocked down at the same time. The experimental results showed that after p53 was knocked down. ACF's ability to regulate SW620 is partially removed. This confirms the view that ACF regulates SW620 cells by regulating p53. In summary, this study found the mechanism by which ACF causes mitochondrial dysfunction and improves the radiosensitivity of colon cancer cells by activating the tumor suppressor protein p53, which may contribute to solving the drug resistance in colon cancer.


Subject(s)
Colonic Neoplasms , Tumor Suppressor Protein p53 , Acriflavine/metabolism , Acriflavine/pharmacology , Acriflavine/therapeutic use , Apoptosis Regulatory Proteins , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Tolerance , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
12.
J Photochem Photobiol B ; 234: 112527, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35914464

ABSTRACT

In recent decades, the laser treatment of cancer has been introduced as a promising treatment option. Because of the maldistribution of optical energy and an ambiguous boundary between the normal and tumor tissues, laser irradiation can stimulate residual cancer cells, leading to a cancer regrowth. As photobiomodulation (PBM) is involved in an extensive range of cellular responses, profound comprehension of photo-stimulated mechanisms against the cancer cells is required to establish a safety margin for PBM. Therefore, we aimed to identify the stimulant effects of PBM at various wavelengths against the tumor cells to establish a safety margin for the laser treatment. CT26 murine colon cancer cells were exposed to either 405 (BL), 635 (VIS), or 808 (NIR) nm laser lights at the fluences of 0, 10, 30, and 50 J/cm2. In addition, CT26 tumor-bearing mice were irradiated with BL, VIS, or NIR at a fluence of 30 J/cm2. Both the proliferation and angiogenesis potential of the CT26 cells and tumors were evaluated using the MTT assay, western blot, and immunohistochemistry (IHC) staining analyses. Although cell viability was not statistically significant, BL significantly induced p-ERK upregulation in the CT26 cells, indicating that PBM with BL can stimulate proliferation. In vivo tests showed that the NIR group exhibited the maximum relative tumor volume, and BL yielded a slight increase compared to the control. In the IHC staining and western blot analyses, both BL and NIR increased the expression of EGFR, VEGF, MMP-9, and HIF-1α, which are related to the proliferation and angiogenesis-related factors. Further investigations will be pursued to clarify the molecular pathways that depend on the cancer cell types and laser wavelengths for the establishment of safety guidelines in clinical environments.


Subject(s)
Colonic Neoplasms , Low-Level Light Therapy , Animals , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Colonic Neoplasms/radiotherapy , Light , Mice
13.
Nano Lett ; 22(15): 6409-6417, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35867897

ABSTRACT

The development of a manageable reactive nitrogen species-potentiated nitrosative stress induction system for cancer therapy has remained elusive. Herein, tailored silica-based nanoscintillators were reported for low-dosage X-ray boosting for the in situ formation of highly cytotoxic peroxynitrite (ONOO-). Significantly, cellular nitrosative stress revolving around the intracellular protein tyrosine nitration through ONOO- pathways was explored. High-energy X-rays were directly deposited on silica-based nanoscintillators, forming the concept of an open source and a reduced expenditure-aggravated DNA damage strategy. Moreover, the resultant ONOO-, along with the released nitric oxide, not only can act as "oxygen suppliers" to combat tumor hypoxia but also can induce mitochondrial damage to initiate caspase-mediated apoptosis, further improving the therapeutic efficacy of radiotherapy. Thus, the design of advanced nanoscintillators with specific enhanced nitrosative stress offers promising potential for postoperative radiotherapy of colon cancer.


Subject(s)
Colonic Neoplasms , Peroxynitrous Acid , Colonic Neoplasms/radiotherapy , Humans , Nitric Oxide/metabolism , Nitrosative Stress , Peroxynitrous Acid/metabolism , Reactive Nitrogen Species/metabolism , Silicon Dioxide
14.
Chem Commun (Camb) ; 58(49): 6942-6945, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35640157

ABSTRACT

Introducing 2,3-dimethyl-1H-imidazol-3-ium iodide (Dmim) as a monodentate ligand during the preparation of ZIF-8 yields ZIF-8 + (50) and ZIF-8 + (38) with cationic 'missing linker' defects. ZIF-8 + (38) adsorbs 125I2 and the resulting radioactive host-guest complex exhibits in vitro cytotoxicity comparable to that of Na125I against colon cancer cell line CT26.


Subject(s)
Colonic Neoplasms , Zeolites , Cations , Colonic Neoplasms/radiotherapy , Humans , Iodine Radioisotopes , Zeolites/pharmacology
15.
Small ; 18(14): e2107656, 2022 04.
Article in English | MEDLINE | ID: mdl-35150039

ABSTRACT

Even though radiotherapy is the most important therapeutic strategy for colon cancer treatment, there is an enormous demand to improve radiosensitivity in solid tumor destruction. For this purpose, a biomimetic nanoplatform based on hollow polydopamine nanoparticles (HP) with homologous targeting and pH-responsive drug release properties is designed. In this work, HP is constructed by using a chelation competition-induced polymerization strategy and then modified with the cancer cell membrane. Hollow polydopamine integrated with Pt nanoparticles (Pt@HP) has a catalase-like activity, which can be used to trigger endogenous H2 O2 into O2 , relieving hypoxia of the tumor microenvironment (TME). With mesoporous shells and large cavities, Pt@HP shows efficient apoptin100-109 (AP) and verteporfin (VP) loading to form AVPt@HP@M. Under X-ray irradiation, AVPt@HP@M exerts a radiosensitization effect via multiple strategies, including relieving hypoxia (Pt NPs), enhancing tumor apoptosis (AP), and X-ray-induced photodynamic therapy (X-PDT) (VP). Further metabonomics analysis shows that the specific mechanism of the AVPt@HP@M is through influencing purine metabolism. Without appreciable systemic toxicity, this nanoplatform highlights a new strategy for effective radiosensitization and provides a reference for treating malignant tumors.


Subject(s)
Colonic Neoplasms , Nanoparticles , Photochemotherapy , Biomimetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Humans , Hypoxia , Indoles , Nanoparticles/therapeutic use , Polymers , Tumor Microenvironment
16.
Med Oncol ; 39(2): 18, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34982268

ABSTRACT

Although radiotherapy is an effective strategy for cancer treatment, tumor resistance to ionizing radiation (IR) and its toxic effects on normal tissues are limiting its use. The aim of this study is to evaluate the anti-cancer effects of mefenamic acid (MEF), as an approved medicine, and its combination with IR against colon tumor cells in mice. Tumor-bearing mice were received MEF at a dose of 25 mg/kg for 6 successive days. The tumor size was measured. In the second experiment, after MEF treatment, tumor-bearing mice locally received an X-ray at dose 6 Gy. Tumor growth and biochemical, histological, and immunohistological assay (caspase-3) were performed. MEF significantly decreased tumor size in mice in comparison to the control group. IR and/or MEF treatment significantly reduced the tumor volume and inhibited tumor growth by 49%, 55%, and 67% by MEF, IR, and MEF + IR groups as compared with the control group. Administration of MEF in combination with radiation had a synergistic effect on enhanced histopathological changes in tumor tissues. MEF treatment in IR exposure mice showed a significant increase in the immunoreactivity of caspase-3 in the colon tumor tissue. MEF has an anti-tumor effect in colon tumor-bearing mice. MEF in combination with IR increased pathological changes and apoptosis in tumor tissues, suggesting that MEF might be clinically useful in the treatment of colon cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemoradiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Mefenamic Acid/therapeutic use , Animals , Caspase 3/metabolism , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Humans , Immunohistochemistry , Male , Mice, Nude , Tumor Burden
17.
Nucleic Acids Res ; 49(19): 11067-11082, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34606602

ABSTRACT

KRAS-activating mutations are oncogenic drivers and are correlated with radioresistance of multiple cancers, including colorectal cancer, but the underlying precise molecular mechanisms remain elusive. Herein we model the radiosensitivity of isogenic HCT116 and SW48 colorectal cancer cell lines bearing wild-type or various mutant KRAS isoforms. We demonstrate that KRAS mutations indeed lead to radioresistance accompanied by reduced radiotherapy-induced mitotic catastrophe and an accelerated release from G2/M arrest. Moreover, KRAS mutations result in increased DNA damage response and upregulation of 53BP1 with associated increased non-homologous end-joining (NHEJ) repair. Remarkably, KRAS mutations lead to activation of NRF2 antioxidant signaling to increase 53BP1 gene transcription. Furthermore, genetic silencing or pharmacological inhibition of KRAS, NRF2 or 53BP1 attenuates KRAS mutation-induced radioresistance, especially in G1 phase cells. These findings reveal an important role for a KRAS-induced NRF2-53BP1 axis in the DNA repair and survival of KRAS-mutant tumor cells after radiotherapy, and indicate that targeting NRF2, 53BP1 or NHEJ may represent novel strategies to selectively abrogate KRAS mutation-mediated radioresistance.


Subject(s)
Colonic Neoplasms/genetics , DNA End-Joining Repair , NF-E2-Related Factor 2/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Radiation Tolerance/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , DNA Breaks, Double-Stranded , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , G1 Phase Cell Cycle Checkpoints/radiation effects , G2 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/radiation effects , Gamma Rays , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mutation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Tumor Suppressor p53-Binding Protein 1/antagonists & inhibitors , Tumor Suppressor p53-Binding Protein 1/metabolism
18.
Br J Radiol ; 94(1128): 20210593, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34520668

ABSTRACT

OBJECTIVE: The aim of the present study was to evaluate the local and regional therapeutic efficacy and abscopal effect of BNCT mediated by boronophenyl-alanine, combined with Bacillus Calmette-Guerin (BCG) as an immunotherapy agent in this model. METHODS: The local effect of treatment was evaluated in terms of tumor response in the irradiated tumor-bearing right hind flank. Metastatic spread to tumor-draining lymph nodes was analyzed as an indicator of regional effect. The abscopal effect of treatment was assessed as tumor growth inhibition in the contralateral (non-irradiated) left hind flank inoculated with tumor cells 2 weeks post-irradiation. The experimental groups BNCT, BNCT + BCG, BCG, Beam only (BO), BO +BCG, SHAM (tumor-bearing, no treatment, same manipulation) were studied. RESULTS: BNCT and BNCT + BCG induced a highly significant local anti-tumor response, whereas BCG alone induced a weak local effect. BCG and BNCT + BCG induced a significant abscopal effect in the contralateral non-irradiated leg. The BNCT + BCG group showed significantly less metastatic spread to tumor-draining lymph nodes vs SHAM and vs BO. CONCLUSION: This study suggests that BNCT + BCG-immunotherapy would induce local, regional and abscopal effects in tumor-bearing animals. BNCT would be the main effector of the local anti-tumor effect whereas BCG would be the main effector of the abscopal effect. ADVANCES IN KNOWLEDGE: Although the local effect of BNCT has been widely evidenced, this is the first study to show the local, regional and abscopal effects of BNCT combined with immunotherapy, contributing to comprehensive cancer treatment with combined therapies.


Subject(s)
Boron Neutron Capture Therapy/methods , Colonic Neoplasms/therapy , Immunotherapy/methods , Animals , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Combined Modality Therapy/methods , Disease Models, Animal , Female , Male , Rats , Treatment Outcome
19.
Cell Death Dis ; 12(7): 694, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257274

ABSTRACT

Hypoxia, a characteristic of most human solid tumors, is a major obstacle to successful radiotherapy. While moderate acute hypoxia increases cell survival, chronic cycling hypoxia triggers adaptation processes, leading to the clonal selection of hypoxia-tolerant, apoptosis-resistant cancer cells. Our results demonstrate that exposure to acute and adaptation to chronic cycling hypoxia alters the balance of Bcl-2 family proteins in favor of anti-apoptotic family members, thereby elevating the apoptotic threshold and attenuating the success of radiotherapy. Of note, inhibition of Bcl-2 and Bcl-xL by BH3-mimetic ABT-263 enhanced the sensitivity of HCT116 colon cancer and NCI-H460 lung cancer cells to the cytotoxic action of ionizing radiation. Importantly, we observed this effect not only in normoxia, but also in severe hypoxia to a similar or even higher extent. ABT-263 furthermore enhanced the response of xenograft tumors of control and hypoxia-selected NCI-H460 cells to radiotherapy, thereby confirming the beneficial effect of combined treatment in vivo. Targeting the Bcl-2 rheostat with ABT-263, therefore, is a particularly promising approach to overcome radioresistance of cancer cells exposed to acute or chronic hypoxia with intermittent reoxygenation. Moreover, we found intrinsic as well as ABT-263- and irradiation-induced regulation of Bcl-2 family members to determine therapy sensitivity. In this context, we identified Mcl-1 as a resistance factor that interfered with apoptosis induction by ABT-263, ionizing radiation, and combinatorial treatment. Collectively, our findings provide novel insights into the molecular determinants of hypoxia-mediated resistance to apoptosis and radiotherapy and a rationale for future therapies of hypoxic and hypoxia-selected tumor cell fractions.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis , Colonic Neoplasms/radiotherapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrolides/metabolism , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Burden/drug effects , Tumor Burden/radiation effects , Tumor Hypoxia , Tumor Microenvironment , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
20.
Cancer Biomark ; 32(2): 199-206, 2021.
Article in English | MEDLINE | ID: mdl-34092618

ABSTRACT

PURPOSE: Insulin like growth factor receptor 1 (IGF-1R) has been documented to play a key role in radiation response, thereby offering an attractive drug target to enhance tumor sensitivity to radiotherapy. Here, we investigated wether knockdown of IGF-1R can sensitize colorectal cancer (CRC) cell lines to radiation. MATERIAL AND METHODS: Human colon carcinoma SW480 and HT-29 cells were transfected with specific small interference RNA (siRNA) to mediate IGF-1R depletion. The expression of IGF-1R mRNA and protein among transfected and untransfected cells was detected by Western blot analysis. Changes in cell proliferation and radiosensitivity were evaluated by the clonogenic survival assay. NVP-ADW742, an IGF-1R inhibitor, in combination with radiation was studied. RAD51, a measure for homologous recombination repair, and 53BP1, a maker for non-homologous end-joining (NHEJ), were determined by immunofluorescence for double-strand breaks (DSB) repair pathways. Cell cycle was also examined in the IGF-1R knockdown and IGF-1R-inhibited cells. RESULTS: CRC cell lines were selectively sensitized to radiation after siRNA-mediated IGF-1R depletion. NVP-ADW742 efficiently increases cancer cell response to radiation. Furthermore, initial formation of RAD51 foci after IR, and 53BP1 foci were significantly reduced in IGF-1R-depleted or with IGF-1R Inhibitor CRC cell lines. Lastly, IGF-1R-depleted or with IGF-1R Inhibitor caused more G2 phase cell arrest. CONCLUSION: Our findings demonstrate that depletion of IGF-1R lead to an increase in radiosensitivity in CRC.


Subject(s)
Colonic Neoplasms/radiotherapy , Radiation Tolerance/genetics , Receptor, IGF Type 1/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Humans , Radiation Tolerance/drug effects , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...