Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.185
Filter
1.
J Cancer Res Ther ; 20(2): 695-705, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38687942

ABSTRACT

BACKGROUND: Tumor-associated macrophages (TAMs) are intimately involved in cancer radiochemotherapy resistance. However, the mechanism by which macrophages affect radiosensitivity through autophagy remains unclear. The purpose of our study was to investigate how activating autophagy in type-II macrophages (M2) by using rapamycin (RAP) would affect the radiosensitivity of colorectal cancer (CRC) xenografts. MATERIALS AND METHODS: A nude mouse CRC model was established by injecting LoVo CRC cells. After tumor formation, supernatant from M2 cells (autophagy-unactivated), autophagy-activated M2 cells, or autophagy-downregulated M2 cells was injected peritumorally. All tumor-bearing mice were irradiated with 8-Gy X-rays twice, and the radiosensitivity of CRC xenografts was analyzed in each group. RESULTS: The mass, volume, and microvessel density (MVD) of tumors in the autophagy-unactivated M2 group significantly increased; however, supernatant from M2 cells that were autophagy-activated by rapamycin significantly decreased tumor weight, volume, and MVD compared with negative control. Combining bafilomycin A1 (BAF-A1) with RAP treatment restored the ability of the M2 supernatant to increase tumor mass, volume, and MVD. Immunohistochemical and Western blot results showed that compared with the negative control group, supernatant from M2 cells that were not activated by autophagy downregulated the expression of Livin and Survivin in tumor tissues; activation of M2 autophagy further downregulated the protein levels. CONCLUSIONS: Therefore, autophagy-activated M2 supernatant can downregulate the expression of the antiapoptotic genes Livin and Survivin in CRC xenografts, improving the radiosensitivity of CRC by inducing apoptosis in combination with radiotherapy and inhibiting the growth of transplanted tumors.


Subject(s)
Autophagy , Colorectal Neoplasms , Mice, Nude , Radiation Tolerance , Sirolimus , Xenograft Model Antitumor Assays , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/therapy , Colorectal Neoplasms/radiotherapy , Colorectal Neoplasms/metabolism , Mice , Autophagy/drug effects , Autophagy/radiation effects , Humans , Radiation Tolerance/drug effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , Cell Line, Tumor , Apoptosis/drug effects , Apoptosis/radiation effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Survivin/metabolism , Survivin/genetics , Mice, Inbred BALB C , Male
2.
Adv Sci (Weinh) ; 11(18): e2400845, 2024 May.
Article in English | MEDLINE | ID: mdl-38520732

ABSTRACT

Complete remission of colorectal cancer (CRC) is still unachievable in the majority of patients by common fractionated radiotherapy, leaving risks of tumor metastasis and recurrence. Herein, clinical CRC samples demonstrated a difference in the phosphorylation of translation initiation factor eIF2α (p-eIF2α) and the activating transcription factor 4 (ATF4), whose increased expression by initial X-ray irradiation led to the resistance to subsequent radiotherapy. The underlying mechanism is studied in radio-resistant CT26 cells, revealing that the incomplete mitochondrial outer membrane permeabilization (iMOMP) triggered by X-ray irradiation is key for the elevated expression of p-eIF2α and ATF4, and therefore radio-resistance. This finding guided to discover that metformin and 2-DG are synergistic in reversing radio resistance by inhibiting p-eIF2α and ATF4. Liposomes loaded with metformin and 2-DG (M/D-Lipo) are thus prepared for enhancing fractionated radiotherapy of CRC, which achieved satisfactory therapeutic efficacy in both local and metastatic CRC tumors by reversing radio-resistance and preventing T lymphocyte exhaustion.


Subject(s)
Colorectal Neoplasms , Liposomes , Metformin , Mitochondria , Radiation Tolerance , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/radiotherapy , Colorectal Neoplasms/pathology , Mice , Animals , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/radiation effects , Metformin/pharmacology , Metformin/therapeutic use , Radiation Tolerance/drug effects , Cell Line, Tumor , Disease Models, Animal , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics
3.
Radiother Oncol ; 194: 110194, 2024 May.
Article in English | MEDLINE | ID: mdl-38447871

ABSTRACT

High precision, image-guided radiotherapy (RT) has increased the therapeutic ratio, enabling higher tumor and lower normal tissue doses, leading to improved patient outcomes. Nevertheless, some patients remain at risk of developing serious side effects.In many clinical situations, the radiation tolerance of normal tissues close to the target volume limits the dose that can safely be delivered and thus the potential for tumor control and cure. This is particularly so in patients being re-treated for tumor progression or a second primary tumor within a previous irradiated volume, scenarios that are becoming more frequent in clinical practice.Various normal tissue 'radioprotective' drugs with the potential to reduce side effects have been studied previously. Unfortunately, most have failed to impact clinical practice because of lack of therapeutic efficacy, concern about concurrent tumor protection or excessive drug-related toxicity. This review highlights the evidence indicating that targeting the CXCL12/CXCR4 pathway can mitigate acute and late RT-induced injury and reduce treatment side effects in a manner that overcomes these previous translational challenges. Pre-clinical studies involving a broad range of normal tissues commonly affected in clinical practice, including skin, lung, the gastrointestinal tract and brain, have shown that CXCL12 signalling is upregulated by RT and attracts CXCR4-expressing inflammatory cells that exacerbate acute tissue injury and late fibrosis. These studies also provide convincing evidence that inhibition of CXCL12/CXCR4 signalling during or after RT can reduce or prevent RT side effects, warranting further evaluation in clinical studies. Greater dialogue with the pharmaceutical industry is needed to prioritize the development and availability of CXCL12/CXCR4 inhibitors for future RT studies.


Subject(s)
Chemokine CXCL12 , Neoplasms , Radiation Injuries , Radiation-Protective Agents , Signal Transduction , Animals , Humans , Chemokine CXCL12/metabolism , Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Radiation Tolerance/drug effects , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Radiotherapy, Image-Guided/methods , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Chemokines, CXC/antagonists & inhibitors
4.
Int J Radiat Biol ; 100(5): 791-801, 2024.
Article in English | MEDLINE | ID: mdl-38442139

ABSTRACT

PURPOSE: Radiotherapy with bladder preservation is highly acceptable among patients bearing bladder cancer (BCa), but the occurrence of secondary tolerance (ARR) during treatment is one of the important reasons for the failure of clinical radiotherapy. COX-2 has been frequently reported to be highly expressed and associated with radio-resistance in various cancers. In this study, the feasibility of Taraxasterol (Tara) as a radiosensitizer was investigated, and the target effect of Tara on COX-2 and its underlying mechanism were explored. METHODS AND MATERIALS: The toxicity of Tara toward BCa cells was detected with the MTT method and cells in response to IR or Tara + IR were compared by clone formation assay. Next, a small RNA interference system (siRNA) was employed to decrease endogenous COX-2 expression in BCa cells, and the stem cell-like features and motion abilities of BCa cells under different treatments were investigated using microsphere formation and transwell chamber assay, respectively. Meanwhile, the expression of a series of inflammation-related molecules and stem cell characteristic molecules was determined by qRT-PCR, western blot and ELISA method. In vivo studies, BCa cells were subcutaneously injected into the right flank of each male mouse. Those mice were then grouped and exposed to different treatment: Tara, IR, IR + Tara and untreated control. The volumes of each tumor were measured every two days and target proteins were detected with immunohistochemical (IHC) staining. RESULTS: The results show that COX-2 decline, due to COX-2 knocking-down or Tara treatment, could greatly enhance BCa cells' radiosensitivity and significantly decrease their migration, invasion and microsphere formation abilities, companied with the reduce of JAK2, phos-STAT3, MMP2 and MMP9 expression. However, Tara could not further reduce the expression of an above molecule of cells in COX-2-deficient BCa cells. Correspondingly, Tara treatment could not further enhance those siCOX-2 BCa cells response to IR. CONCLUSIONS: Our data support that Tara can improve the radiosensitivity of BCa cells by targeting COX-2/PGE2. The mechanism may involve regulating STAT3 phosphorylation, DNA damage response protein activation, and expression of MMP2/MMP9.


Subject(s)
Cyclooxygenase 2 , Janus Kinase 2 , Radiation Tolerance , STAT3 Transcription Factor , Urinary Bladder Neoplasms , Janus Kinase 2/metabolism , Humans , Cyclooxygenase 2/metabolism , Urinary Bladder Neoplasms/radiotherapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , STAT3 Transcription Factor/metabolism , Mice , Radiation Tolerance/drug effects , Dinoprostone/metabolism , Signal Transduction/drug effects , Sterols/pharmacology , Triterpenes/pharmacology , Triterpenes/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Male
5.
Int J Radiat Biol ; 100(5): 715-723, 2024.
Article in English | MEDLINE | ID: mdl-38421209

ABSTRACT

PURPOSE: DNA damage response (DDR) is the principal mechanism regulating genomic stability and cell cycle checkpoint activation by coordinating DNA repair and apoptotic pathways. Ataxia telangiectasia and Rad3-related protein (ATR) play a significant role in the DDR due to its capability to detect a wide spectrum of DNA damage. Therefore, targeting DDR, specifically ATR, is a promising therapeutic strategy in cancer treatment. Furthermore, the inhibition of ATR sensitizes cancer cells to radiotherapy (RT). Herein, we, for the first time, investigated the synergistic effects of Elimusertib (BAY-1895344) as a highly potent selective ATR inhibitor with RT combination in triple-negative breast cancer (TNBC), in vitro. METHODS: MDA-MB-231 TNBC cells were firstly treated with different concentrations of Elimusertib for 24 h and then exposed to 4 and 8 Gy of X-ray irradiation. After post-irradiation for 72 h, WST-1, Annexin V, cell cycle, acridine orange/propidium iodide, mitochondria staining and western blot analysis were conducted. RESULTS: Our findings showed that 4 Gy irradiation and lower doses (especially 2 and 4 nM) of Elimusertib combination exerted a considerable anticancer activity at 72 h post-irradiation through apoptotic cell death, marked nuclear and mitochondrial damages and the suppression of ATR-Chk1 based DDR mechanism. CONCLUSION: ATR inhibition by Elimusertib in combination with RT may be a promising new treatment strategy in the treatment of TNBC. However, further experiments should be performed to elucidate the underlying molecular mechanisms of the therapeutic efficacy of this combination treatment and its association with DNS repair mechanisms in TNBC, in vitro and in vivo.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Radiation Tolerance , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/radiotherapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Radiation Tolerance/drug effects , Apoptosis/drug effects , Apoptosis/radiation effects , Female , DNA Damage
6.
J Transl Med ; 21(1): 738, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37858134

ABSTRACT

BACKGROUND: Autophagy is involved in nasopharyngeal carcinoma (NPC) radioresistance. Replication protein A 1 (RPA1) and RPA3, substrates of the RPA complex, are potential therapeutic targets for reversing NPC radioresistance. Nevertheless, the role of RPA in autophagy is not adequately understood. This investigation was performed to reveal the cytotoxic mechanism of a pharmacologic RPA inhibitor (RPAi) in NPC cells and the underlying mechanism by which RPAi-mediated autophagy regulates NPC radiosensitivity. METHODS AND RESULTS: We characterized a potent RPAi (HAMNO) that was substantially correlated with radiosensitivity enhancement and proliferative inhibition of in vivo and in NPC cell lines in vitro. We show that the RPAi induced autophagy at multiple levels by inducing autophagic flux, AMPK/mTOR pathway activation, and autophagy-related gene transcription by decreasing glycolytic function. We hypothesized that RPA inhibition impaired glycolysis and increased NPC dependence on autophagy. We further demonstrated that combining autophagy inhibition with chloroquine (CQ) treatment or genetic inhibition of the autophagy regulator ATG5 and RPAi treatment was more effective than either approach alone in enhancing the antitumor response of NPC to radiation. CONCLUSIONS: Our study suggests that HAMNO is a potent RPAi that enhances radiosensitivity and induces autophagy in NPC cell lines by decreasing glycolytic function and activating autophagy-related genes. We suggest a novel treatment strategy in which pharmacological inhibitors that simultaneously disrupt RPA and autophagic processes improve NPC responsiveness to radiation.


Subject(s)
Antineoplastic Agents , Autophagy , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Radiation Tolerance , Replication Protein A , Humans , Antineoplastic Agents/therapeutic use , Apoptosis , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Replication Protein A/antagonists & inhibitors , Replication Protein A/genetics , Autophagy-Related Protein 5/genetics
7.
Biomed Pharmacother ; 165: 115107, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37423171

ABSTRACT

The antitumor effects of traditional drugs have received increasing attention and active antitumor components extracted from traditional drugs have shown good efficacy with minimal adverse events. Cepharanthine(CEP for short) is an active component derived from the Stephania plants of Menispermaceae, which can regulate multiple signaling pathways alone or in combination with other therapeutic drugs to inhibit tumor cell proliferation, induce apoptosis, regulate autophagy, and inhibit angiogenesis, thereby inhibiting tumor progression. Therefore, we retrieved studies concerning CEP's antitumor effects in recent years and summarized the antitumor mechanism and targets, in order to gain new insights and establish a theoretical basis for further development and application of CEP.


Subject(s)
Antineoplastic Agents , Benzodioxoles , Benzylisoquinolines , Benzylisoquinolines/chemistry , Benzylisoquinolines/pharmacology , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Humans , Cell Line, Tumor , Apoptosis/drug effects , Radiation Tolerance/drug effects , Autophagy/drug effects , Angiogenesis/drug effects
8.
Int J Mol Sci ; 23(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36430560

ABSTRACT

(1) Destabilization of microtubule dynamics is a primary strategy to inhibit fast growing tumor cells. The low cytotoxic derivative of microtubule inhibitor D-24851, named BPR0C261 exhibits antitumor activity via oral administration. In this study, we investigated if BPR0C261 could modulate the radiation response of human non-small cell lung cancer (NSCLC) cells with or without p53 expression. (2) Different doses of BPR0C261 was used to treat human NSCLC A549 (p53+/+) cells and H1299 (p53-/-) cells. The cytotoxicity, radiosensitivity, cell cycle distribution, DNA damage, and protein expression were evaluated using an MTT assay, a colony formation assay, flow cytometry, a comet assay, and an immunoblotting analysis, respectively. (3) BPR0C261 showed a dose-dependent cytotoxicity on A549 cells and H1299 cells with IC50 at 0.38 µM and 0.86 µM, respectively. BPR0C261 also induced maximum G2/M phase arrest and apoptosis in both cell lines after 24 h of treatment with a dose-dependent manner. The colony formation analysis demonstrated that a combination of low concentration of BPR0C261 and X-rays caused a synergistic radiosensitizing effect on NSCLC cells. Additionally, we found that a low concentration of BPR0C261 was sufficient to induce DNA damage in these cells, and it increased the level of DNA damage induced by a fractionation radiation dose (2 Gy) of conventional radiotherapy. Furthermore, the p53 protein level of A549 cell line was upregulated by BPR0C261. On the other hand, the expression of PTEN tumor suppressor was found to be upregulated in H1299 cells but not in A549 cells under the same treatment. Although radiation could not induce PTEN in H1299 cells, a combination of low concentration of BPR0C261 and radiation could reverse this situation. (4) BPR0C261 exhibits specific anticancer effects on NSCLC cells by the enhancement of DNA damage and radiosensitivity with p53-dependent and p53-independent/PTEN-dependent manners. The combination of radiation and BPR0C261 may provide an important strategy for the improvement of radiotherapeutic treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Radiation Tolerance , Tumor Suppressor Protein p53 , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/genetics , Microtubules/drug effects , Microtubules/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use
9.
Cell Chem Biol ; 29(10): 1517-1531.e7, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36206753

ABSTRACT

Beyond synthesizing telomere repeats, the telomerase reverse transcriptase (TERT) also serves multiple other roles supporting cancer growth. Blocking telomerase to drive telomere erosion appears impractical, but TERT's non-canonical activities have yet to be fully explored as cancer targets. Here, we used an irreversible TERT inhibitor, NU-1, to examine impacts on resistance to conventional cancer therapies. In vitro, inhibiting TERT sensitized cells to chemotherapy and radiation. NU-1 delayed repair of double-strand breaks, resulting in persistent DNA damage signaling and cellular senescence. Although NU-1 alone did not impact growth of syngeneic CT26 tumors in BALB/c mice, it dramatically enhanced the effects of radiation, leading to immune-dependent tumor elimination. Tumors displayed persistent DNA damage, suppressed proliferation, and increased activated immune infiltrate. Our studies confirm TERT's role in limiting genotoxic effects of conventional therapy but also implicate TERT as a determinant of immune evasion and therapy resistance.


Subject(s)
Radiation Tolerance , Telomerase , Animals , Mice , Cellular Senescence/drug effects , DNA Damage/drug effects , Radiation Tolerance/drug effects , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Telomere
10.
Dokl Biochem Biophys ; 506(1): 185-190, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36303049

ABSTRACT

The effect of xylazine-zoletyl anesthesia on the radiosensitivity of mice irradiated with protons, carbon ions in two regions of the Bragg curve, and X-ray radiation was studied according to the criteria of 30-day survival, dynamics of death, and the average life span of dead mice. The maximum effect of anesthesia by 3.3 times was observed by a decrease in the death of animals during irradiation with carbon ions at the Bragg peak; in the case of irradiation before the peak, the effect was 1.2 times. In the case of proton irradiation at the Bragg peak, the protective effect of anesthesia by a factor of 1.7 was observed only at a dose of 8.5 Gy. When mice were irradiated with X-rays in the dose range of 6.0-8.5 Gy, the anesthesia effect coefficient was 1.7-2. According to the 30-day survival method, it was shown that the use of a xylazine-zoletil mixture significantly changes the radiosensitivity of mice depending on the radiation dose and the radiation source quality.


Subject(s)
Anesthesia , Dose-Response Relationship, Radiation , Radiation Tolerance , Xylazine , Animals , Mice , Anesthesia/methods , Carbon , Ions , Protons , Radiation Tolerance/drug effects , X-Rays , Xylazine/pharmacology
11.
Br J Cancer ; 127(5): 927-936, 2022 09.
Article in English | MEDLINE | ID: mdl-35618789

ABSTRACT

PURPOSE: Radiation therapy (RT) and hormone receptor (HR) inhibition are used for the treatment of HR-positive breast cancers; however, little is known about the interaction of the androgen receptor (AR) and estrogen receptor (ER) in response to RT in AR-positive, ER-positive (AR+/ER+) breast cancers. Here we assessed radiosensitisation of AR+/ER+ cell lines using pharmacologic or genetic inhibition/degradation of AR and/or ER. METHODS: Radiosensitisation was assessed with AR antagonists (enzalutamide, apalutamide, darolutamide, seviteronel, ARD-61), ER antagonists (tamoxifen, fulvestrant) or using knockout of AR. RESULTS: Treatment with AR antagonists or ER antagonists in combination with RT did not result in radiosensitisation changes (radiation enhancement ratios [rER]: 0.76-1.21). Fulvestrant treatment provided significant radiosensitisation of CAMA-1 and BT-474 cells (rER: 1.06-2.0) but not ZR-75-1 cells (rER: 0.9-1.11). Combining tamoxifen with enzalutamide did not alter radiosensitivity using a 1 h or 1-week pretreatment (rER: 0.95-1.14). Radiosensitivity was unchanged in AR knockout compared to Cas9 cells (rER: 1.07 ± 0.11), and no additional radiosensitisation was achieved with tamoxifen or fulvestrant compared to Cas9 cells (rER: 0.84-1.19). CONCLUSION: While radiosensitising in AR + TNBC, AR inhibition does not modulate radiation sensitivity in AR+/ER+ breast cancer. The efficacy of ER antagonists in combination with RT may also be dependent on AR expression.


Subject(s)
Breast Neoplasms , Radiation Tolerance , Receptors, Androgen , Receptors, Estrogen , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Androgens , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , Humans , Naphthalenes , Piperidines , Pyrrolidines , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Thiazoles , Triazoles
12.
Int J Oncol ; 60(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35179215

ABSTRACT

Atypical teratoid rhabdoid tumor (ATRT) is a highly aggressive pediatric brain tumor. Despite radiation, aggressive chemotherapy and autologous stem cell rescue, children usually have a poor survival time. In the present study, the role of TP53/MDM2 interaction in ATRT was investigated. A functional genomic screen identified the TP53/MDM2 axis as a therapeutic target in the central nervous system (CNS) ATRT. Gene expression analysis revealed that all ATRT sub­groups expressed high levels of MDM2, which is a negative regulator of TP53. Using cell viability, colony formation and methylcellulose assays it was found that genetic MDM2 inhibition with short hairpin RNA or chemical MDM2 inhibition with small molecule inhibitors, Nutlin3 and idasanutlin (RG7388) decreased the growth of ATRT cell lines. Furthermore, idasanutlin significantly decreased the growth of intracranial orthotopic ATRT brain tumors, as evaluated using T2 MRI, and prolonged survival time relative to control animals. MRI of intracranial tumors showed that diffusion coefficient, an effective marker for successful treatment, significantly increased with idasanutlin treatment showing tumor necrosis/apoptosis. Immunohistochemistry revealed an increased number of caspase­3­positive cells in the idasanutlin treatment group, confirming the induction of apoptosis in vivo. Using flow cytometry and western blot analysis we show that inhibition of MDM2 enhanced radiation sensitivity in vitro by potentiating DNA damage via the induction of the TP53/Bax/Puma proapoptotic axis. Furthermore, DNA damage was associated with increased mitochondrial reactive oxygen species accumulation. The present study demonstrated that MDM2 expression level was increased in ATRT patient samples and MDM2 inhibition suppressed ATRT cell growth in vitro, and leads to apoptosis in vivo. MDM2 inhibition potentiates DNA damage and sensitizes ATRT cells to radiation. These findings highlight the TP53/MDM2 axis as a rational therapeutic target in CNS ATRT.


Subject(s)
Proto-Oncogene Proteins c-mdm2/drug effects , Radiation Tolerance/drug effects , Rhabdoid Tumor/radiotherapy , Tumor Suppressor Protein p53/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Colorado , Humans , Radiation Tolerance/genetics , Teratoma/radiotherapy
13.
Biomed Pharmacother ; 147: 112686, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35124385

ABSTRACT

Cancer is one of the main causes of human mortality and brain tumors, including invasive pituitary adenomas, medulloblastomas and glioblastomas are common brain malignancies with poor prognosis. Therefore, the development of innovative management strategies for refractory cancers and brain tumors is important. In states of mitochondrial dysfunction - commonly encountered in malignant cells - cells mostly shift to anaerobic glycolysis by increasing the expression of LDHA (Lactate Dehydrogenase-A) gene. Oxamate, an isosteric form of pyruvate, blocks LDHA activity by competing with pyruvate. By blocking LDHA, it inhibits protumorigenic cascades and also induces ROS (reactive oxygen species)-induced mitochondrial apoptosis of cancer cells. In preclinical studies, oxamate blocked the growth of invasive pituitary adenomas, medulloblastomas and glioblastomas. Oxamate also increases temozolomide and radiotherapy sensitivity of glioblastomas. Oxamate is highly polar, which may preclude its clinical utilization due to low penetrance through cell membranes. However, this obstacle could be overcome with nanoliposomes. Moreover, different oxamate analogs were developed which inhibit LDHC4, an enzyme also involved in cancer progression and germ cell physiology. Lastly, phenformin, an antidiabetic agent, exerts anticancer effects via complex I inhibition in the mitochondria and leading the overproduction of ROS. Oxamate combination with phenformin reduces the lactic acidosis-causing side effect of phenformin while inducing synergistic anticancer efficacy. In sum, oxamate as a single agent and more efficiently with phenformin has high potential to slow the progression of aggressive cancers with special emphasis to brain tumors.


Subject(s)
Brain Neoplasms/pathology , L-Lactate Dehydrogenase/antagonists & inhibitors , Oxamic Acid/pharmacology , Animals , Cell Line, Tumor , Glycolysis/physiology , Humans , L-Lactate Dehydrogenase/metabolism , Mitochondria/metabolism , Neoplasms/pathology , Phenformin/pharmacology , Radiation Tolerance/drug effects , Reactive Oxygen Species/metabolism , Temozolomide/pharmacology
14.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35163494

ABSTRACT

Usher syndrome (USH) is a rare autosomal recessive disease characterized by the combination of hearing loss, visual impairment due to retinitis pigmentosa, and in some cases vestibular dysfunctions. Studies published in the 1980s reported that USH is associated with cellular radiosensitivity. However, the molecular basis of this particular phenotype has not yet been documented. The aim of this study was therefore to document the radiosensitivity of USH1-a subset of USH-by examining the radiation-induced nucleo-shuttling of ATM (RIANS), as well as the functionality of the repair and signaling pathways of the DNA double-strand breaks (DSBs) in three skin fibroblasts derived from USH1 patients. The clonogenic cell survival, the micronuclei, the nuclear foci formed by the phosphorylated forms of the X variant of the H2A histone (É£H2AX), the phosphorylated forms of the ATM protein (pATM), and the meiotic recombination 11 nuclease (MRE11) were used as cellular and molecular endpoints. The interaction between the ATM and USH1 proteins was also examined by proximity ligation assay. The results showed that USH1 fibroblasts were associated with moderate but significant radiosensitivity, high yield of micronuclei, and impaired DSB recognition but normal DSB repair, likely caused by a delayed RIANS, suggesting a possible sequestration of ATM by some USH1 proteins overexpressed in the cytoplasm. To our knowledge, this report is the first radiobiological characterization of cells from USH1 patients at both molecular and cellular scales.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Radiation Tolerance/genetics , Usher Syndromes/enzymology , Usher Syndromes/genetics , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , Diphosphonates/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/radiation effects , Histones/metabolism , Humans , Kinetics , MRE11 Homologue Protein/metabolism , Micronuclei, Chromosome-Defective/radiation effects , Models, Biological , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Subcellular Fractions/radiation effects
15.
Cells ; 11(2)2022 01 17.
Article in English | MEDLINE | ID: mdl-35053421

ABSTRACT

Recurrence in hepatocellular carcinoma (HCC) after conventional treatments is a crucial challenge. Despite the promising progress in advanced targeted therapies, HCC is the fourth leading cause of cancer death worldwide. Radionuclide therapy can potentially be a practical targeted approach to address this concern. Rhenium-188 (188Re) is a ß-emitting radionuclide used in the clinic to induce apoptosis and inhibit cell proliferation. Although adherent cell cultures are efficient and reliable, appropriate cell-cell and cell-extracellular matrix (ECM) contact is still lacking. Thus, we herein aimed to assess 188Re as a potential therapeutic component for HCC in 2D and 3D models. The death rate in treated Huh7 and HepG2 lines was significantly higher than in untreated control groups using viability assay. After treatment with 188ReO4, Annexin/PI data indicated considerable apoptosis induction in HepG2 cells after 48 h but not Huh7 cells. Quantitative RT-PCR and western blotting data also showed increased apoptosis in response to 188ReO4 treatment. In Huh7 cells, exposure to an effective dose of 188ReO4 led to cell cycle arrest in the G2 phase. Moreover, colony formation assay confirmed post-exposure growth suppression in Huh7 and HepG2 cells. Then, the immunostaining displayed proliferation inhibition in the 188ReO4-treated cells on 3D scaffolds of liver ECM. The PI3-AKT signaling pathway was activated in 3D culture but not in 2D culture. In nude mice, Huh7 cells treated with an effective dose of 188ReO4 lost their tumor formation ability compared to the control group. These findings suggest that 188ReO4 can be a potential new therapeutic agent against HCC through induction of apoptosis and cell cycle arrest and inhibition of tumor formation. This approach can be effectively combined with antibodies and peptides for more selective and personalized therapy.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Radioisotopes/pharmacology , Rhenium/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , G2 Phase/drug effects , Humans , Inhibitory Concentration 50 , Mice, Nude , Mitosis/drug effects , Phenotype , Radiation Tolerance/drug effects
16.
Anticancer Drugs ; 33(1): e622-e627, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34407046

ABSTRACT

Brain metastasis is a common cause of death in HER2-positive breast cancer patients. Currently, it is mainly treated by whole-brain radiotherapy. Pyrotinib is an irreversible pan-ErbB inhibitor, which has demonstrated promising tumor-suppressing activity and acceptable tolerance in previous phase trials. In the present study, we evaluated the efficacy of pyrotinib on HER2-positive brain metastatic breast cancer patients treated with whole-brain radiotherapy. A total of 20 such patients were separated into pyrotinib plus capecitabine and capecitabine-only groups in a 1:1 ratio. All patients met either the primary or secondary endpoints. Oral admission of pyrotinib together with radiotherapy can significantly increase the overall response rate, progression-free survival, time to progression and duration of response of HER2+ brain metastatic breast cancer patients, without causing extra adverse events. In addition, pyrotinib can enhance the radiosensitivity of in-vitro cultured HER2+ breast cancer cell lines. The outcome of our study suggests that pyrotinib might be an effective medication to enhance the tumor radiosensitivity of HER2-positive brain metastatic breast cancer patients.


Subject(s)
Acrylamides/therapeutic use , Aminoquinolines/therapeutic use , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Radiation Tolerance/drug effects , Acrylamides/administration & dosage , Acrylamides/adverse effects , Adult , Aged , Aminoquinolines/administration & dosage , Aminoquinolines/adverse effects , Cell Line, Tumor , Female , Humans , Middle Aged , Progression-Free Survival , Receptor, ErbB-2/antagonists & inhibitors
17.
Lab Invest ; 102(3): 298-311, 2022 03.
Article in English | MEDLINE | ID: mdl-34773069

ABSTRACT

Pancreatic cancer (PC) is highly resistant to chemo and radiotherapy. Radiation-induced fibrosis (RIF) is a major cause of clinical concern for various malignancies, including PC. In this study, we aimed to evaluate the radiosensitizing and anti-RIF potential of fluvastatin in PC. Short-term viability and clonogenic survival assays were used to evaluate the radiosensitizing potential of fluvastatin in multiple human and murine PC cell lines. The expression of different proteins was analyzed to understand the mechanisms of fluvastatin-mediated radiosensitization of PC cells and its anti-RIF effects in both mouse and human pancreatic stellate cells (PSCs). Finally, these effects of fluvastatin and/or radiation were assessed in an immune-competent syngeneic murine model of PC. Fluvastatin radiosensitized multiple PC cell lines, as well as radioresistant cell lines in vitro, by inhibiting radiation-induced DNA damage repair response. Nonmalignant cells, such as PSCs and NIH3T3 cells, were less sensitive to fluvastatin-mediated radiosensitization than PC cells. Interestingly, fluvastatin suppressed radiation and/or TGF-ß-induced activation of PSCs, as well as the fibrogenic properties of these cells in vitro. Fluvastatin considerably augmented the antitumor effect of external radiation therapy and also suppressed intra-tumor RIF in vivo. These findings suggested that along with radiation, fluvastatin co-treatment may be a potential therapeutic approach against PC.


Subject(s)
Fluvastatin/pharmacology , Pancreatic Neoplasms/pathology , Radiation Tolerance/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/radiation effects , Fibrosis/prevention & control , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Zebrafish/embryology
18.
Cancer Rep (Hoboken) ; 5(12): e1553, 2022 12.
Article in English | MEDLINE | ID: mdl-34533293

ABSTRACT

BACKGROUND: Ionizing radiation (IR) is a standard modality for the management of solid tumors. Apart from its killing effects, IR can induce pro-survival factors leading to radioresistance of cancer. Mechanistic understanding of radiation resistance is warranted to overcome the pro-survival effects of IR. AIM: The aim of this study was to investigate the role of upstream stimulatory factor-1 (USF-1) in the induction of radioresistance in prostate cancer and its targeting by histone deacetylase (HDAC) inhibitors to reverse resistance. METHODS AND RESULTS: This study reports here that USF-1 is a marker for radioresistance in PC-3 cells. Using protein-DNA array analysis, it was documented that DNA binding activity of USF-1 was elevated following IR in PC-3 cells. Novel HDAC inhibitors downregulated USF-1 binding either alone or in combination with IR. A 5 Gy dose of IR induced the expression of target genes of USF-1 (human telomerase reverse transcriptase [hTERT], IGF2R, CyclinB1, and Cdk1), however, HDAC inhibitors alone or in combination with IR reduced their expression as measured by real time RT PCR analysis. Furthermore, immunofluorescence analysis revealed that while USF-1 localized primarily in the nucleus following IR, it localized in the cytoplasm when treated with HDAC inhibitors/combination. Maximum effects of modulation of USF-1 expression (overexpression or suppression) were observed on hTERT activity as determined by dual-luciferase reporter assay. To further confirm the role of USF-1 in radioresistance, cell growth was analyzed using the real-time cell electronic sensing (RT-CES) system. This study found that USF-1-transfected cells proliferated faster than the vector-transfected cells with or without treatments with HDAC inhibitors/IR/combination. Colony forming assay also confirmed that USF-1 overexpression led to increased survival following IR. Importantly, colony-forming assay demonstrated that HDAC inhibitors reversed the radioresistance in both PC-3 and DU-145 cells. CONCLUSION: These studies demonstrate that HDAC inhibitors reverse the radioresistance in prostate cancer through down-modulation of USF-1-mediated transactivation of target genes involved in cell proliferation and cell cycle.


Subject(s)
DNA-Binding Proteins , Histone Deacetylase Inhibitors , Prostatic Neoplasms , Radiation Tolerance , Upstream Stimulatory Factors , Humans , Male , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism , Radiation Tolerance/drug effects , PC-3 Cells , Down-Regulation
19.
Cancer Rep (Hoboken) ; 5(12): e1326, 2022 12.
Article in English | MEDLINE | ID: mdl-34472223

ABSTRACT

BACKGROUND: Calreticulin (CRT), an endoplasmic reticulum-resident protein generally overexpressed in cancer cells, is associated with radiation resistance. CRT shows higher transacetylase activity, as shown by us earlier, in the presence of the polyphenolic acetates (like 7, 8-diacetoxy-4-methylcoumarin, DAMC) and modifies the activity of a number of proteins, thereby influencing cell signaling. AIM: To investigate the relationship between CRT expression and radiation response in a human glioma cell line and to evaluate the radiomodifying effects of DAMC. METHODS AND RESULTS: Studies were carried out in an established human glioma cell line (BMG-1) and its isogenic clone overexpressing CRT (CROE, CRT-overexpressing cells) by analyzing clonogenic survival, cell proliferation, micronuclei analysis, and protein levels by Western blotting as parameters of responses. CRT overexpression conferred resistance against radiation-induced cell death in CROE cells (D37  = 7.35 Gy, D10  = 12.6 Gy and D0  = 7.25 Gy) as compared to BMG-1 cells (D37  = 5.70 Gy, D10  = 9.2 Gy and D0  = 5.6 Gy). A lower level of radiation-induced micronuclei formation observed in CROE cells suggested that reduced induction and/or enhanced DNA repair partly contributed to the enhanced radioresistance. Consistent with this suggestion, we noted that CRT-mediated radioresistance was coupled with enhanced grp78 level and reduced P53 activation-mediated prodeath signaling, while no changes were noted in acetylation of histone H4. DAMC-enhanced radiation-induced delayed (secondary) apoptosis, which was higher in CROE cells. CONCLUSION: CRT overexpression confers resistance against radiation-induced death of human glioma cells, which can be overcome by the polyphenolic acetate DAMC.


Subject(s)
Acetates , Calreticulin , Coumarins , Glioma , Polyphenols , Radiation Tolerance , Radiation-Sensitizing Agents , Humans , Acetates/pharmacology , Calreticulin/genetics , Calreticulin/metabolism , Cell Line , Glioma/drug therapy , Glioma/metabolism , Polyphenols/pharmacology , Coumarins/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radiation Tolerance/drug effects
20.
Exp Cell Res ; 410(1): 112946, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34826424

ABSTRACT

The principle underlying radiotherapy is to kill cancer cells while minimizing the harmful effects on non-cancer cells, which has still remained as a major challenge. In relation, ferroptosis has recently been proposed as a novel mechanism of radiation-induced cell death. In this study, we investigated and demonstrated the role of Hemin as an iron overloading agent in the generation of reactive oxygen species (ROS) induced by ionizing radiation in lung cancer and non-cancer cells. It was found that the presence of Hemin in irradiated lung cancer cells enhanced the productivity of initial ROS, resulting in lipid peroxidation and subsequent ferroptosis. We observed that application of Hemin as a co-treatment increased the activity of GPx4 degradation in both cancer and normal lung cells. Furthermore, Hemin protected normal lung cells against radiation-induced cell death, in that it suppressed ROS after radiation, and boosted the production of bilirubin which was a lipophilic ROS antioxidant. In addition, we demonstrated significant FTH1 expression in normal lung cells when compared to lung cancer cells, which prevented iron from playing a role in increasing IR-induced cell death. Our findings demonstrated that Hemin had a dual function in enhancing the radiosensitivity of ferroptosis in lung cancer cells while promoting cell survival in normal lung cells.


Subject(s)
Ferritins/genetics , Hemin/pharmacology , Lung Neoplasms/radiotherapy , Oxidoreductases/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , A549 Cells , Animals , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Hemin/chemistry , Heterografts , Humans , Iron/metabolism , Lipid Peroxidation/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Radiation Tolerance/drug effects , Radiation, Ionizing , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...