Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cancer Discov ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552003

RESUMEN

Resistance to poly (ADP-ribose) polymerase inhibitors (PARPi) limits the therapeutic efficacy of PARP inhibition in treating breast cancer susceptibility gene 1 (BRCA1)-deficient cancers. Here we reveal that BRCA1 has a dual role in regulating ferroptosis. BRCA1 promotes the transcription of voltage-dependent anion channel 3 (VDAC3) and glutathione peroxidase 4 (GPX4); consequently, BRCA1 deficiency promotes cellular resistance to erastin-induced ferroptosis but sensitizes cancer cells to ferroptosis induced by GPX4 inhibitors (GPX4i). In addition, nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and defective GPX4 induction unleash potent ferroptosis in BRCA1-deficient cancer cells upon PARPi and GPX4i co-treatment. Finally, we show that xenograft tumors derived from BRCA1-mutant breast cancer patients with PARPi resistance exhibit decreased GPX4 expression and high sensitivity to PARP and GPX4 co-inhibition. Our results show that BRCA1 deficiency induces a ferroptosis vulnerability to PARP and GPX4 co-inhibition and inform a therapeutic strategy for overcoming PARPi resistance in BRCA1-deficient cancers.

2.
Eur J Oncol Nurs ; 68: 102469, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38039708

RESUMEN

PURPOSE: Cancer-related fatigue (CRF) is a highly prevalent and debilitating symptom reported by breast cancer survivors (BCS). CRF has been associated with the co-occurrence of anxiety, depression, poor sleep quality, cognitive impairment, which are collectively termed as psychoneurophysiological (PNP) symptoms. CRF and these PNP symptoms are often reported during and after treatment with long-lasting distress. It is unclear how CRF and these PNP symptoms influence each other. This study aimed to explore predictive factors (i.e., PNP symptoms and social-demographic factors) of CRF, and test exploratory path models of the relationships of CRF with PNP symptoms (depression, anxiety, sleep disturbance, pain, and cognitive function) in BCS. METHODS: This paper is part of a larger descriptive, correlational, and cross-sectional study. Validated and reliable instruments assessed CRF, depression, anxiety, sleep disturbance, pain, and cognitive function. Descriptive statistics, Pearson correlation, multiple linear regression models, and path analysis were employed. RESULTS: Patients (N = 373) who reported less bodily pain had worst CRF (r = -0.45, p < .01). Significant predictors of CRF included depression, sleep disorder, bodily pain, perceived cognitive ability, and dispositional (state) optimism. Depression alone accounted for 31% of the variance in CRF. An integrative path model with bodily pain, neuropathic pain, CRF, and depression showed a good fit across different indices (CFI = 0.993, RMSEA = 0.047, 90% CI 0-0.12, SRMR = 0.027). CONCLUSIONS: This study identified significant predictors of CRF and revealed a good fit mediation model with significant pathways for CRF, suggesting that a common etiology may underpin the co-occurrence of CRF with PNP symptoms (pain and depression). However, further investigation with longitudinal design is necessary to explore the causal relationships of these symptoms. Evidence-based strategies/interventions are needed to reduce or eliminate the burden of these symptoms on the lives of BCS.


Asunto(s)
Neoplasias de la Mama , Supervivientes de Cáncer , Neuralgia , Humanos , Femenino , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/psicología , Supervivientes de Cáncer/psicología , Estudios Transversales , Fatiga/diagnóstico , Fatiga/epidemiología , Fatiga/etiología , Depresión/epidemiología , Depresión/etiología , Calidad de Vida
4.
Am J Cancer Res ; 13(4): 1209-1239, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37168336

RESUMEN

Nuclear epidermal growth factor receptor (EGFR) has been shown to be correlated with drug resistance and a poor prognosis in patients with cancer. Previously, we have identified a tripartite nuclear localization signal (NLS) within EGFR. To comprehensively determine the functions and underlying mechanism of nuclear EGFR and its clinical implications, we aimed to explore the nuclear export signal (NES) sequence of EGFR that is responsible for interacting with the exportins. We combined in silico prediction with site-directed mutagenesis approaches and identified a putative NES motif of EGFR, which is located in amino acid residues 736-749. Mutation at leucine 747 (L747) in the EGFR NES led to increased nuclear accumulation of the protein via a less efficient release of the exportin CRM1. Interestingly, L747 with serine (L747S) and with proline (L747P) mutations were found in both tyrosine kinase inhibitor (TKI)-treated and -naïve patients with lung cancer who had acquired or de novo TKI resistance and a poor outcome. Reconstituted expression of the single NES mutant EGFRL747P or EGFRL747S, but not the dual mutant along with the internalization-defective or NLS mutation, in lung cancer cells promoted malignant phenotypes, including cell migration, invasiveness, TKI resistance, and tumor initiation, supporting an oncogenic role of nuclear EGFR. Intriguingly, cells with germline expression of the NES L747 mutant developed into B cell lymphoma. Mechanistically, nuclear EGFR signaling is required for sustaining nuclear activated STAT3, but not for Erk. These findings suggest that EGFR functions are compartmentalized and that nuclear EGFR signaling plays a crucial role in tumor malignant phenotypes, leading to tumorigenesis in human cancer.

5.
Nat Cancer ; 3(10): 1211-1227, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36253486

RESUMEN

Poly(ADP-ribose) polymerase (PARP) inhibitors have demonstrated promising clinical activity in multiple cancers. However, resistance to PARP inhibitors remains a substantial clinical challenge. In the present study, we report that anaplastic lymphoma kinase (ALK) directly phosphorylates CDK9 at tyrosine-19 to promote homologous recombination (HR) repair and PARP inhibitor resistance. Phospho-CDK9-Tyr19 increases its kinase activity and nuclear localization to stabilize positive transcriptional elongation factor b and activate polymerase II-dependent transcription of HR-repair genes. Conversely, ALK inhibition increases ubiquitination and degradation of CDK9 by Skp2, an E3 ligase. Notably, combination of US Food and Drug Administration-approved ALK and PARP inhibitors markedly reduce tumor growth and improve survival of mice in PARP inhibitor-/platinum-resistant tumor xenograft models. Using human tumor biospecimens, we further demonstrate that phosphorylated ALK (p-ALK) expression is associated with resistance to PARP inhibitors and positively correlated with p-Tyr19-CDK9 expression. Together, our findings support a biomarker-driven, combinatorial treatment strategy involving ALK and PARP inhibitors to induce synthetic lethality in PARP inhibitor-/platinum-resistant tumors with high p-ALK-p-Tyr19-CDK9 expression.


Asunto(s)
Quinasa de Linfoma Anaplásico , Antineoplásicos , Neoplasias de la Mama , Quinasa 9 Dependiente de la Ciclina , Animales , Femenino , Humanos , Ratones , Quinasa de Linfoma Anaplásico/metabolismo , Antineoplásicos/farmacología , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 9 Dependiente de la Ciclina/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor B de Elongación Transcripcional Positiva , Tirosina/química , Tirosina/metabolismo , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Estados Unidos
6.
Am J Cancer Res ; 12(5): 2363-2375, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693073

RESUMEN

Immunotherapies that block PD-L1/PD-1 immune checkpoint proteins represent a landmark breakthrough in cancer treatment. Although the role of PD-L1 in suppressing T cell activity has been extensively studied, its cancer cell-intrinsic functions are not well understood. Herein, we demonstrated that PD-L1 is important for the repair of DNA damage in cancer cells. Mechanically, depletion of PD-L1 led to the downregulation of the critical molecules involved in the homologous recombination (HR) repair pathway, such as ATM and BRCA1, but did not obviously affect the non-homologous end joining (NHEJ) pathway. Notably, PD-L1 silence sensitized cancer cells to chemotherapy agents and the inhibitor of DNA-PK, which is an important kinase for NHEJ. Furthermore, PD-L1 depletion potentiated DNA damage-induced cGAS-STING pathway and induction of IFNß. The regulation of DNA repair and cGAS-STING pathway by PD-L1 represents its connection with innate immunity that can be exploited to enhance the efficacy of existing immunotherapy. Our findings thus expand the focus of PD-L1 from tumor antigen-specific CD8+ T cells to innate immunity, and support targeting tumor-intrinsic PD-L1 combined with DNA-PK inhibition for tumor eradication, through promoting synthetic lethality and innate immune response.

7.
Am J Cancer Res ; 12(1): 337-354, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141022

RESUMEN

Acquired resistance and clonal heterogeneity are critical challenges in cancer treatment, and the lack of effective computational tools hampers the discovery of new treatments to overcome resistance. Using high-throughput transcriptomic databases of compound perturbation profiles, we have developed a bioinformatic strategy for identifying candidate drugs to overcome resistance with combinatorial therapy. We devised this strategy during an investigation into the acquired resistance against PARP inhibitors (PARPi) in a triple-negative inflammatory breast cancer cell line. In this study, we derived multiple PARPi-resistant clones and characterized their transcriptomic adaptations compared to the parental clone. The transcriptomes of the resistant clones showed substantial heterogeneity, highlighting the importance of characterizing multiple clones from the same tumour. Surprisingly, we found that these transcriptomic changes may not actually confer PARPi resistance, but they may nevertheless induce a shared secondary vulnerability. By modeling our data in relation to transcriptomic perturbation profiles of compounds, we uncovered deficiencies in Ras signaling that resulted from transcriptional adaptation to long-term PARPi treatment across multiple resistant clones. Due to these induced deficiencies, we predicted that the resistant clones would be sensitive to pharmacological reinforcement of PARPi-induced transcriptional adaptation. We then experimentally validated this predicted vulnerability that is shared by multiple resistant clones. Our results thus provide a promising paradigm for integrating transcriptomic data with compound perturbation profiles in order to identify drugs that can exploit an induced vulnerability and overcome therapeutic resistance, thus providing another strategy towards precision oncology.

8.
Nurs Res ; 70(6): 475-480, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34380980

RESUMEN

BACKGROUND: Cancer-related fatigue (CRF) is a highly prevalent, debilitating, and persistent symptom experienced by patients receiving cancer treatments. Up to 71% of men with prostate cancer receiving radiation therapy experience acute and persistent CRF. There is neither an effective therapy nor a diagnostic biomarker for CRF. This pilot study aimed to discover potential biomarkers associated with chronic CRF in men with prostate cancer receiving radiation therapy. METHODS: We used a longitudinal repeated-measures research design. Twenty men with prostate cancer undergoing radiation therapy completed all study visits. CRF was evaluated by a well-established and validated questionnaire, the Patient-Reported Outcomes Measurement Information System for Fatigue (PROMIS-F) Short Form. In addition, peripheral blood mononuclear cells were harvested to quantify ribonucleic acid (RNA) gene expression of mitochondria-related genes. Data were collected before, during, on completion, and 24 months postradiation therapy and analyzed using paired t-tests and repeated-measures analysis of variance. RESULTS: The mean of the PROMIS-F T score was significantly increased over time in patients with prostate cancer, remaining elevated at 24 months postradiation therapy compared to baseline. A significant downregulated BC1 ubiquinol-cytochrome c reductase synthesis-like (BCS1L) was observed over time during radiation therapy and at 24 months postradiation therapy. An increased PROMIS-F score was trended with downregulated BCS1L in patients 24 months after completing radiation therapy. DISCUSSION: This is the first evidence to describe altered messenger RNA for BCS1L in chronic CRF using the PROMIS-F measure with men receiving radiation therapy for prostate cancer. CONCLUSION: Our results suggest that peripheral blood mononuclear cell messenger RNA for BCS1L is a potential biomarker and therapeutic target for radiation therapy-induced chronic CRF in this clinical population.


Asunto(s)
Biomarcadores/sangre , Metabolismo Energético , Fatiga/diagnóstico , Fatiga/etiología , Leucocitos Mononucleares , Neoplasias de la Próstata/complicaciones , Neoplasias de la Próstata/radioterapia , Anciano , Enfermedad Crónica , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Proyectos Piloto , Encuestas y Cuestionarios
9.
FEBS J ; 288(9): 2884-2887, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33570247

RESUMEN

The efficacy of the combination of a PARP inhibitor (PARPi) and an EZH2 inhibitor has been investigated in breast cancer cells with either BRCA1 mutation or BRCA2 mutation. However, earlier studies focused on the efficacy of this combination against BRCA-mutated but not BRCA-proficient breast cancer. Yang et al. observed that PARP1 depletion combined with EZH2 depletion via PRC2 depletion did not affect the growth of BRCA1/2 wild-type breast cancer cells in vitro. Moreover, Yang et al. reported that this combination stimulated synthetic viability of BRCA1/2-proficient breast cancer cells in vivo by regulating the tumor microenvironment to induce angiogenesis and differentiation of M2-type macrophages. The findings of Yang et al. provided evidence that both in vitro and animal models should be employed in the studies of PARPi combination therapies in order to involve the alteration of the tumor microenvironment in these investigations. These studies of PARP inhibition combined with EZH2 inhibition in breast cancer showed that this combination may benefit breast cancer patients carrying BRCA1-mutated tumor, but the combination may also enhance recurrence of BRCA2-mutated tumor and may even promote BRCA-proficient cancer cell survival. Therefore, BRCA1 mutation status should be used to select breast cancer patients for PARPi and EZH2 inhibitor combination treatment in clinical trials in the future.


Asunto(s)
Neoplasias de la Mama , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Humanos , Mutación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Microambiente Tumoral
10.
Am J Cancer Res ; 11(1): 236-250, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33520371

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer that lack effective therapeutic strategies. The response rate of PDAC for treatment with gemcitabine, a current first-line chemotherapeutic for this tumor, is lower than 20%. Identifying key targetable molecules that mediate gemcitabine resistance and developing novel strategies for precision PDAC medicine are urgently needed. Most PDACs have either intratumoral hypoxia or high reactive oxygen species (ROS) production; cytotoxic chemotherapy can elevate ROS production in PDACs. Although excessive ROS production leads to oxidative damage of macromolecules such as DNA, pancreatic cancer cells can survive high DNA damage stress levels. Therefore, identifying molecular mechanisms of overcoming ROS-induced stress in pancreatic cancer cells is important for developing novel therapeutic strategies. ROS-induced DNA damage is predominantly repaired via poly (ADP-ribose) polymerase 1 (PARP1)-mediated DNA repair mechanisms. A recent clinical trial reported that PARP inhibitors are effective in treating pancreatic patients carrying BRCA mutations. However, only less than 10% of pancreatic cancer patients bearing BRCA mutated tumors. Activation of the receptor tyrosine kinase c-MET positively correlates with poor prognosis for PDAC, and our previous study showed that nuclear c-MET can phosphorylate PARP1 at tyrosine 907 under ROS stimulation to promote DNA repair. As described herein, we proposed to expand PARP inhibitor-targeted therapy to more pancreatic cancer patients regardless of BRCA mutation status by combining olaparib, a PARP inhibitor, with c-MET inhibitors as we demonstrated in our previous studies in breast cancer. In this prospective study, we found that ROS-inducing chemotherapeutic drugs such as gemcitabine and doxorubicin stimulated nuclear accumulation of c-MET in BxPC-3 and L3.6pl pancreatic cancer cells. We further showed that combining a c-MET inhibitor with gemcitabine or a PARP inhibitor induced more DNA damage than monotherapy did. Moreover, we demonstrated the synergistic antitumor effects of c-MET inhibitors combined with a PARP inhibitor or gemcitabine in eliminating pancreatic cancer cells. These data suggested that accumulation of ROS in pancreatic cancer cells promotes nuclear localization of c-MET, resulting in resistance to both chemotherapy and PARP inhibitors. Our findings suggest that combining c-MET inhibitors with PARP inhibitors or gemcitabine is a novel, rational therapeutic strategy for advanced pancreatic cancer.

11.
Materials (Basel) ; 13(17)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32859067

RESUMEN

Peri-implantitis is the pathological condition of connective tissue inflammation and the progressive loss of supporting bone around dental implants. One of the primary causes of peri mucositis evolving into peri-implantitis is bacterial infection, including infection from Porphyromonas gingivalis. Enhancing the surface smoothness of implants helps to prevent P. gingivalis adhesion to the implant's surface. Interaction analyses between bacteria and the surface roughness of zirconia (Zr) discs subjected to a glow discharge plasma (GDP) treatment compared with non-plasma-treated autoclaved control Zr discs were done. Examinations of the material prosperities revealed that the GDP-treated Zr group had a smoother surface for a better wettability. The GDP-treated Zr discs improved the proliferation of the osteoblast-like cells MG-63, and the osteoblastic differentiation was assessed through alkaline phosphatase detection and marker gene bone sialoprotein (Bsp) and osteocalcin (OC) induction. Scanning electron microscopy demonstrated a relatively low P. gingivalis adhesion on GDP-treated Zr disks, as well as lower colonization of P. gingivalis compared with the control. Our findings confirmed that the GDP treatment of Zr discs resulted in a significant reduction of P. gingivalis adhesion and growth, demonstrating a positive correlation between surface roughness and bacteria adhesion. Therefore, the GDP treatment of Zr dental implants can provide a method for reducing the risk of peri-implantitis.

12.
Adv Cancer Res ; 147: 59-107, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32593407

RESUMEN

Signaling functions of plasma membrane-localized receptor tyrosine kinases (RTKs) have been extensively studied after they were first described in the mid-1980s. Plasma membrane RTKs are activated by extracellular ligands and cellular stress stimuli, and regulate cellular responses by activating the downstream effector proteins to initiate a wide range of signaling cascades in the cells. However, increasing evidence indicates that RTKs can also be transported into the intracellular compartments where they phosphorylate traditional effector proteins and non-canonical substrate proteins. In general, internalization that retains the RTK's transmembrane domain begins with endocytosis, and endosomal RTK remains active before being recycled or degraded. Further RTK retrograde transport from endosome-Golgi-ER to the nucleus is primarily dependent on membranes vesicles and relies on the interaction with the COP-I vesicle complex, Sec61 translocon complex, and importin. Internalized RTKs have non-canonical substrates that include transcriptional co-factors and DNA damage response proteins, and many nuclear RTKs harbor oncogenic properties and can enhance cancer progression. Indeed, nuclear-localized RTKs have been shown to positively correlate with cancer recurrence, therapeutic resistance, and poor prognosis of cancer patients. Therefore, understanding the functions of nuclear RTKs and the mechanisms of nuclear RTK transport will further improve our knowledge to evaluate the potential of targeting nuclear RTKs or the proteins involved in their transport as new cancer therapeutic strategies.


Asunto(s)
Núcleo Celular/enzimología , Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Endocitosis , Humanos , Neoplasias/enzimología , Neoplasias/patología , Fosforilación , Transporte de Proteínas , Transducción de Señal
13.
Am J Cancer Res ; 10(2): 564-571, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32195027

RESUMEN

Non-small cell lung cancer (NSCLC) patients with c-MET dysregulation may benefit from c-MET inhibitors therapy as inhibition of c-MET activity has emerged as a therapeutic approach against this disease. Although several c-MET inhibitors have been evaluated in multiple clinical trials in lung cancer, their benefits so far have been modest. Thus, furthering our understanding of the mechanisms contributing to the lack of success of c-MET inhibitors in clinical trials is essential toward the development of rational and effective combination strategies. Here we show that exposure of NCSLC cell lines to c-MET inhibitor tivantinib increases their expression of PD-L1, which in turn causes cells to become more resistant to T-cell killing. Mechanistically, inhibition of c-MET suppresses p-GSK3ß, leading to the stabilization of PD-L1 similar to that observed in liver cancer cells. Collectively, our findings suggest a potential crosstalk between c-MET inhibition and immune escape and provide a rationale for the combination therapy of c-MET inhibitors and immune checkpoint blockade in NSCLC.

14.
Am J Cancer Res ; 10(2): 648-661, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32195033

RESUMEN

The limited treatment options and therapeutic failure due to acquired resistance for patients with triple-negative breast cancer (TNBC) represent a significant challenge. Inhibitors against poly (ADP-ribose) polymerase (PARP), olaparib and talazoparib, were recently approved for the treatment of metastatic breast cancer (including TNBC) in patients with germline BRCA1/2 mutations. Despite impressive response rates of ~60%, the prolongation in median progression-free survival with a PARPi is modest, suggesting the emergence of resistance. Several studies have reported that receptor tyrosine kinases (RTKs), such as c-MET (also known as hepatocyte growth factor receptor), are involved in resistance to various anti-neoplastic agents, including PARPi. However, the mechanism by which c-MET contributes to acquired resistance to PARPi in TNBC is not fully understood. In this study, we show that hyperactivated c-Met is detected in TNBC cells with acquired resistance to PARPi, and the combination of talazoparib and crizotinib (a multi-kinase inhibitor that inhibits c-MET) synergistically inhibits proliferation in these cells. Unexpectedly, depleting c-MET had limited effect on talazoparib sensitivity in PARPi-resistant cells. Interestingly, we found evidence of epidermal growth factor receptor (EGFR) hyperactivation and interaction of EGFR/c-Met in these cells. Notably, combining EGFR and PARP inhibitors resulted in greater inhibition of proliferation in c-MET-depleted TNBC cells, and combined c-MET and EGFR inhibition increased sensitivity to talazoparib in TNBC cells with acquired resistance to PARPi. Our findings suggest that combined inhibition of c-MET and EGFR could potentially re-sensitize TNBC to the cytotoxic effects of PARPi.

15.
Nat Commun ; 10(1): 5654, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31827085

RESUMEN

Poly(ADP-ribose)ylation (PARylation) by PAR polymerase 1 (PARP1) and PARylation removal by poly(ADP-ribose) glycohydrolase (PARG) critically regulate DNA damage responses; yet, conflicting reports obscure PARG biology and its impact on cancer cell resistance to PARP1 inhibitors. Here, we found that PARG expression is upregulated in many cancers. We employed chemical library screening to identify and optimize methylxanthine derivatives as selective bioavailable PARG inhibitors. Multiple crystal structures reveal how substituent positions on the methylxanthine core dictate binding modes and inducible-complementarity with a PARG-specific tyrosine clasp and arginine switch, supporting inhibitor specificity and a competitive inhibition mechanism. Cell-based assays show selective PARG inhibition and PARP1 hyperPARylation. Moreover, our PARG inhibitor sensitizes cells to radiation-induced DNA damage, suppresses replication fork progression and impedes cancer cell survival. In PARP inhibitor-resistant A172 glioblastoma cells, our PARG inhibitor shows comparable killing to Nedaplatin, providing further proof-of-concept that selectively inhibiting PARG can impair cancer cell survival.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glicósido Hidrolasas/antagonistas & inhibidores , Neoplasias/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Humanos , Neoplasias/enzimología , Neoplasias/metabolismo , Neoplasias/fisiopatología , Poli ADP Ribosilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Bibliotecas de Moléculas Pequeñas/química
16.
Cancer Manag Res ; 11: 6703-6717, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31410061

RESUMEN

Introduction: Cancer-related fatigue (CRF) is the most debilitating symptom with the greatest adverse side effect on quality of life. The etiology of this symptom is still not understood. The purpose of this study was to examine the relationship between mitochondrial gene expression, mitochondrial oxidative phosphorylation, electron transport chain complex activity, and fatigue in prostate cancer patients undergoing radiotherapy (XRT), compared to patients on active surveillance (AS). Methods: The study used a matched case-control and repeated-measures research design. Fatigue was measured using the revised Piper Fatigue Scale from 52 patients with prostate cancer. Mitochondrial oxidative phosphorylation, electron-transport chain enzymatic activity, and BCS1L gene expression were determined using patients' peripheral mononuclear cells. Data were collected at three time points and analyzed using repeated measures ANOVA. Results: The fatigue score was significantly different over time between patients undergoing XRT and AS (P<0.05). Patients undergoing XRT experienced significantly increased fatigue at day 21 and day 42 of XRT (P<0.01). Downregulated mitochondrial gene (BC1, ubiquinol-cytochrome c reductase, synthesis-like, BCS1L, P<0.05) expression, decreased OXPHOS-complex III oxidation (P<0.05), and reduced activity of complex III were observed over time in patients with XRT. Moreover, increased fatigue was significantly associated with downregulated BCS1L and decreased complex III oxidation in patients undergoing XRT. Conclusion: Our results suggest that BCS1L and complex III in mitochondrial mononuclear cells are potential biomarkers and feasible therapeutic targets for acute XRT-induced fatigue in this clinical population.

17.
Am J Cancer Res ; 9(3): 608-618, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30949414

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are promising targeted therapeutics for breast and ovarian cancers bearing a germline BRCA1/2 mutation (BRCA m), and several have already received regulatory approval in the United States. In patients with a BRCA m cancer, PARPi can increase the burden of unrepaired DNA double-strand breaks by blocking PARP activity and trapping PARP1 onto damaged DNA. Resistance to PARP inhibitors can block the formation of DNA double-strand breaks through BRCA-related DNA repair pathway. MET is a hyper-activated receptor tyrosine kinase expressed in multiple cancer types and the activation contributes to resistance to DNA damage-inducing therapeutic drugs. Our previous study showed that MET inhibition by pan-kinase inhibitors has synergism with PARPi in suppressing growth of breast cancer in vitro and in xenograft tumor models. In this study, we validated the inhibitory effect of novel inhibitors, HS10241 (selective MET inhibitor) and HS10160 (PARPi), to their target respectively in triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC) cells. We further demonstrated that these two inhibitors function synergistically in eliminating TNBC and HGSOC cells; combining with HS10241 increased DNA double-strand breaks induced by HS10160 in cancer cells; and PARP1 tyrosine (Y)-907 phosphorylation (PARP1 p-Y907) can be an effective biomarker as an indicator of MET-mediated PARPi in HGSOC. Our results suggest that the combination of HS10241 and HS10160 may benefit patients bearing tumors overexpressing MET as well as those resistant to single-agent PARPi treatment.

18.
J Biol Chem ; 294(21): 8516-8528, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30962283

RESUMEN

Reactive oxygen species (ROS) are cellular by-products produced from metabolism and also anticancer agents, such as ionizing irradiation and chemotherapy drugs. The ROS H2O2 has high rates of production in cancer cells because of their rapid proliferation. ROS oxidize DNA, protein, and lipids, causing oxidative stress in cancer cells and making them vulnerable to other stresses. Therefore, cancer cell survival relies on maintaining ROS-induced stress at tolerable levels. Hepatocyte growth factor receptor (c-MET) is a receptor tyrosine kinase overexpressed in malignant cancer types, including breast cancer. Full-length c-MET triggers a signal transduction cascade from the plasma membrane that, through downstream signaling proteins, up-regulates cell proliferation and migration. Recently, c-MET was shown to interact and phosphorylate poly(ADP-ribose) polymerase 1 in the nucleus and to induce poly(ADP-ribose) polymerase inhibitor resistance. However, it remains unclear how c-MET moves from the cell membrane to the nucleus. Here, we demonstrate that H2O2 induces retrograde transport of membrane-associated full-length c-MET into the nucleus of human MCF10A and MCF12A or primary breast cancer cells. We further show that knocking down either coatomer protein complex subunit γ1 (COPG1) or Sec61 translocon ß subunit (SEC61ß) attenuates the accumulation of full-length nuclear c-MET. However, a c-MET kinase inhibitor did not block nuclear c-MET transport. Moreover, nuclear c-MET interacted with KU proteins in breast cancer cells, suggesting a role of full-length nuclear c-MET in ROS-induced DNA damage repair. We conclude that a membrane-bound retrograde vesicle transport mechanism facilitates membrane-to-nucleus transport of c-MET in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/enzimología , Membrana Celular/enzimología , Núcleo Celular/enzimología , Peróxido de Hidrógeno/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/patología , Núcleo Celular/genética , Núcleo Celular/patología , Proteína Coatómero/genética , Proteína Coatómero/metabolismo , Daño del ADN , Reparación del ADN , Femenino , Humanos , Proteínas Proto-Oncogénicas c-met/genética , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo
19.
Cancer Res ; 79(4): 819-829, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30573522

RESUMEN

PARP1 inhibitors (PARPi) are currently used in the clinic for the treatment of ovarian and breast cancers, yet their therapeutic efficacy against hepatocellular carcinoma (HCC) has been disappointing. To ensure therapeutic efficacy of PARPi against HCC, a disease often diagnosed at intermediate to advanced stages with no effective treatment options, it is critical to identify not only biomarkers to predict PARPi resistance but also rational treatments to overcome this. Here, we report that a heterodimer of EGFR and MET interacts with and phosphorylates Y907 of PARP1 in the nucleus, which contributes to PARPi resistance. Inhibition of both EGFR and MET sensitized HCC cells to PARPi, and both EGFR and MET are known to be overexpressed in HCC. This report provides an explanation for the poor efficacy of PARPi against HCC and suggests combinatorial treatment consisting of EGFR, MET, and PARP inhibitors may be an effective therapeutic strategy in HCC. SIGNIFICANCE: Regulation of PARP by the c-MET and EGFR heterodimer suggests a potentially effective combination therapy to sensitize HCC to PARPi.


Asunto(s)
Carcinoma Hepatocelular/patología , Resistencia a Antineoplásicos , Neoplasias Hepáticas/patología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-met/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Manag Res ; 10: 6367-6377, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30568498

RESUMEN

INTRODUCTION: Fatigue experienced by cancer patients is one of the most common symptoms with the greatest adverse effect on quality of life, but arguably the least understood. The purpose of this study was to explore changes in integrated mitochondrial function and fatigue in non-metastatic prostate cancer patients receiving localized radiation therapy (XRT). MATERIALS AND METHODS: We proposed a mitochondrial bioenergetics mechanism of radiation-induced fatigue linking impaired oxidative phosphorylation (OXPHOS) through complex III and decreased adenosine triphosphate (ATP) production as consequences of XRT. Integrated mitochondrial function was measured as mitochondrial OXPHOS from patients' peripheral blood mononuclear cells. Fatigue was measured using the revised Piper Fatigue Scale. Data were collected before (day 0) and at day 21 of XRT. RESULTS: At day 21 of XRT, fatigue symptom intensified in 15 prostate cancer patients (P<0.05). Mitochondrial OXPHOS complex III-linked and uncoupled complex III rates were significantly decreased in mononuclear cells at day 21 during XRT compared to that before XRT (P<0.05). Additionally, increased fatigue appeared to be associated with decreased OXPHOS complex III-linked respiration in patients undergoing XRT. CONCLUSION: Fatigue was associated with OXPHOS complex III-linked oxidation and a defect in oxidation starting at complex III in mononuclear cell mitochondria was revealed at day 21 of XRT in 15 prostate cancer patients. Complex III is a potential target for pharmacological and, in particular, nutraceutical interventions, eg, Q10, for design of interventions for CRF.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA