Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 585: 216645, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38280477

RESUMEN

The TP53 gene, encoding the p53 protein, has been a focal point of research since its 1979 discovery, playing a crucial role in tumor suppression. Ferroptosis, a distinct form of cell death characterized by lipid peroxide accumulation, has gained prominence since its recognition in 2012. Recent studies have unveiled an intriguing connection between p53 and ferroptosis, with implications for cancer therapy. Recent research underscores p53 as a novel target for cancer therapy, influencing key metabolic processes in ferroptosis. Notably, p53 represses the expression of the cystine-glutamate antiporter SLC7A11, supporting p53-mediated tumor growth suppression. Furthermore, under metabolic stress, p53 mitigates ferroptosis sensitivity, aiding cancer cells in coping and delaying cell death. This dynamic interplay between p53 and ferroptosis has far-reaching implications for various diseases, particularly cancer. This review provides a comprehensive overview of ferroptosis in cancer cells, elucidating p53's role in regulating ferroptosis, and explores the potential of targeting p53 to induce ferroptosis for cancer therapy. Understanding this complex relationship between p53 and ferroptosis offers a promising avenue for developing innovative cancer treatments.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Genes p53 , Medicina de Precisión , Ferroptosis/genética , Especies Reactivas de Oxígeno/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética
2.
Eur J Pharmacol ; 941: 175497, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36621602

RESUMEN

Cancer often perturbs lipid metabolism, which leads to the alteration of metabolism intermediates, contributing to their deregulated growth and metastasis. Alteration of lipid metabolism shifting to contain more polyunsaturated fatty acids (PUFAs) in membrane phospholipids (PLs) also leads to cancer therapy resistance. High amounts of PL-PUFAs render cancer cells more vulnerable to lipid peroxidation (LPO), predisposing them towards ferroptosis, a new form of iron-dependent oxidative regulated cell death. The commitment of cancer undergoing ferroptotic cell death depends on the adaptive lipidome remodeling, LPO patterns, and LPO scavenging ability in heterogeneous cancer cells. Ferroptosis is receiving attention in cancer research as treating cancers, altering membrane lipid homeostasis, and refractory from conventional therapies. Therefore, a better understanding of the molecular underpinning of lipid metabolism alterations may provide new opportunities for solving cancer resistance. This review intends to understand altered lipid metabolism in cancers and discuss lipid composition and metabolic processes associated with ferroptosis induction in cancers.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Metabolismo de los Lípidos , Peroxidación de Lípido , Oxidación-Reducción , Ácidos Grasos Insaturados
4.
Mol Psychiatry ; 26(9): 5087-5096, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33483691

RESUMEN

The fine-tuning of neuroinflammation is crucial for brain homeostasis as well as its immune response. The transcription factor, nuclear factor-κ-B (NFκB) is a key inflammatory player that is antagonized via anti-inflammatory actions exerted by the glucocorticoid receptor (GR). However, technical limitations have restricted our understanding of how GR is involved in the dynamics of NFκB in vivo. In this study, we used an improved lentiviral-based reporter to elucidate the time course of NFκB and GR activities during behavioral changes from sickness to depression induced by a systemic lipopolysaccharide challenge. The trajectory of NFκB activity established a behavioral basis for the NFκB signal transition involved in three phases, sickness-early-phase, normal-middle-phase, and depressive-like-late-phase. The temporal shift in brain GR activity was differentially involved in the transition of NFκB signals during the normal and depressive-like phases. The middle-phase GR effectively inhibited NFκB in a glucocorticoid-dependent manner, but the late-phase GR had no inhibitory action. Furthermore, we revealed the cryptic role of basal GR activity in the early NFκB signal transition, as evidenced by the fact that blocking GR activity with RU486 led to early depressive-like episodes through the emergence of the brain NFκB activity. These results highlight the inhibitory action of GR on NFκB by the basal and activated hypothalamic-pituitary-adrenal (HPA)-axis during body-to-brain inflammatory spread, providing clues about molecular mechanisms underlying systemic inflammation caused by such as COVID-19 infection, leading to depression.


Asunto(s)
Depresión/metabolismo , FN-kappa B , Receptores de Glucocorticoides , Animales , Encéfalo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Ratones , FN-kappa B/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Glucocorticoides/metabolismo
5.
J Cancer Res Clin Oncol ; 146(10): 2497-2507, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32620987

RESUMEN

PURPOSE: Tumor explant culture systems can mimic the in vivo tumor microenvironment, proposing as a substitute for preclinical studies for prediction of individual treatment response. Therefore, our study evaluated the potential usefulness of ex vivo tumor explants culture assembled into the cell sheets by anticancer drug screening in patients with head and neck squamous cell carcinoma (HNSCC). METHODS: Our model included tumor explants incorporated into cell sheet composing of epithelium and subepithelial stroma using tumor and mucosal samples obtained from the HNSCC patients who underwent surgery. Cell growth, viability, and hypoxia were measured by cell counting kit-8, live/dead assay, propidium iodide, and LOX-1 staining, and were compared among the different treatment groups with vehicle, cisplatin or docetaxel. RESULTS: Tumor explants stably survived in the cell sheet over 10 days after explantation, whereas most of the explants in non-matrix culture became nonviable within 5-8 days with the significant daily decrease of viability. The live tissue areas of tumor explants in the cell sheet maintained over 30 days without significant changes although hypoxic cell areas gradually increased up to 5 days. Tissue viability and live cancer tissue areas significantly decreased after the treatment of cisplatin or docetaxel in the dose and time-dependent manners. CONCLUSION: Our cell sheet-based tumor explants model might be applied to the reliable ex vivo screening for anticancer chemotherapeutics for HNSCC.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Cisplatino/farmacología , Docetaxel/farmacología , Relación Dosis-Respuesta a Droga , Neoplasias de Cabeza y Cuello/sangre , Humanos , Técnicas de Cultivo de Órganos/métodos , Carcinoma de Células Escamosas de Cabeza y Cuello/sangre , Células Tumorales Cultivadas
6.
Theranostics ; 10(17): 7775-7786, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685019

RESUMEN

Rationale: Loss of iron-sulfur cluster function predisposes cancer cells to ferroptosis by upregulating iron-starvation response, but the role of glutaredoxin 5 (GLRX5) silencing in ferroptosis remains unknown. We examined the role of GLRX5 functional loss in promoting ferroptosis in cisplatin-resistant head and neck cancer (HNC) cells. Methods: The effects of sulfasalazine treatment and GLRX5 gene silencing were tested on HNC cell lines and mouse tumor xenograft models. These effects were analyzed concerning cell viability and death, lipid reactive oxygen species (ROS) and mitochondrial iron production, labile iron pool, mRNA/protein expression, and malondialdehyde assays. Results: Cyst(e)ine deprivation, erastin, or sulfasalazine induced ferroptosis in HNC cells, which was relatively less sensitive in cisplatin-resistant HNC cells. Sulfasalazine or cyst(e)ine deprivation-induced ferroptosis resulted from increased lipid peroxidation and intracellular free iron, which were significantly promoted by short-interfering RNA or short hairpin RNA (shRNA) targeting GLRX5 (P<0.05). GLRX5 silencing activated iron-starvation response and boosted up intracellular free iron through the iron-responsive element-binding activity of increased iron regulatory protein (increased transferrin receptor and decreased ferritin). These effects were rescued by resistant GLRX5 cDNA but not by catalytically inactive mutant GLRX5 K101Q. The same results were noted in an in vivo mouse model transplanted with vector or shGLRX5-transduced HNC cells and treated with sulfasalazine. Conclusion: Our data suggest that inhibition of GLRX5 predisposes therapy-resistant HNC cells to ferroptosis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Glutarredoxinas/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Hierro/metabolismo , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mutación , Interferencia de ARN , Sulfasalazina/farmacología , Sulfasalazina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Redox Biol ; 30: 101418, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31931284

RESUMEN

Ferroptosis is a new form of regulated cell death driven by iron-dependent lipid peroxidation. Glutaminolysis and tricarboxylic acid cycle are involved in ferroptosis, but the underlying metabolic process remains unclear. We examined the role of dihydrolipoamide dehydrogenase (DLD) in ferroptosis induction in head and neck cancer (HNC). The effects of cystine deprivation or sulfasalazine treatment and of DLD gene silencing/overexpression were tested on HNC cell lines and mouse tumor xenograft models. These effects were analyzed with regard to cell death, lipid reactive oxygen species (ROS) and mitochondrial iron production, mitochondrial membrane potential, mRNA/protein expression, and α-ketoglutarate dehydrogenase (KGDH)/succinate/aconitase activities. Cystine deprivation induced ferroptosis via glutaminolysis. Cystine deprivation or import inhibition using sulfasalazine induced cancer cell death and increased lipid ROS and mitochondrial iron levels, which had been significantly decreased by short-interfering RNA (siRNA) or short hairpin RNA (shRNA) targeting DLD (P < 0.01) but not by dihydrolipoyl succinyltransferase. The same results were noted in an in vivo mouse model transplanted with vector or shDLD-transduced HN9 cells. After cystine deprivation or sulfasalazine treatment, mitochondrial membrane potential, mitochondrial free iron level, KGDH activity, and succinate content significantly increased (P < 0.001), which had been blocked by DLD siRNA or shRNA and were consequently rescued by resistant DLD cDNA. Cystine deprivation caused iron starvation response and mitochondrial iron accumulation for Fenton reaction and ferroptosis. Our data suggest a close association of DLD with cystine deprivation- or import inhibition-induced ferroptosis.


Asunto(s)
Cistina/deficiencia , Dihidrolipoamida Deshidrogenasa/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Sulfasalazina/farmacología , Animales , Línea Celular Tumoral , Dihidrolipoamida Deshidrogenasa/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Ferroptosis , Silenciador del Gen , Neoplasias de Cabeza y Cuello/genética , Humanos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo
8.
Head Neck ; 41(3): 774-779, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30537435

RESUMEN

BACKGROUND: This study examined the potential use of our newly developed prevascularized oral mucosal cell sheet for the treatment of skin wounds. METHODS: Mucosal cell sheets containing cultured keratinocytes and plasma fibrin without (K sheet) or with a mixture of fibroblasts and endothelial progenitor cells (PV sheet) were transplanted into full-thickness skin excisional wounds of nude mice. RESULTS: This technique was successful for in vitro culture; expanding keratinocytes, fibroblasts, and endothelial progenitor cells; and generating prevascularized mucosal cell sheets. Cell sheets promoted in vivo wound healing with rapid wound closure and less scarring compared to controls. This result was more apparent in the PV than the K sheet (P < .05). Wounds covered with cell sheets showed less expression of TGFB1, ACTA2, and FN1 mRNAs than the controls (P < .05). CONCLUSION: The prevascularized mucosal cell sheet showed in vivo efficacy and tissue plasticity in cutaneous wounds by promoting accelerated healing.


Asunto(s)
Células Progenitoras Endoteliales/trasplante , Fibroblastos/trasplante , Queratinocitos/trasplante , Mucosa Bucal/citología , Úlcera Cutánea/terapia , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Membranas Artificiales , Ratones , Ratones Desnudos , Úlcera Cutánea/etiología , Úlcera Cutánea/patología
9.
J Biomed Mater Res B Appl Biomater ; 107(6): 1779-1785, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30419151

RESUMEN

The current use of steroids or pharmacological immunomodulators for the treatment of intractable oral ulceration is ineffective, necessitating newer cell-based therapeutic approaches. We examined the potential efficacy of an oral mucosa equivalent developed in this study in an in vivo model of repeat major oral ulceration mimicking the intractable oral ulceration observed clinically. Oral mucosal samples and plasma fibrin were obtained from Sprague-Dawley rats. The oral mucosa equivalents were prepared with cultured mucosal keratinocytes and plasma fibrin mixed with cultured fibroblasts. Ulcers were chemically induced on the rat buccal mucosa thrice in 3 weeks and covered with or without mucosa equivalents. Gross and microscopic findings and mRNA expression levels were compared between the ulcer control and mucosa equivalent groups. Oral mucosal keratinocytes and fibroblasts were cultured in vitro to achieve high viability and colony-forming efficiency. The equivalents showed epithelial and subepithelial structures similar to those of oral mucosa and exhibited high p63 positivity. In the in vivo study, ulceration was resolved earlier without significant granulation or scarring in the equivalent group than in control group (p < 0.05). Microscopic examinations revealed rapid re-epithelialization and less fibrosis in the equivalent group than in the control group (p < 0.05). Mucosa equivalent-covered ulcers showed histological characteristics similar to those of the normal buccal mucosa and exhibited lower expression of TGFB1, ACTA2, and FN1 mRNAs than the control group. The in vitro-engineered oral mucosa equivalent promotes ulcer healing without scarring and functional deficits. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1779-1785, 2019.


Asunto(s)
Mucosa Bucal/metabolismo , Úlceras Bucales/metabolismo , Úlceras Bucales/terapia , Cicatrización de Heridas , Animales , Fibrina/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Mucosa Bucal/patología , Úlceras Bucales/patología , Ratas , Ratas Sprague-Dawley
10.
Theranostics ; 8(20): 5703-5712, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555575

RESUMEN

Pre-vascularised cell sheets have been used to promote early angiogenesis and graft survival. However, the use of pre-vascularised mucosal cell sheets for burn wounds has been rarely evaluated. Therefore, we examined the applicability of an oral pre-vascularised mucosal cell sheet that we had previously developed for the treatment of cutaneous burn wounds. Methods: Mucosal keratinocytes, fibroblasts, and endothelial progenitor cells were isolated from the oral mucosa and peripheral blood and were expanded in vitro. Mucosal cell sheets were generated by seeding cultured keratinocytes onto a mixture of fibroblasts, endothelial cells, and fibrin. Third-degree burn wounds were created on the backs of rats and were covered with the cell sheets, skin grafts, or silastic sheets as a control. Gross and microscopic findings and gene expression profiles of wounds were compared among the groups. Results: CD31-positive microvessels were observed in the fibrin-matrix layer of the cell sheet. In the cutaneous burn wound model, the cell sheets promoted wound healing, with accelerated wound closure and less scarring than with silastic sheets and skin grafts. The cell sheets had more microvessels and proliferating cells and less neutrophil infiltration and fibrotic features than the controls or skin grafts. The cell sheet induced higher mRNA expression of KRT14, VEGFA, IL10, and AQP3 and lower mRNA expression of TGFB1, IL6, ICAM1, ACTA2, and FN1 than did the controls or skin grafts. Conclusions: The pre-vascularised mucosal cell sheet promotes cutaneous burn wound healing.


Asunto(s)
Quemaduras/terapia , Mucosa Bucal/fisiología , Trasplante de Piel/métodos , Ingeniería de Tejidos/métodos , Cicatrización de Heridas , Animales , Células Sanguíneas/fisiología , Quemaduras/patología , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/fisiología , Fibroblastos/fisiología , Perfilación de la Expresión Génica , Histocitoquímica , Queratinocitos/fisiología , Ratas , Resultado del Tratamiento
11.
Free Radic Biol Med ; 129: 454-462, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30339884

RESUMEN

Glutathione peroxidase 4 (GPX4) is a regulator of ferroptosis (iron-dependent, non-apoptotic cell death); its inhibition can render therapy-resistant cancer cells susceptible to ferroptosis. However, some cancer cells develop mechanisms protective against ferroptosis; understanding these mechanisms could help overcome chemoresistance. In this study, we investigated the molecular mechanisms underlying resistance to ferroptosis induced by GPX4 inhibition in head and neck cancer (HNC). The effects of two GPX4 inhibitors, (1S, 3R)-RSL3 and ML-162, and of trigonelline were tested in HNC cell lines, including cisplatin-resistant (HN3R) and acquired RSL3-resistant (HN3-rslR) cells. The effects of the inhibitors and trigonelline, as well as of inhibition of the p62, Keap1, or Nrf2 genes, were assessed by cell viability, cell death, lipid ROS production, and protein expression, and in mouse tumor xenograft models. Treatment with RSL3 or ML-162 induced the ferroptosis of HNC cells to varying degrees. RSL3 or ML-162 treatment increased the expression of p62 and Nrf2 in chemoresistant HN3R and HN3-rslR cells, inactivated Keap1, and increased expression of the phospho-PERK-ATF4-SESN2 pathway. Transcriptional activation of Nrf2 was associated with resistance to ferroptosis. Overexpression of Nrf2 by inhibiting Keap1 or Nrf2 gene transfection rendered chemosensitive HN3 cells resistant to RSL3. However, Nrf2 inhibition or p62 silencing sensitized HN3R cells to RSL3. Trigonelline sensitized chemoresistant HNC cells to RSL3 treatment in a mouse model transplanted with HN3R. Thus, activation of the Nrf2-ARE pathway contributed to the resistance of HNC cells to GPX4 inhibition, and inhibition of this pathway reversed the resistance to ferroptosis in HNC.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Glutatión Peroxidasa/genética , Neoplasias de Cabeza y Cuello/genética , Hierro/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Alcaloides/farmacología , Animales , Carbolinas/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Glutatión Peroxidasa/antagonistas & inhibidores , Glutatión Peroxidasa/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Desnudos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
12.
Theranostics ; 8(14): 3964-3973, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30083273

RESUMEN

Epithelial cancer grows in vivo in a microenvironment that comprises tumour, stroma, and immune cells. A three-dimensional (3D) culture model might be able to mimic the tumour microenvironment in vivo; therefore, we developed a new 3D epithelial cancer model using in vitro cell-sheet engineering and compared the results of treatment with several chemotherapeutic drugs among the 3D cell-sheet model, spheroid culture, and 2D cell culture. Methods: The cell sheet comprised keratinocytes and a plasma fibrin matrix containing fibroblasts. Cancer spheroids with or without cancer-associated fibroblasts (CAFs) were interposed between the keratinocytes and fibrin layer. Cell growth, viability, and hypoxia were measured using the cell counting kit-8, LIVE/DEAD assay, and propidium iodide and LOX-1 staining. The morphology, invasion, and mRNA and protein expression were compared among the different cell culture models. Results: Enhanced resistance to sorafenib and cisplatin by cancer spheroids and CAFs was more easily observed in the 3D than in the 2D model. Invasion by cancer-CAF spheroids into the fibrin matrix was more clearly observed in the 3D cell sheet. The expansion of viable cancer cells increased in the 3D cell sheet, particularly in those with CAFs, which were significantly inhibited by treatment with 10 µM sorafenib or 20 µM cisplatin (P < 0.05). TGF-ß1, N-cadherin, and vimentin mRNA and protein levels were higher in the 3D cell-sheet model. Conclusions: The 3D cell sheet-based cancer model could be applied to in vitro observation of epithelial cancer growth and invasion and to anticancer drug testing.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Epitelio/efectos de los fármacos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Técnicas de Cultivo de Órganos/métodos , Esferoides Celulares/efectos de los fármacos , Humanos
13.
Cancer Lett ; 432: 180-190, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-29928961

RESUMEN

Sulfasalazine has been repurposed to induce ferroptotic cancer cell death via inhibition of xc--cystine/glutamate antiporter (xCT). However, cancer cells are capable of developing mechanisms to evade cell death. Therefore, we sought to determine the molecular mechanisms underlying resistance to sulfasalazine-induced ferroptosis in head and neck cancer (HNC). The effects of sulfasalazine and pioglitazone were tested in various HNC cell lines. The effects of these drugs and inhibition and overexpression of CISD2 gene were determined by evaluating viability, cell death, lipid ROS production, mitochondrial iron, and mouse tumor xenograft models. SAS induced ferroptotic cell death in HNC at different levels. CISD2 expression showed an association between its expression and ferroptosis resistance. CISD2 overexpression conferred resistance to ferroptosis by sulfasalazine. Silencing CISD2 gene rendered resistant HNC cells susceptible to sulfasalazine-induced ferroptosis, with increased levels of lipid ROS and mitochondrial ferrous iron. Pioglitazone induced over-accumulation of mitochondrial iron and ROS and sensitized resistant HNC cells to sulfasalazine treatment in vitro and in a mouse tumor-xenograft model. CISD2 inhibition overcomes HNC resistance to ferroptotic cell death induced by sulfasalazine via increased accumulation of mitochondrial ferrous iron and lipid ROS.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Resistencia a Antineoplásicos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Hierro/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Sulfasalazina/farmacología , Animales , Apoptosis , Proliferación Celular , Glutatión/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Head Neck ; 40(2): 394-401, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28990282

RESUMEN

BACKGROUND: We developed a highly efficient in vitro-engineered mucosa equivalent using completely autologous mucosa and blood and investigated its feasibility and efficacy for oral surgical wound healing. METHODS: Small oral mucosa samples were obtained from surgical patients, and keratinocytes and fibroblasts were primarily grown in media without animal products for generating 3D cell sheets. RESULTS: Morphological characteristics of the cell sheets were comparable to those of human mucosa, although p63-positive cells were more numerous in cell sheets. In addition, cell sheets were flexible, expandable, and easy to handle or transfer. In further in vivo rat experiments with deep wounding of the buccal mucosa and soft tissues, controls had significantly thinner epithelium and thicker collagen densities than those with cell sheets. CONCLUSION: Autologous cell sheets can be engineered in vitro from oral keratinocytes, fibroblasts, and fibrin, and can be used clinically to accelerate healing of oral soft tissue defects.


Asunto(s)
Mucosa Bucal/citología , Herida Quirúrgica/fisiopatología , Ingeniería de Tejidos , Trasplante de Tejidos , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Fibroblastos , Técnicas In Vitro , Queratinocitos , Modelos Animales , Ratas , Ratas Sprague-Dawley
15.
Oral Oncol ; 75: 81-88, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29224829

RESUMEN

OBJECTIVES: Wound healing is generally faster and associated with less scarring in the oral mucosa than in the skin. Although rarely studied, oral mucosa equivalents may contribute to rapid, scarless cutaneous wound healing. Therefore, we examined the potential utility of our newly developed oral mucosal cell sheet in skin wound healing. MATERIALS AND METHODS: Oral mucosa and skin samples were obtained from surgical patients and Sprague-Dawley rats. Keratinocytes and fibroblasts were primarily cultured for in vitro cell expansion. Mucosa and skin equivalents were produced with a mixture of cultured fibroblasts and autologous fibrin from plasma and seeding keratinocytes. Mucosal and skin cell sheets were transplanted in full-thickness excisional wounds of rat skin with control wounds. Gross, histological, and molecular characteristics of wound healing according to different postsurgical days were compared in control and cell sheet-covered wounds. RESULTS: Keratinocytes and fibroblasts derived from the oral mucosa were cultured faster than those derived from the skin. The in vitro-engineered oral mucosa and skin equivalents were successfully produced using complete autologous mucosa or skin and plasma fibrin, showing similarity to the histological characteristics of the skin or mucosa. In the in vivo rat model, the oral mucosal and skin cell sheet promoted wound healing with early wound closure and less scarring. The cell sheet-treated wounds showed lower TGF-ß1, α-smooth muscle actin, and fibronectin mRNA expression than the control wounds. CONCLUSIONS: The oral mucosal cell sheet demonstrated in vivo tissue plasticity through good adaptation to skin wounds, contributing to accelerated and scarless healing.


Asunto(s)
Plasticidad de la Célula , Cicatriz/prevención & control , Mucosa Bucal/citología , Piel/fisiopatología , Cicatrización de Heridas/fisiología , Actinas/metabolismo , Animales , Células Cultivadas , Fibroblastos/citología , Fibronectinas/genética , Humanos , Queratinocitos/citología , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Piel/metabolismo , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta1/genética
16.
Sci Rep ; 7(1): 10667, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878261

RESUMEN

Cell sheets with pre-vascularization have recently been developed but remain relatively untested in oral wound healing. Therefore, we examined the potential utility of our newly developed pre-vascularized mucosal cell sheets in oral wound healing. Mucosal keratinocytes, fibroblasts, and endothelial progenitor cells were primarily cultured for in vitro cell expansion from mucosa and blood of Sprague-Dawley rats. Mucosal cell sheets were generated using cultured keratinocytes and plasma fibrin (K sheet) or keratinocytes and a mixture of fibrin, fibroblasts, and endothelial cells (PV sheet). Autologous sheets were transplanted on deep wounds in the buccal region of rats. The gross and histological characteristics of wound healing were compared among control wound, K sheet, and PV sheet groups. We successfully cultured and expanded keratinocytes, fibroblasts, and endothelial progenitor cells in vitro for generating mucosal cell sheets with or without pre-vascularization. In the in vivo oral wound model, compared with the control wound, the PV sheet group exhibited rapid wound closure more prominently than the K sheet group. The histological healing in the PV sheet group was similar to that in rat normal buccal mucosa without fibrosis. The pre-vascularized mucosal cell sheet exhibited in vivo efficacy in oral wound healing by promoting accelerated healing.


Asunto(s)
Regeneración Tisular Dirigida , Mucosa Bucal/patología , Cicatrización de Heridas , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratas
17.
Oral Oncol ; 69: 84-91, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28559026

RESUMEN

OBJECTIVES: Severe oral mucosal and tissue defects can lead to pain, infection, and later undesirable healing of scarring and adhesion, resulting in a poor quality of life. In vitro-engineered oral mucosal equivalents for covering such defects are an alternative to avoiding the donor site morbidity of conventional skin or tissue grafts. We examined the efficacy of our newly developed three-dimensional mucosal cell sheets in an in vivo tongue wound model mimicking the surgical extirpation of tongue cancer. MATERIALS AND METHODS: Small oral mucosal and autologous fibrin samples were obtained from surgical patients and Sprague-Dawley rats. The fibrin was mixed with fibroblasts and seeded with keratinocytes that had been primarily cultured for in vitro cell expansion. The three-dimensional autologous cell sheets, cultured in air-lift interface inserts, were transplanted into deep wounds of the rat ventral tongue. Gross and microscopic findings of the postsurgical wounds were compared between wound control and cell sheet groups. RESULTS: The cell sheets were flexible, expandable, and easy to transfer, and had histological characteristics similar to that of the normal oral mucosa, with high p63 positivity. They promoted oral wound healing with earlier re-epithelialization and less fibrosis than that in the wound control. The cell sheet-healed tongue had similar histology to that of a normal tongue. CONCLUSIONS: Our engineered cell sheets have potential applicability for the rapid healing of oral mucosal and soft tissue defects, without scarring, adhesion, and functional deficits. CONDENSED ABSTRACT: The efficacy of in vitro-engineered mucosal equivalents, using completely autologous mucosa and plasma, was examined. Transplantation of the autologous cell sheets into deep wounds of the rat ventral tongue promoted oral wound healing with earlier re-epithelialization and less fibrosis than that in controls. Healed and normal tongues showed similar histology.


Asunto(s)
Mucosa Bucal/citología , Trasplante de Tejidos/métodos , Cicatrización de Heridas , Animales , Humanos , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Herida Quirúrgica , Lengua
18.
Redox Biol ; 13: 219-227, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28582730

RESUMEN

Reactivation of p53 and induction of tumor cell apoptosis (RITA) is a small molecule that blocks p53-MDM2 interaction, thereby reactivating p53 in tumors. RITA can induce exclusive apoptosis in cancer cells independently of the p53 pathway; however, the resistance of cancer cells remains a major drawback. Here, we found a novel resistance mechanism of RITA treatment and an effective combined treatment to overcome RITA resistance in head and neck cancer (HNC) cells. The effects of RITA and 3-methyladenine (3-MA) were tested in different HNC cell lines, including cisplatin-resistant and acquired RITA-resistant HNC cells. The effects of each drug alone and in combination were assessed by measuring cell viability, apoptosis, cell cycle, glutathione, reactive oxygen species, protein expression, genetic inhibition of p62 and Nrf2, and a mouse xenograft model of cisplatin-resistant HNC. RITA induced apoptosis of HNC cells at different levels without significantly inhibiting normal cell viability. Following RITA treatment, RITA-resistant HNC cells exhibited a sustained expression of other autophagy-related proteins, overexpressed p62, and displayed activation of the Keap1-Nrf2 antioxidant pathway. The autophagy inhibitor 3-MA sensitized resistant HNC cells to RITA treatment via the dual inhibition of molecules related to the autophagy and antioxidant systems. Silencing of the p62 gene augmented the combined effects. The effective antitumor activity of RITA plus 3-MA was also confirmed in vivo in mouse xenograft models transplanted with resistant HNC cells, showing increased oxidative stress and DNA damage. The results indicate that RITA plus 3-MA can help overcome RITA resistance in HNC cells. CONDENSED ABSTRACT: This study revealed a novel RITA resistant mechanism associated with the sustained induction of autophagy, p62 overexpression, and Keap1-Nrf2 antioxidant system activation. The combined treatment of RITA with the autophagy inhibitor 3-methyladenine overcomes RITA resistance via dual inhibition of autophagy and antioxidant systems in vitro and in vivo.


Asunto(s)
Adenina/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Resistencia a Antineoplásicos , Furanos/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Estrés Oxidativo , Adenina/administración & dosificación , Adenina/farmacología , Adenina/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas Relacionadas con la Autofagia/metabolismo , Línea Celular Tumoral , Furanos/administración & dosificación , Furanos/uso terapéutico , Glutatión/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
19.
Free Radic Biol Med ; 104: 1-9, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28057599

RESUMEN

The nonsteroidal anti-inflammatory drug aspirin and the multikinase inhibitor sorafenib have both shown experimental and clinical anticancer activities. The present study investigated whether aspirin and sorafenib synergize to potentiate cisplatin treatment in resistant head and neck cancer (HNC) cells. The effects of aspirin, sorafenib and cisplatin, and combinations thereof were assessed by measuring cell viability, death, glutathione (GSH) and reactive oxygen species (ROS) levels, protein and mRNA expression, genetic inhibition and overexpression of cystine-glutamate antiporter (xCT) and tumor xenograft mouse models. Even at low concentrations, aspirin plus sorafenib synergized to induce cell death of cisplatin-resistant HNC cells. The combination of aspirin and sorafenib induced xCT inhibition, GSH depletion, and ROS accumulation in cancer cells. Genetic and pharmacological inhibition of xCT potentiated the cytotoxic effects of aspirin plus sorafenib; this effect was diminished by xCT overexpression. Low-dose aspirin plus sorafenib enhanced the cytotoxicity of cisplatin in resistant HNC cells through xCT inhibition and oxidant and DNA damage. The in vivo effects of aspirin plus sorafenib on cisplatin therapy were also confirmed in resistant HNC xenograft models, in terms of growth inhibition, GSH depletion, and increased γH2AX formation and apoptosis in tumors. Aspirin and sorafenib synergize to potentiate the cytotoxicity of cisplatin in resistant HNC cells. This therapeutic strategy may help to eliminate resistant HNC.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Aspirina/administración & dosificación , Cisplatino/administración & dosificación , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Animales , Antioxidantes/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/biosíntesis , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Niacinamida/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Sorafenib , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Antioxid Redox Signal ; 27(2): 106-114, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-27733046

RESUMEN

AIMS: The glutathione (GSH), thioredoxin (Trx), and Nrf2 systems represent a major defense against reactive oxygen species (ROS), the cellular imbalance of which in cancer promotes growth and therapeutic resistance. This study investigated whether targeting the GSH, Trx, and Nrf2 antioxidant systems effectively eliminated head and neck cancer (HNC). RESULTS: At high concentrations, auranofin, but not buthionine sulfoximine (BSO) alone, decreased the viability of HNC, whereas even at low concentrations, auranofin plus BSO synergized to kill HNC cells. Dual silencing of the genes for GCLM and TrxR1 induced GSH depletion, Trx activity inhibition, and ROS accumulation, synergistically killing HNC cells. Inhibition of the GSH and Trx systems resulted in activation of the Nrf2-antioxidant response element (ARE) pathway, which may result in suboptimal GSH and Trx inhibition where HNC is resistant. Genetic inhibition of Nrf2 and/or HO-1 or trigonelline enhanced growth suppression, ROS accumulation, and cell death from GSH and Trx inhibition. The in vivo effects of GSH, Trx, and Nrf2 system inhibition were confirmed in a mouse HNC xenograft model by achieving growth inhibition >60% compared with those of control. Innovations: This study is the first to show that triple inhibition of GSH, Trx, and Nrf2 pathways could be an effective method to overcome the resistance of HNC. CONCLUSIONS: Inhibition of the Nrf2-ARE pathway in addition to dual inhibition of the GSH and Trx antioxidant systems can effectively eliminate resistant HNC. Antioxid. Redox Signal. 27, 106-114.


Asunto(s)
Auranofina/administración & dosificación , Butionina Sulfoximina/administración & dosificación , Glutatión/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Tiorredoxinas/metabolismo , Animales , Auranofina/farmacología , Butionina Sulfoximina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA