Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119752, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38776987

RESUMEN

Preserving a functional mitochondrial network is crucial for cellular well-being, considering the pivotal role of mitochondria in ensuring cellular survival, especially under stressful conditions. Mitophagy, the selective removal of damaged mitochondria through autophagy, plays a pivotal role in preserving cellular homeostasis by preventing the production of harmful reactive oxygen species from dysfunctional mitochondria. While the involvement of mitophagy in neurodegenerative diseases has been thoroughly investigated, it is becoming increasingly evident that mitophagy plays a significant role in cancer biology. Perturbations in mitophagy pathways lead to suboptimal mitochondrial quality control, catalyzing various aspects of carcinogenesis, including establishing metabolic plasticity, stemness, metabolic reconfiguration of cancer-associated fibroblasts, and immunomodulation. While mitophagy performs a delicate balancing act at the intersection of cell survival and cell death, mounting evidence indicates that, particularly in the context of stress responses induced by cancer therapy, it predominantly promotes cell survival. Here, we showcase an overview of the current understanding of the role of mitophagy in cancer biology and its potential as a target for cancer therapy. Gaining a more comprehensive insight into the interaction between cancer therapy and mitophagy has the potential to reveal novel targets and pathways, paving the way for enhanced treatment strategies for therapy-resistant tumors in the near future.

2.
bioRxiv ; 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38464251

RESUMEN

The androgen receptor (AR) is a ligand-responsive transcription factor that binds at enhancers to drive terminal differentiation of the prostatic luminal epithelia. By contrast, in tumors originating from these cells, AR chromatin occupancy is extensively reprogrammed to drive hyper-proliferative, metastatic, or therapy-resistant phenotypes, the molecular mechanisms of which remain poorly understood. Here, we show that the tumor-specific enhancer circuitry of AR is critically reliant on the activity of Nuclear Receptor Binding SET Domain Protein 2 (NSD2), a histone 3 lysine 36 di-methyltransferase. NSD2 expression is abnormally gained in prostate cancer cells and its functional inhibition impairs AR trans-activation potential through partial off-loading from over 40,000 genomic sites, which is greater than 65% of the AR tumor cistrome. The NSD2-dependent AR sites distinctly harbor a chimeric AR-half motif juxtaposed to a FOXA1 element. Similar chimeric motifs of AR are absent at the NSD2-independent AR enhancers and instead contain the canonical palindromic motifs. Meta-analyses of AR cistromes from patient tumors uncovered chimeric AR motifs to exclusively participate in tumor-specific enhancer circuitries, with a minimal role in the physiological activity of AR. Accordingly, NSD2 inactivation attenuated hallmark cancer phenotypes that were fully reinstated upon exogenous NSD2 re-expression. Inactivation of NSD2 also engendered increased dependency on its paralog NSD1, which independently maintained AR and MYC hyper-transcriptional programs in cancer cells. Concordantly, a dual NSD1/2 PROTAC degrader, called LLC0150, was preferentially cytotoxic in AR-dependent prostate cancer as well as NSD2-altered hematologic malignancies. Altogether, we identify NSD2 as a novel subunit of the AR neo-enhanceosome that wires prostate cancer gene expression programs, positioning NSD1/2 as viable paralog co-targets in advanced prostate cancer.

3.
Front Bioeng Biotechnol ; 11: 1213932, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37701494

RESUMEN

Targeted delivery of site-specific therapeutic agents is an effective strategy for osteoarthritis treatment. The lack of blood vessels in cartilage makes it difficult to deliver therapeutic agents like peptides to the defect area. Therefore, nucleus-targeting zwitterionic carbon nano-dots (CDs) have immense potential as a delivery vehicle for effective peptide delivery to the cytoplasm as well as nucleus. In the present study, nucleus-targeting zwitterionic CDs have been synthesized as delivery vehicle for peptides while also working as nano-agents towards optical monitoring of cartilage healing. The functional groups of zwitterion CDs were introduced by a single-step microwave assisted oxidation procedure followed by COL II peptide conjugation derived from Capra auricular cartilage through NHS/EDC coupling. The peptide-conjugated CDs (PCDs) allows cytoplasmic uptake within a short period of time (∼30 m) followed by translocation to nucleus after ∼24 h. Moreover, multicolor fluorescence of PCDs improves (blue, green, and read channel) its sensitivity as an optical code providing a compelling solution towards enhanced non-invasive tracking system with multifunctional properties. The PCDs-based delivery system developed in this study has exhibited superior ability to induce ex-vivo chondrogenic differentiation of ADMSCs as compared to bare CDs. For assessment of cartilage regeneration potential, pluronic F-127 based PCDs hydrogel was injected to rabbit auricular cartilage defects and potential healing was observed after 60 days. Therefore, the results confirm that PCDs could be an ideal alternate for multimodal therapeutic agents.

4.
Nat Commun ; 14(1): 5253, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37644036

RESUMEN

Loss of the tumor suppressive activity of the protein phosphatase 2A (PP2A) is associated with cancer, but the underlying molecular mechanisms are unclear. PP2A holoenzyme comprises a heterodimeric core, a scaffolding A subunit and a catalytic C subunit, and one of over 20 distinct substrate-directing regulatory B subunits. Methylation of the C subunit regulates PP2A heterotrimerization, affecting B subunit binding and substrate specificity. Here, we report that the leucine carboxy methyltransferase (LCMT1), which methylates the L309 residue of the C subunit, acts as a suppressor of androgen receptor (AR) addicted prostate cancer (PCa). Decreased methyl-PP2A-C levels in prostate tumors is associated with biochemical recurrence and metastasis. Silencing LCMT1 increases AR activity and promotes castration-resistant prostate cancer growth. LCMT1-dependent methyl-sensitive AB56αCme heterotrimers target AR and its critical coactivator MED1 for dephosphorylation, resulting in the eviction of the AR-MED1 complex from chromatin and loss of target gene expression. Mechanistically, LCMT1 is regulated by S6K1-mediated phosphorylation-induced degradation requiring the ß-TRCP, leading to acquired resistance to anti-androgens. Finally, feedforward stabilization of LCMT1 by small molecule activator of phosphatase (SMAP) results in attenuation of AR-signaling and tumor growth inhibition in anti-androgen refractory PCa. These findings highlight methyl-PP2A-C as a prognostic marker and that the loss of LCMT1 is a major determinant in AR-addicted PCa, suggesting therapeutic potential for AR degraders or PP2A modulators in prostate cancer treatment.


Asunto(s)
Neoplasias de la Próstata , Proteína Fosfatasa 2 , Humanos , Masculino , Antagonistas de Andrógenos , Leucina , Metiltransferasas , Próstata , Neoplasias de la Próstata/genética , Proteína Fosfatasa 2/genética
5.
Exp Cell Res ; 424(1): 113488, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36736226

RESUMEN

Glioma is difficult-to-treat because of its infiltrative nature and the presence of the blood-brain barrier. Temozolomide is the only FDA-approved drug for its management. Therefore, finding a novel chemotherapeutic agent for glioma is of utmost importance. Magnolol, a neolignan, has been known for its apoptotic role in glioma. In this work, we have explored a novel anti-glioma mechanism of Magnolol associated with its role in autophagy modulation. We found increased expression levels of Beclin-1, Atg5-Atg12, and LC3-II and lower p62 expression in Magnolol-treated glioma cells. PI3K/AKT/mTOR pathway proteins were also downregulated in Magnolol-treated glioma cells. Next, we treated the glioma cells with Insulin, a stimulator of PI3K/AKT/mTOR signaling, to confirm that Magnolol induced autophagy by inhibiting this pathway. Insulin reversed the effect on Magnolol-mediated autophagy induction. We also established the same in in vivo glioma model where Magnolol showed an anti-glioma effect by inducing autophagy. To confirm the cytotoxic effect of Magnolol-induced autophagy, we used Chloroquine, a late-stage autophagy inhibitor. Chloroquine efficiently reversed the anti-glioma effects of Magnolol both in vitro and in vivo. Our study revealed the cytotoxic effect of Magnolol-induced autophagy in glioma, which was not previously reported. Additionally, Magnolol showed no toxicity in non-cancerous cell lines as well as rat organs. Thus, we concluded that Magnolol is an excellent candidate for developing new therapeutic strategies for glioma management.


Asunto(s)
Antineoplásicos , Glioma , Insulinas , Lignanos , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Antineoplásicos/farmacología , Lignanos/farmacología , Lignanos/uso terapéutico , Glioma/tratamiento farmacológico , Glioma/metabolismo , Autofagia , Cloroquina/farmacología , Cloroquina/uso terapéutico , Insulinas/farmacología , Insulinas/uso terapéutico , Línea Celular Tumoral , Apoptosis
6.
Cell Rep ; 39(11): 110971, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705030

RESUMEN

Ewing sarcoma (EwS) is a highly aggressive tumor of bone and soft tissues that mostly affects children and adolescents. The pathognomonic oncofusion EWSR1::FLI1 transcription factor drives EwS by orchestrating an oncogenic transcription program through de novo enhancers. By integrative analysis of thousands of transcriptomes representing pan-cancer cell lines, primary cancers, metastasis, and normal tissues, we identify a 32-gene signature (ESS32 [Ewing Sarcoma Specific 32]) that stratifies EwS from pan-cancer. Among the ESS32, LOXHD1, encoding a stereociliary protein, is the most highly expressed gene through an alternative transcription start site. Deletion or silencing of EWSR1::FLI1 bound upstream de novo enhancer results in loss of the LOXHD1 short isoform, altering EWSR1::FLI1 and HIF1α pathway genes and resulting in decreased proliferation/invasion of EwS cells. These observations implicate LOXHD1 as a biomarker and a determinant of EwS metastasis and suggest new avenues for developing LOXHD1-targeted drugs or cellular therapies for this deadly disease.


Asunto(s)
Proteínas Portadoras , Elementos de Facilitación Genéticos , Proteínas de Fusión Oncogénica , Sarcoma de Ewing , Adolescente , Proteínas Portadoras/genética , Línea Celular Tumoral , Niño , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología
7.
Exp Cell Res ; 417(1): 113195, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35561786

RESUMEN

The Transforming growth factor-ß1 (TGF- ß1) in the tumor microenvironment (TME) is the major cytokine that acts as a mediator of tumor-stroma crosstalk, which in fact has a dual role in either promoting or suppressing tumor development. The cancer-associated fibroblasts (CAFs) are the major cell types in the TME, and the interaction with most of the epithelial cancers is the prime reason for cancer survival. However, the molecular mechanisms, associated with the TGF- ß1 induced tumor promotion through tumor-CAF crosstalk are not well understood. In the Reverse Warburg effect, CAFs feed the adjacent cancer cells by lactate produced during the aerobic glycolysis. We hypothesized that the monocarboxylate transporter, MCT4 which is implicated in lactate efflux from the CAFs, must be overexpressed in the CAFs. Contextually, to explore the role of TGF- ß1 in the hypoxia-induced autophagy in CAFs, we treated CoCl2 and external TGF- ß1 to the human dermal fibroblasts and L929 murine fibroblasts. We demonstrated that hypoxia accelerated the TGF- ß1 signaling and subsequent transformation of normal fibroblasts to CAFs. Moreover, we elucidated that synergistic induction of autophagy by hypoxia and TGF- ß1 upregulate the aerobic glycolysis and MCT4 expression in CAFs. Furthermore, we showed a positive correlation between glucose consumption and MCT4 expression in the CAFs. Autophagy was also found to be involved in the EMT in hypoxic CAFs. Collectively, these findings reveal the unappreciated role of autophagy in TME, which enhances the CAF transformation and that promotes tumor migration and metastasis via the reverse Warburg effect.


Asunto(s)
Autofagia , Fibroblastos Asociados al Cáncer , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Neoplasias , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Hipoxia/metabolismo , Ácido Láctico/metabolismo , Ratones , Neoplasias/patología , Microambiente Tumoral , Regulación hacia Arriba
9.
Comput Biol Med ; 141: 105052, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34836625

RESUMEN

BACKGROUND: Aloe vera extract and its bioactive compounds possess anti-proliferative properties against cancer cells. However, no detailed molecular mechanism of action studies has been reported. We have now employed a computational approach to scrutinize the molecular mechanism of lead bioactive compounds from Aloe vera that potentially inhibit DNA synthesis. METHODS: Initially, the anti-proliferative activity of Aloe vera extract was examined in human breast cancer cells (in vitro/in vivo). Later on, computational screening of bioactive compounds from Aloe vera targeting DNA was performed by molecular docking and molecular dynamics simulation. RESULTS: In-vitro and in-vivo studies confirm that Aloe vera extract effectively suppresses the growth of breast cancer cells without significant cytotoxicity towards non-cancerous normal immortal cells. Computational screening predicts that growth suppression may be due to the presence of DNA intercalating bioactive compounds (riboflavin, daidzin, aloin, etc.) contained in Aloe vera. MM/PBSA calculation showed that riboflavin has a higher binding affinity at the DNA binding sites compared to standard drug daunorubicin. CONCLUSIONS: These observations support the hypothesis that riboflavin may be exploited as an anti-proliferative DNA intercalating agent to prevent cancer and is worthy of testing for the management of cancer by performing more extensive pre-clinical and if validated clinical trials.


Asunto(s)
Aloe , Neoplasias , Aloe/química , ADN , Humanos , Simulación del Acoplamiento Molecular , Extractos Vegetales/farmacología
10.
Biomed Mater ; 16(4)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33621207

RESUMEN

The use of medicinal plants is as ancient as human civilization. The development of phytochemistry and pharmacology facilitates the identification of natural bioactive compounds and their mechanisms of action, including against cancer. The efficacy and the safety of a bioactive compound depend on its optimal delivery to the target site. Most natural bioactive compounds (phenols, flavonoids, tannins, etc) are unable to reach their target sites due to their low water solubility, less cellular absorption, and high molecular weight, leading to their failure into clinical translation. Therefore, many scientific studies are going on to overcome the drawbacks of natural products for clinical applications. Several studies in India, as well as worldwide, have proposed the development of natural products-based nanoformulations to increase their efficacy and safety profile for cancer therapy by improving the delivery of natural bioactive compounds to their target site. Therefore, we are trying to discuss the development of natural products-based nanoformulations in India to improve the efficacy and safety of natural bioactive compounds against cancer.


Asunto(s)
Productos Biológicos , Neoplasias , Plantas Medicinales , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Flavonoides , Humanos , Neoplasias/tratamiento farmacológico , Solubilidad
11.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188416, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32822826

RESUMEN

One of the undeniable issues with cancer eradication is the evolution of chemoresistance in due course of treatment, and the mechanisms of chemoresistance have been the subject of extensive research for several years. The efficacy of chemotherapy is hindered by cancer epithelium, mostly in a cell-autonomous mechanism. However, recently the valid experimental evidence showed that the surrounding tumor microenvironment (TME) is equivalently responsible for the induction of chemoresistance. Of the verities of cells in the tumor microenvironment, cancer-associated fibroblasts (CAFs) are the major cellular component of TME and act as a key regulator in the acquisition of cancer chemoresistance by providing a protective niche to the cancer cells against the anti-cancer drugs. Moreover, the symbiotic relationship between the tumor and CAFs to obtain key resources such as growth factors and nutrients for optimal tumor growth and proliferation favors the chemoresistance phenotype. Here, in this review, we provide an up-to-date overview of our knowledge of the role of the CAFs in inducing chemoresistance and tumor progression. We also further delineated the emerging events leading to the CAF origins and activation of normal fibroblasts to CAFs. Along with this, we also discuss the novel area of research confined to the CAF targeted therapies of cancer. The identification of CAF-specific markers may allow unveiling new targets and avenues for blunting or reverting the detrimental pro-tumorigenic potential of CAFs in the foreseeable future.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Resistencia a Antineoplásicos , Neoplasias/metabolismo , Biomarcadores/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/patología , Nutrientes/metabolismo , Microambiente Tumoral
12.
ACS Appl Bio Mater ; 3(9): 6284-6296, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-35021759

RESUMEN

In recent years, graphene-based materials complexed with drugs have been developed for application in cancer therapy, aimed at gaining synergistic effect. Here, we have prepared graphene oxide (GO) and graphene quantum dots (GQDs) with curcumin (Cur) in three different ratios (1:1, 1:3, and 1:5 w/v). We showed a successful complexation of GO and GQDs with Cur through various spectroscopy and microscopy techniques. The optical density of the complex through UV-vis spectroscopy showed less than 10% (for GQDs-Cur) and less than 20% (for GO-Cur) aggregation in 48 h, which confirms the stability of the complex. The UV-vis result estimates the loading efficiency of Cur to be 80 ± 1 and 83 ± 1% for GO-Cur and GQDs-Cur respectively. We tested the complexes GO-Cur and GQDs-Cur in different ratios as an anticancer drug against human breast cancer cell lines MCF-7 and MDA-MB-468 through the MTT assay. Following 48 h of incubation with the cell lines, a cell viability of more than 75% was observed in the case of GQDs & GO, while it was 40% in the case of Cur at a concentration of 100 µg/mL. The 1:1, 1:3, and 1:5 ratios of complexes enforced cell death ∼60, ∼80, and ∼95% at 100 µg/mL after 48 h of treatment, respectively. The optical images of cancerous cells treated with GO, GQDs, Cur, GO-Cur, and GQDs-Cur, at three different time intervals (0, 24, and 48 h), corroborated well with the results from the MTT assay in terms of the percentage of dead cells. The fluorescence images show a successful delivery of Cur drug inside the cancerous cell. The possible mechanism of killing of the cancerous cell with the complexes GO-Cur and GQDs-Cur is discussed. Moreover, this study opens a window to determine the mechanism of killing the cancerous cell.

13.
Semin Cancer Biol ; 66: 59-74, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31430557

RESUMEN

Autophagy is an evolutionary conserved catabolic process that regulates the cellular homeostasis by targeting damaged cellular contents and organelles for lysosomal degradation and sustains genomic integrity, cellular metabolism, and cell survival during diverse stress and adverse conditions. Recently, the role of autophagy is extremely debated in the regulation of cancer initiation and progression. Although autophagy has a dichotomous role in the regulation of cancer, growing numbers of studies largely indicate the pro-survival role of autophagy in cancer progression and metastasis. In this review, we discuss the detailed mechanisms of autophagy, the role of pro-survival autophagy that positively drives several classical as well as emerging hallmarks of cancer for tumorigenic progression, and also we address various autophagy inhibitors that could be harnessed against pro-survival autophagy for effective cancer therapeutics. Finally, we highlight some outstanding problems that need to be deciphered extensively in the future to unravel the role of autophagy in tumor progression.


Asunto(s)
Autofagia/fisiología , Neoplasias/patología , Animales , Carcinogénesis/patología , Supervivencia Celular/fisiología , Progresión de la Enfermedad , Humanos
14.
Pharmacol Res ; 148: 104416, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31470079

RESUMEN

Aloe vera (Aloe barbadensis Miller) is a perennial succulent medicinal plant. It has been used as a traditional or folk medicine for thousands of years and claimed that it possesses wound and burn healing activities, and anti-inflammatory as well as immunomodulatory effects. In recent years, the use of Aloe vera has been growing as a dietary supplement. The pre-clinical studies over the last couple of decades uncover the potential therapeutic activities of Aloe vera and its bioactive compounds, especially against neoplastic disease. Such investigations indicate the possible preventive as well as therapeutic effects of Aloe vera against cancer. Here, we discuss the crucial bioactive compounds of Aloe vera that have been harnessed against cancer and also address several mechanisms of action of these lead bioactive compounds compared to other standard drugs involved in cancer prevention and treatment.


Asunto(s)
Aloe/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Quemaduras/tratamiento farmacológico , Humanos , Plantas Medicinales/química , Cicatrización de Heridas/efectos de los fármacos
15.
Biochim Biophys Acta Mol Cell Res ; 1866(6): 1004-1018, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30878502

RESUMEN

Estrogen receptor (ER) antagonist, tamoxifen has been universally used for the treatment of the ER-positive breast cancer; however, the inevitable emergence of resistance to tamoxifen obstructs the successful treatment of this cancer. So, there is an immediate requirement for the search of a novel therapeutic target for treatment of this cancer. Acquired tamoxifen-resistant breast cancer cell lines MCF-7 (MCF-7/TAM-R) and T47D (T47D/TAM-R) showed higher apoptotic resistance accompanied by induction of pro-survival autophagy compared to their parental cells. Besides, tamoxifen resistance was associated with reduced production of ATP and with overexpression of glycolytic pathways, leading to induced autophagy to meet the energy demand. Further, our study revealed that LDHA; one of the key molecules of glycolysis in association with Beclin-1 induced pro-survival autophagy in tamoxifen-resistant breast cancer. Mechanistically, pharmacological and genetic inhibition of LDHA reduced the pro-survival autophagy, with the restoration of apoptosis and reverting back the EMT like phenomena noticed in tamoxifen-resistant breast cancer. In total, targeting LDHA opened a novel strategy to interrupt autophagy and tamoxifen resistance in breast cancer.


Asunto(s)
Beclina-1/genética , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos , L-Lactato Deshidrogenasa/genética , Tamoxifeno/farmacología , Autofagia , Línea Celular Tumoral , Supervivencia Celular , Transición Epitelial-Mesenquimal , Femenino , Glucólisis , Humanos , Células MCF-7 , Complejo de la Endopetidasa Proteasomal/metabolismo
16.
Mol Pharm ; 16(1): 24-40, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30513203

RESUMEN

The effective delivery of target-specific siRNA to the brain by exploiting the exosomes derived from dendritic cells renders the paradigm shift for the prospective use of nanosized exosomes as a delivery system. Although the in vivo targeting strategies by other nanovesicles like liposomes exist, still this novel exosome-based delivery approach holds an inclusive dominance of in vivo security and reduced immunogenicity. Achieving promising exosome-based delivery strategies warrants more desirable exploration of their biology. Over the years, the invention of novel production, characterization, targeting strategies, and cargo loading techniques of exosome improved its ability to reach clinics. Essentially, exosome-based delivery of therapeutics assures to conquer the major hurdles, like delivery of cargos across impermeable biological barriers, like the blood-brain barrier, biocompatibility, increased solubility, metabolic stability, improved circulation time, target specific delivery, and pharmacokinetics, and thereby enhanced the efficacy of loaded therapeutic agents. In this article, we cover the current status of exosome as a delivery vehicle for therapeutics and the challenges that need to be overcome, and we also discuss future perspectives of this exciting field of research to transform it from bench to clinical reality.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Exosomas/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Humanos , Modelos Teóricos , ARN Interferente Pequeño/metabolismo
17.
Oncogene ; 37(33): 4546-4561, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29743594

RESUMEN

Although there is a strong correlation between multinucleated cells (MNCs) and cancer chemo-resistance in variety of cancers, our understanding of how multinucleated cells modulate the tumor micro-environment is limited. We captured multinucleated cells from triple-negative chemo-resistant breast cancers cells in a time frame, where they do not proliferate but rather significantly regulate their micro-environment. We show that oxidatively stressed MNCs induce chemo-resistance in vitro and in vivo by secreting VEGF and MIF. These factors act through the RAS/MAPK pathway to induce chemo-resistance by upregulating anti-apoptotic proteins. In MNCs, elevated reactive oxygen species (ROS) stabilizes HIF-1α contributing to increase production of VEGF and MIF. Together the data indicate, that the ROS-HIF-1α signaling axis is very crucial in regulation of chemo-resistance by MNCs. Targeting ROS-HIF-1α in future may help to abrogate drug resistance in breast cancer.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Cancer Metastasis Rev ; 37(4): 749-766, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29536228

RESUMEN

Resistance to therapy is one of the prime causes for treatment failure in cancer and recurrent disease. In recent years, autophagy has emerged as an important cell survival mechanism in response to different stress conditions that are associated with cancer treatment and aging. Autophagy is an evolutionary conserved catabolic process through which damaged cellular contents are degraded after uptake into autophagosomes that subsequently fuse with lysosomes for cargo degradation, thereby alleviating stress. In addition, autophagy serves to maintain cellular homeostasis by enriching nutrient pools. Although autophagy can act as a double-edged sword at the interface of cell survival and cell death, increasing evidence suggest that in the context of cancer therapy-induced stress responses, it predominantly functions as a cell survival mechanism. Here, we provide an up-to-date overview on our current knowledge of the role of pro-survival autophagy in cancer therapy at the preclinical and clinical stages and delineate the molecular mechanisms of autophagy regulation in response to therapy-related stress conditions. A better understanding of the interplay of cancer therapy and autophagy may allow to unveil new targets and avenues for an improved treatment of therapy-resistant tumors in the foreseeable future.


Asunto(s)
Neoplasias/patología , Neoplasias/terapia , Autofagia/fisiología , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia
19.
Neoplasia ; 20(3): 263-279, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29462756

RESUMEN

Target-specific treatment modalities are currently not available for triple-negative breast cancer (TNBC), and acquired chemotherapy resistance is a primary obstacle for the treatment of these tumors. Here we employed derivatives of BT-549 and MDA-MB-468 TNBC cell lines that were adapted to grow in the presence of either 5-Fluorouracil, Doxorubicin or Docetaxel in an aim to identify molecular pathways involved in the adaptation to drug-induced cell killing. All six drug-adapted BT-549 and MDA-MB-468 cell lines displayed cross resistance to chemotherapy and decreased apoptosis sensitivity. Expression of the anti-apoptotic co-chaperone BAG3 was notably enhanced in two thirds (4/6) of the six resistant lines simultaneously with higher expression of HSP70 in comparison to parental controls. Doxorubicin-resistant BT-549 (BT-549rDOX20) and 5-Fluorouracil-resistant MDA-MB-468 (MDA-MB-468r5-FU2000) cells were chosen for further analysis with the autophagy inhibitor Bafilomycin A1 and lentiviral depletion of ATG5, indicating that enhanced cytoprotective autophagy partially contributes to increased drug resistance and cell survival. Stable lentiviral BAG3 depletion was associated with a robust down-regulation of Mcl-1, Bcl-2 and Bcl-xL, restoration of drug-induced apoptosis and reduced cell adhesion in these cells, and these death-sensitizing effects could be mimicked with the BAG3/Hsp70 interaction inhibitor YM-1 and by KRIBB11, a selective transcriptional inhibitor of HSF-1. Furthermore, BAG3 depletion was able to revert the EMT-like transcriptional changes observed in BT-549rDOX20 and MDA-MB-468r5-FU2000 cells. In summary, genetic and pharmacological interference with BAG3 is capable to resensitize TNBC cells to treatment, underscoring its relevance for cell death resistance and as a target to overcome therapy resistance of breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Autofagia/genética , Resistencia a Antineoplásicos/genética , Neoplasias de la Mama Triple Negativas/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
20.
Cell Prolif ; 51(1)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29171106

RESUMEN

OBJECTIVE: We inspected the relevance of CD44, ABCB1 and ADAM17 in OSCC stemness and deciphered the role of autophagy/mitophagy in regulating stemness and chemoresistance. MATERIAL AND METHODS: A retrospective analysis of CD44, ABCB1 and ADAM17 with respect to the various clinico-pathological factors and their correlation was analysed in sixty OSCC samples. Furthermore, the stemness and chemoresistance were studied in resistant oral cancer cells using sphere formation assay, flow cytometry and florescence microscopy. The role of autophagy/mitophagy was investigated by transient transfection of siATG14, GFP-LC3, tF-LC3, mKeima-Red-Mito7 and Western blot analysis of autophagic and mitochondrial proteins. RESULTS: In OSCC, high CD44, ABCB1 and ADAM17 expressions were correlated with higher tumour grades and poor differentiation and show significant correlation in their co-expression. In vitro and OSCC tissue double labelling confirmed that CD44+ cells co-expresses ABCB1 and ADAM17. Further, cisplatin (CDDP)-resistant FaDu cells displayed stem-like features and higher CD44, ABCB1 and ADAM17 expression. Higher autophagic flux and mitophagy were observed in resistant FaDu cells as compared to parental cells, and inhibition of autophagy led to the decrease in stemness, restoration of mitochondrial proteins and reduced expression of CD44, ABCB1 and ADAM17. CONCLUSION: The CD44+ /ABCB1+ /ADAM17+ expression in OSCC is associated with stemness and chemoresistance. Further, this study highlights the involvement of mitophagy in chemoresistance and autophagic regulation of stemness in OSCC.


Asunto(s)
Proteína ADAM17/metabolismo , Autofagia/fisiología , Resistencia a Antineoplásicos , Receptores de Hialuranos/metabolismo , Neoplasias de la Boca/metabolismo , Células Madre Neoplásicas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...