Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci ; 341: 122506, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38373620

RESUMEN

Most human malignancies are attributed to exposure to infectious organisms such as viruses. Certain infections that can induce cancer can evade the immune system, leading to persistent inflammation that facilitates uncontrolled cell growth. Moreover, these pathogens can increase the likelihood of oncogenic transformation, leading to cancer development. Despite significant advancements in medicine, oncological research continues to seek innovative treatment techniques in light of the constraints imposed by traditional therapeutic agents. Virus-based therapy is a novel treatment method that has garnered significant interest due to its broad range of applications. Virotherapy employs oncolytic viruses that are genetically modified to target tumor cells specifically, undergo replication inside them and destroy the malignant cells. Additionally, this therapeutic approach elicits an anticancer response by boosting the patient's immune system. In addition, viruses are commonly employed as targeted delivery vectors for the precise transportation of various genes, medicinal compounds and immune-stimulating substances. Furthermore, virotherapy offers more excellent anticancer activity in combination with established treatment modalities such as immune therapy, chemotherapy and radiation therapy. This review presents a concise overview of the roles played by infectious agents, such as viruses in cancer progression. In addition, we have thoroughly summarized the advancements in utilizing viruses for their oncolytic properties in conjunction with established cancer treatment modalities such as chemotherapy, radiation and immunotherapy.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Viroterapia Oncolítica/métodos , Neoplasias/terapia , Neoplasias/patología , Inmunoterapia/métodos
2.
Med Oncol ; 41(2): 49, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184505

RESUMEN

Recently, we reported that a combination of a natural, bioactive compound Resveratrol (RES) and a PARP inhibitor Olaparib (OLA) deregulated the homologous recombination (HR) pathway, and enhanced apoptosis in BRCA1-wild-type, HR-proficient breast cancer cells. Upon DNA damage, chromatin relaxation takes place, which allows the DNA repair proteins to access the DNA lesion. But whether chromatin remodeling has any role in RES + OLA-mediated HR inhibition is not known. By using in vitro and ex vivo model systems of breast cancer, we have investigated whether RES + OLA inhibits chromatin relaxation and thereby blocks the HR pathway. It was found that RES + OLA inhibited PARP1 activity, terminated PARP1-BRCA1 interaction, and deregulated the HR pathway only in the chromatin fraction of MCF-7 cells. DR-GFP reporter plasmid-based HR assay demonstrated marked reduction in HR efficiency in I-SceI endonuclease-transfected cells treated with OLA. RES + OLA efficiently trapped PARP1 at the DNA damage site in the chromatin of MCF-7 cells. Unaltered expressions of HR proteins were found in the chromatin of PARP1-silenced MCF-7 cells, which confirmed that RES + OLA-mediated DNA damage response was PARP1-dependent. Histone Acetyltransferase (HAT) activity and histone H4 acetylation assays showed reduction in HAT activity and H4 acetylation in RES + OLA-treated chromatin fraction of cells. Western blot analysis showed that the HAT enzyme TIP60, P400 and acetylated H4 were downregulated after RES + OLA exposure. In the co-immunoprecipitation assay, it was observed that RES + OLA caused abolition of PARP1-TIP60-BRCA1 interaction, which suggested the PARP1-dependent TIP60-BRCA1 association. Unaltered expressions of PAR, BRCA1, P400, and acetylated H4 in the chromatin of TIP60-silenced MCF-7 cells further confirmed the role of TIP60 in PARP1-mediated HR activation in the chromatin. Similar results were obtained in ex vivo patient-derived primary breast cancer cells. Thus, the present study revealed that RES + OLA treatment inhibited PARP1 activity in the chromatin, and blocked TIP60-mediated chromatin relaxation, which, in turn, affected PARP1-dependent TIP60-BRCA1 association, resulting in deregulation of HR pathway in breast cancer cells.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Cromatina , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Resveratrol/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Reparación del ADN por Recombinación
3.
J Nutr Biochem ; 125: 109568, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38185347

RESUMEN

Tumor associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) in the tumor microenvironment secrete several cytokines, which involved in tumor initiation, progression, metastatic outgrowth and angiogenesis. However, the association between TAMs and CAFs in the context of tumor development remain unclear. Here, we studied the relationship between TAMs and CAFs along with the involvement of cytokines in the production of cancer-stem-like-cells (CSCs) in oral cancer cells and explored the potential anticancer effects of Nano-formulated Resveratrol (Res-NP) using an activated macrophage-M1 (AM-M1) and activated fibroblast cells as the model system. IL-6 secretion was found to be enhanced in the conditioned-medium (CM) when AM-M1 cells + CAFs-like cells were cocultured together. CSCs-enriched population was developed after addition of CM of AM-M1 +CAFs in H-357 cells and patient-derived-primary-oral-cancer cells. AM-M1 cells+ CAFs-like cells secreted IL-6 enhanced CSCs growth, proliferation, metastasis, and angiogenesis. IL-6 was found to promote PD-L1 expression in CSCs-enriched cells via JAK2/STAT3 pathway, as evident from the enhanced expression of p-JAK2 and p-STAT3. Nevertheless, Res-NP inhibited CSCs proliferation and reduced the expression of metastatic and angiogenic markers, in ovo blood vascularization, NO production and MMPs expression. Res-NP delinked the association between AM-M1 and CAFs by blocking IL-6 production and also disrupted the potential connection between IL-6 and PD-L1 with considerable decrease in p-JAK2 and p-STAT3 expressions. IL-6 depletion inhibited stemness and angiogenesis in oral CSCs by downregulating PD-L1 via JAK2/STAT3 cascade. Similar observations were also observed in Res-NP treated xenograft mice. Thus, data demonstrate that CSCs growth is dependent on IL-6/PD-L1 axis. Res-NP deregulates the association between AM-M1 and CAFs along-with attenuates carcinogenesis in in vitro, in ovo, ex vivo and in vivo model systems by inhibiting PD-L1 via IL-6/JAK2/STAT3 axis.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Boca , Humanos , Animales , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Interleucina-6/metabolismo , Resveratrol/farmacología , Macrófagos Asociados a Tumores/metabolismo , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Neoplasias de la Boca/metabolismo , Microambiente Tumoral , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo
4.
Nanomedicine (Lond) ; 19(7): 581-596, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38293827

RESUMEN

Aim: This study aimed to determine if quinacrine-gold hybrid nanoparticles (QAuNPs) + near-infrared (NIR) deregulate HSP-70/P300 complex-mediated H3K14 acetylation in estrogen receptor/progesterone receptor (ER/PR+) breast cancer stem cells (CSCs). Materials & methods: Various cells and mouse-based systems were used as models. Results: QAuNP + NIR treatment reduced the nuclear translocation of HSP-70, affected the histone acetyltransferase activity of P300 and specifically decreased H3K14 acetylation in ER/PR+ breast CSCs. Finally, HSP-70 knockdown showed a reduction in P300 histone acetyltransferase activity, decreased H3K14 acetylation and inhibited activation of the TGF-ß gene. Conclusion: This study revealed that QAuNP + NIR irradiation inhibits oncogenic activation of the TGF-ß gene by decreasing H3K14 acetylation mediated through the HSP-70/P300 nuclear complex in ER/PR+ breast CSCs.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Ratones , Acetilación , Oro , Histona Acetiltransferasas , Células Madre Neoplásicas , Quinacrina/farmacología , Factor de Crecimiento Transformador beta , Humanos , Femenino
5.
Med Oncol ; 40(12): 351, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37940725

RESUMEN

The presence of cancer stem cells (CSCs) in the tumor microenvironment (TME) is majorly responsible for the development and recurrence of cancer. Earlier reports suggested that upon DNA damage, poly-(ADP-ribose) polymerase-1 (PARP-1) helps in chromatin modulation and DNA repair process, thereby promoting CSC survival. But whether a combination of DNA damaging agents along with PARP inhibitors can modulate chromatin assembly, inhibit DNA repair processes, and subsequently target CSCs is not known. Hence, we have investigated the effect of nontoxic bioactive compound quinacrine (QC) and a potent PARP inhibitor Talazoparib in patient-derived oral mucosa CSCs (OM-CSCs) and in vivo xenograft mice preclinical model systems. Data showed that QC + Talazoparib inhibited the PARP-1-mediated chromatin remodelers' recruitment and deregulated HAT activity of GCN5 (general control nonderepressible-5) and P300 at DNA damage site, thereby preventing the access of repair proteins to the damaged DNA. Additionally, this combination treatment inhibited topoisomerase activity, induced topological stress, and induced apoptosis in OM-CSCs. Similar results were observed in an in vivo xenograft mice model system. Collectively, the data suggested that QC + Talazoparib treatment inhibited BER pathway, induced genomic instability and triggered apoptosis in OM-CSCs through the deregulation of PARP-1-mediated chromatin remodelers (GCN5 and P300) activity. Schematic representation of QC + Talazoparib-induced apoptosis in oral mucosa CSCs. (1) Induction of DNA damage takes place after QC treatment (2) PARP1-mediated PARylation at the site of DNA damage, which recruits multiple chromatin remodelers (3) Acetylation at the histone tails relax the structure of chromatin and recruits the BER pathway proteins at the site of DNA damage. (4) BER pathway activated at the site of DNA damage. (5) CSCs survive after successful repair of DNA damage. (6) Treatment of QC-treated CSCs with PARP inhibitor Talazoparib (7) Inhibition of PARylation results in failure of chromatin remodelers to interact with PARP1. (8) Inhibition of acetylation status leads to chromatin compaction. (9) BER pathway proteins are not recruited at the site of DNA damage, resulting in inhibition of BER pathway and accumulation of unrepaired DNA damage, leading to apoptosis and cell death.


Asunto(s)
Antineoplásicos , Quinacrina , Humanos , Animales , Ratones , Quinacrina/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Mucosa Bucal , Reparación del ADN , Antineoplásicos/farmacología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Daño del ADN , Cromatina , ADN/farmacología , Apoptosis
6.
Expert Opin Ther Targets ; 27(10): 999-1015, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37787493

RESUMEN

OBJECTIVE: Sensitization of mismatch repair (MMR)-deficient colorectal cancer (CRC) cells by 5-Fluorouracil (5-FU) is well-documented. But not much is known about the treatment of MMR-proficient CRC cancer stem cells (CRC-CSCs). Here, we investigated whether a PARP inhibitor (ABT-888) can enhance the 5-FU-mediated apoptosis in CRC-CSCs through MMR pathway inhibition. METHODS: The anti-cancer action of 5-FU+ABT-888 combination in CRC-CSCs has been studied by using in vitro, ex vivo, and in vivo preclinical model systems. RESULTS: 5-FU caused DNA damage in CRC-CSCs, and ABT-888 enhanced the accumulation of DNA mismatches by downregulating the MMR pathway, triggering S-phase arrest, and finally apoptosis and cell death in 5-FU-pre-treated MMR-proficient-CRC-CSCs at much lower concentrations than their individual treatments. After 5-FU treatment, PARylated-PARP1 activated MMR pathway by interacting with MSH6. But, upon ABT-888 treatment in 5-FU-pre-exposed CSCs, PARylation was inhibited, as a result of which PARP1 could not interact with MSH6, and other MMR proteins were downregulated. The role of MSH6 in PARP1-mediated MMR activation, was confirmed by silencing MSH6 gene, which resulted in MMR pathway shutdown. Similar results were obtained in ex vivo and in vivo model systems. CONCLUSIONS: 5-FU+ABT-888 combination enhanced CRC-CSCs death by increasing DNA damage accumulation and simultaneously inhibiting the MMR pathway in MMR-proficient cells. But this study does not discuss whether the combination treatment will increase the sensitivity of MMR-deficient CSCs, for which further research will be performed in the future.


5-FU is a well-known drug commonly used to treat colorectal cancer and it causes DNA damage inside the cancer cells. The limitation of 5-FU treatment is the development of chemoresistance due to the high DNA repair capacity of cancer stem cells present in the tumor microenvironment. In this study, a novel chemotherapeutic approach has been developed to target colorectal cancer stem cells by using a combination of 5-FU and a PARP1 inhibitor (ABT-888). Here, 5-FU caused DNA damage and ABT-888 enhanced the accumulation of the DNA lesions by inhibiting the MMR repair pathway in 5-FU-pre-treated MMR-proficient-CRC-CSCs. This resulted in S-phase arrest, induction of apoptosis, and finally CSCs death.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Humanos , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN , Células Madre Neoplásicas
7.
Phytomedicine ; 117: 154914, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37321076

RESUMEN

BACKGROUND: Breast cancer stem cells (BCSCs) have a critical role in progression of breast cancer by inducing angiogenesis. Several therapeutic strategies have been designed for the treatment of breast cancer by specifically preventing angiogenesis. But there is a dearth of study regarding the treatment procedure which can specifically target and kill the BCSCs and cause lesser harm to healthy cells of the body. A plant-based bioactive compound Quinacrine (QC) specifically kills cancer stem cells (CSCs) without harming healthy cells and also inhibits cancer angiogenesis but the detailed mechanistic study of its anti-CSCs and anti-angiogenic activity is yet to explore. HYPOTHESIS: Earlier report showed that both cMET and ABCG2 play an essential role in cancer angiogenesis. Both are present on the cell surface of CSCs and share an identical ATP-binding domain. Interestingly, QC a plant based and bioactive compound which was found to inhibit the function of CSCs marker cMET and ABCG2. These relevant evidence led us to hypothesize that cMET and ABCG2 may interact with each other and induce the production of angiogenic factors, resulting in activation of cancer angiogenesis and QC might disrupt the interaction between them to stop this phenomena. METHODS: Co-immunoprecipitation assay, immunofluorescence assay, and western blotting were performed by using ex vivo patient-derived breast cancer-stem-cells (PDBCSCs) and human umbilical vein endothelial cells (HUVECs). In silico study was carried out to check the interaction between cMET and ABCG2 in presence or absence of QC. Tube formation assay using HUVECs and in ovo Chorioallantoic membrane (CAM) assay using chick fertilized eggs were performed to monitor angiogenesis. In vivo patient-derived xenograft (PDX) mice model was used to validate in silico and ex vivo results. RESULTS: Data revealed that in a hypoxic tumor microenvironment (TME), cMET and ABCG2 interact with each other and upregulate HIF-1α/VEGF-A axis to induce breast cancer angiogenesis. In silico and ex vivo study showed that QC disrupted the interaction between cMET and ABCG2 to inhibit the angiogenic response in endothelial cells by reducing the secretion of VEGF-A from PDBCSCs within the TME. Knockdown of cMET, ABCG2 or both, significantly downregulated the expression of HIF-1α and reduced the secretion of pro-angiogenic factor VEGF-A in the TME of PDBCSCs. Additionally, when PDBCSCs were treated with QC, similar experimental results were obtained. CONCLUSION: In silico, in ovo, ex vivo and in vivo data confirmed that QC inhibited the HIF-1α/VEGF-A mediated angiogenesis in breast cancer by disrupting the interaction between cMET and ABCG2.


Asunto(s)
Neoplasias de la Mama , Quinacrina , Humanos , Animales , Ratones , Femenino , Quinacrina/farmacología , Quinacrina/metabolismo , Quinacrina/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias de la Mama/patología , Células Endoteliales/metabolismo , Células Madre Neoplásicas/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Línea Celular Tumoral , Microambiente Tumoral , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias/metabolismo
8.
J Cell Commun Signal ; 17(4): 1371-1388, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37162635

RESUMEN

A trans-membrane receptor tyrosine kinase, cMET, belonging to the MET proto-oncogene family, is responsible for cancer metastasis and angiogenesis. But not much is known about the role of cMET in growth and progression of cancer stem cells (CSCs). Earlier studies have shown that Quinacrine (QC), a bioactive agent, has anti-CSCs activity. Here, the role of QC in deregulation of cMET-mediated metastasis and angiogenesis has been systematically evaluated in vitro in highly metastatic breast CSCs (mBCSCs), ex vivo in patient-derived breast cancer stem cells (PDBCSCs) and in vivo in xenograft mice model systems. Cell proliferation, migration, invasion and representative metastasis markers were upregulated in cMET-overexpressed cells and QC exposure inhibited these processes in both mBCSCs and PDBCSCs. Interestingly, metastasis was significantly inhibited by QC in cMET-overexpressed cells but comparatively lesser significant alteration of the process was noted in cMET-silenced cells. Increase in vascularization (in in ovo CAM assay), and cell-cell tube formation (in HUVECs), and enhanced MMP9 and MMP2 enzymatic activities (in gelatin zymography) were noted after cMET overexpression but these processes got reversed after cMET knockdown or QC treatment in cMET-overexpressed cells. QC inhibited angiogenesis significantly in cMET-overexpressed cells, but lesser significant change was observed in cMET-silenced cells. Reduction in tumor volume and decreased expression of metastatic and angiogenic markers were also noted in xenograft mice after QC treatment. Furthermore, QC inhibited cMET activity by dephosphorylation of its tyrosine residues (Y1234 and Y1356) and downregulation of its downstream cascade. Thus, QC inhibited the cMET-mediated metastasis and angiogenesis in in vitro, in ovo, in vivo and ex vivo model systems. Ligand (HGF) binding leads to receptor dimerization and phosphorylation of tyrosine kinase domain of cMET. This activates the cMET signaling cascade. The representative downstream metastasis and angiogenesis-related proteins get upregulated and induce the metastasis and angiogenesis process. But after the QC treatment, cMET get dephosphorylated and inactivated. As a result, the downstream signaling proteins of cMET along with the other representative metastatic and angiogenic factors get downregulated. These lead to inhibition of cMET-mediated metastasis and angiogenesis. (Created with BioRender.com).

9.
Nanomedicine (Lond) ; 18(1): 19-33, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36916388

RESUMEN

Aim: This study aimed to explore the antiangiogenic mechanism of quinacrine-gold hybrid nanoparticle (QAuNP) and near-infrared (NIR) radiation in patient-derived primary breast cancer stem cells. Materials & methods: Various cell-based in ovo angiogenesis and in vivo patient-derived xenograft mouse systems were used as models for the study. Results: The experimental results showed that QAuNP + NIR treatment deregulated the HSP-70/TGF-ß physical interaction in primary breast cancer stem cells. Reduced TGF-ß secretion in the tumor microenvironment inhibited angiogenesis activation in endothelial cells by deregulating the TGF-ß-mediated PI3K/AKT/mTOR cascade. Conclusion: This study revealed that QAuNP + NIR irradiation downregulated HSP-70 expression, inhibited the HSP-70/TGF-ß interaction, reduced the secretion of TGF-ß in the tumor microenvironment and ultimately inhibited TGF-ß-mediated angiogenesis.


This study discovered that the formation of blood vessels in breast cancer is significantly reduced when hybrid nanoparticles and infrared laser therapy are used to treat breast cancer stem cells. The secretory cytokines in the tumor microenvironment primarily responsible for developing blood vessels in the tumor are dramatically reduced by treatment. As a result, the tumor's blood vessel growth is reduced, making it difficult for the cancer cells to get the nutrients and oxygen they need to survive.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Oro , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas , Quinacrina/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Espectroscopía Infrarroja Corta , Proteínas HSP70 de Choque Térmico/metabolismo
10.
Biomater Sci ; 11(10): 3431-3449, 2023 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-36974930

RESUMEN

Viruses have a worldwide impact on healthcare and social and economic growth because they are the largest cause of mortality due to infectious diseases. Furthermore, the long-term conventional drug use comes with substantial risks to public health, such as the rapid evolution of drug resistance and the emergence of secondary side effects. Therefore, it is necessary to develop new methods for the treatment of virus-related diseases. In this case, the use of nanomaterial-based nanomedicines possesses tremendous advantages over the traditional treatment approach. Nanomaterial-based drug delivery systems have unique features that make them promising candidates in the pursuit of therapeutic benefits. In this review, we present the various biocompatible nanomaterials that show promise as nanomedicines for anti-viral therapy. Also, we include how current developments in nanomedicine are being used to treat and prevent the most common viral illnesses such as the flu, HIV, SARS-CoV-2, monkeypox, and human papillomaviruses.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Virosis , Humanos , Nanomedicina/métodos , SARS-CoV-2 , Sistemas de Liberación de Medicamentos/métodos , Virosis/tratamiento farmacológico
11.
Curr Pharm Biotechnol ; 24(11): 1365-1375, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36734900

RESUMEN

Cancer patients frequently report experiencing pain as one of their symptoms. Cancerrelated pain is often caused by the tumor itself, especially when the tumor is pressing on nerves. In addition to the pain caused by the tumor itself, patients also experience discomfort from the treatment, such as surgery, chemotherapy, radiation therapy, and the diagnostic procedures. The majority of today's pain therapies rely on opioid analgesics, which have not been shown to be effective. The adverse effects of opioids and their addictive properties call for the development of innovative treatment techniques. Nanotechnology offers answers to the issues raised above, which are related to the utilization of more conventional modes of therapy. These nanotechnology-based nanotherapeutics reduce the systemic toxicity, offering outstanding selectiveness and prolonged release of the analgesic drugs at the target site. Thus, these reduce cancer-induced pain in the patients. In this article, we will explain the mechanism behind the most common types of pain that are caused by cancer, including neuropathic, somatic, and visceral pain. In addition, a comprehensive discussion is held on the use of various nanotherapeutics as analgesic drug carriers, as well as on their impacts and the potential opportunities that lie ahead in the field of cancer pain treatment.


Asunto(s)
Neoplasias , Manejo del Dolor , Humanos , Manejo del Dolor/efectos adversos , Analgésicos/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/etiología , Analgésicos Opioides , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Nanotecnología
12.
J Nutr Biochem ; 113: 109257, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36572069

RESUMEN

Cancer-associated fibroblasts (CAFs) are one of the highly abundant components in the tumor microenvironment (TME). They secrete several cytokines, which amplified tumor progression, invasion, stemness, metastasis, and angiogenesis. Here, we evaluate the potentiality of cytokines for the formation of cancer stem cells (CSCs) in oral cancer cells niche and investigate the anti-inflammatory and anti-carcinogenic effect of Resveratrol-nanoparticle (Res-NP). We first differentiated quiescent human fibroblasts into CAFs in vitro in response to PDGF-B and TGF-ß stimulation and these CAFs were found to increase CXCL-12 and IL-6 secretion. CSCs-enriched population was created by incubating H-357 cells with CAFs and cytokine-enriched CAFs-conditioned media (CAFs-CM). Likewise, CSCs-populated environment was also generated after incubating CAFs-CM to patient-derived primary oral cancer cells. It was noted that CXCL-12 and IL-6 secreted from CAFs significantly promoted CSCs growth, proliferation, aggressiveness, metastasis, and angiogenesis. However, Res-NP reduced CSCs growth and proliferation by abrogating the secretion of CXCL-12 and IL-6. A significant decrease in the expression of metastatic and angiogenic markers, in ovo blood vascularization, intracellular NO generation, MMPs expression and tube formation was found upon Res-NP treatment. Reduction of representative CSCs and angiogenesis markers were also noted after Res-NP treatment in xenograft mice model. CXCL-12 physically interact with IL-6 and this interaction was diminished after Res-NP treatment. Moreover, the expression of CD133 and VEGF-A were down-regulated either on Res-NP or CXCL-12/IL-6-specific inhibitors treated CSCs-enriched cells. Thus, the data suggest that CSCs growth is CXCL-12 and IL-6 dependent and Res-NP obstruct carcinogenesis and metastasis by inhibiting CXCL-12 and IL-6 production in in vitro, in vivo, in ovo, and ex vivo systems.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Boca , Nanopartículas , Humanos , Animales , Ratones , Citocinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Interleucina-6/metabolismo , Resveratrol/farmacología , Neoplasias de la Boca/metabolismo , Línea Celular Tumoral , Microambiente Tumoral
13.
Exp Cell Res ; 420(1): 113338, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36075449

RESUMEN

Although sensitization of BRCA-mutated, homologous recombination (HR)-deficient breast cancer cells through PARP inhibitor is widely studied, not much is known about the treatment of BRCA-wild-type, HR-proficient breast cancer. Here, we aim to investigate whether a bioactive compound, Resveratrol (RES), can induce DNA double-strand breaks in HR-proficient breast cancer cells and Olaparib (OLA), a PARP inhibitor, can enhance the RES-mediated apoptosis by deregulating the HR repair pathway. The detailed mechanism of anti-cancer action of RES + OLA combination in breast cancer has been evaluated using in vitro, ex vivo, and in vivo preclinical model systems. OLA increased RES-mediated DNA damage, downregulated the HR pathway proteins, caused a late S/G2 cell cycle arrest, enhanced apoptosis and cell death in RES pre-treated breast cancer cells at much lower concentrations than their individual treatments. Direct measurement of HR pathway activity using a GFP plasmid-based assay demonstrated reduced HR efficiency in I-SceI endonuclease-transfected cells treated with OLA. Moreover, RES + OLA treatment also caused significant reduction in PARP1-mediated PARylation and efficiently trapped PARP1 at the DNA damage site. Upon RES treatment, PARylated PARP1 was found to interact with BRCA1, which then activated other HR pathway proteins. But after addition of OLA in RES pre-treated cells, PARP1 could not interact with BRCA1 due to inhibition of PARylation. This resulted in deregulation of HR pathway. To further confirm the role of BRCA1 in PARP1-mediated HR pathway activation, BRCA1 was knocked down that caused complete inhibition of HR pathway activity, and further enhanced apoptosis after RES + OLA treatment in BRCA1-silenced cells. In agreement with in vitro data, similar experimental results were obtained in ex vivo patient-derived breast cancer cells and in vivo xenograft mice. Thus, RES + OLA combination treatment enhanced breast cancer cell death by causing excessive DNA damage and also simultaneously inhibiting the HR pathway.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Antineoplásicos/farmacología , Apoptosis , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , ADN/farmacología , Endonucleasas/genética , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Ftalazinas , Piperazinas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Reparación del ADN por Recombinación , Resveratrol/farmacología
14.
J Cancer Res Clin Oncol ; 148(12): 3521-3535, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35962813

RESUMEN

PURPOSE: Inhibition of Poly (ADP-ribose) Polymerases (PARP) results in the blocking of DNA repair cascades that eventually leads to apoptosis and cancer cell death. PARP inhibitors (PARPi) exhibit their actions either by inhibiting PARP-induced PARylation and/or by trapping PARP at the DNA damage site. But, the mechanism of PARPi-mediated induction of cellular toxicity via PARP-trapping is largely unknown. METHODS: The cellular toxicity of PARPi [Talazoparib (BMN) and/or Olaparib (Ola)] was investigated in oral cancer cells and the underlying mechanism was studied by using in vitro, in silico, and in vivo preclinical model systems. RESULTS: The experimental data suggested that induction of DNA damage is imperative for the optimal effectiveness of PARPi. Curcumin (Cur) exhibited maximum DNA damaging capacity in comparison to Resveratrol and 5-Flurouracil. Combination of BMN + Ola induced cell death in Cur pre-treated cells at much lower concentrations than their individual treatments. BMN + Ola treatment deregulated the BER cascade, potentiated PARP-trapping, caused cell cycle arrest and apoptosis in Cur pre-treated cells in a much more effective manner than their individual treatments. In silico data indicated the involvement of different amino acid residues which might play important roles in enhancing the BMN + Ola-mediated PARP-trapping. Moreover, in vivo mice xenograft data also suggested the BMN + Ola-mediated enhancement of apoptotic potentiality of Cur. CONCLUSION: Thus, induction of DNA damage was found to be essential for optimal functioning of PARPi and BMN + Ola combination treatment enhanced the apoptotic potentiality of Cur in cancer cells by enhancing the PARP-trapping activity via modulation of BER cascade.


Asunto(s)
Curcumina , Neoplasias de la Boca , Humanos , Animales , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Curcumina/farmacología , Resveratrol/farmacología , Ribosa/farmacología , Línea Celular Tumoral , Apoptosis , Poli(ADP-Ribosa) Polimerasas , Neoplasias de la Boca/tratamiento farmacológico , ADN , Aminoácidos/farmacología , Adenosina Difosfato/farmacología
15.
Nanomedicine ; 40: 102502, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34843984

RESUMEN

Cancer stem cells (CSCs) are the tumor cell subpopulations that can self-renew, differentiate, initiate and maintain tumor growth. CSCs are frequently drug-resistant, resulting in tumor recurrence, metastasis, and angiogenesis. Herein, using in vitro oral squamous cell carcinoma (OSCC) CSCs and in vivo xenograft mice model, we have systematically studied the apoptotic potentiality of quinacrine-gold hybrid nanoparticle (QAuNP) and its underlying mechanism after NIR irradiation. QAuNP + NIR caused DNA damage and induced apoptosis in SCC-9-CSCs by deregulating mitochondrial membrane potential (ΔΨm) and activation of ROS. Upregulation of CASPASE-3 and DR-5/DR-4 and reduction of heat shock protein (HSP-70) up to 5-fold were also noticed upon the treatment. The increased expression of DR-5 and CASPASE-3 and decreased expression of HSP-70, CD-44 and Ki-67 were also noted in the xenograft mice treated with QAuNP + NIR + TRAIL. Thus, data suggest that the combined treatment enhances apoptosis in OSCC-CSCs by modulating HSP-70 in the DISC.


Asunto(s)
Antineoplásicos , Carcinoma de Células Escamosas , Neoplasias de la Boca , Nanopartículas , Animales , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Oro/uso terapéutico , Humanos , Ratones , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/radioterapia , Células Madre Neoplásicas/patología , Quinacrina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Vascul Pharmacol ; 140: 106865, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33945869

RESUMEN

Tumor-induced lymphangiogenesis promotes tumor progression by generating new lymphatic vessels that helps in tumor dissemination to regional lymph nodes and distant sites. Recently, the role of Nectin-4 in cancer metastasis and angiogenesis has been studied, but its role in lymphangiogenesis is unknown. Here, we systematically delineated the role of Nectin-4 in lymphangiogenesis and its regulation in invasive duct carcinoma (IDC). Nectin-4 expression positively correlated with occurrence risk factors associated with breast cancer (alcohol, smoke, lifestyle habit, etc), CXCR4 expression, and LYVE-1-lymphatic vessel density (LVD). LVD was significantly higher in axillary lymph node (ALN) than primary tumor. Depleting Nectin-4, VEGF-C or both attenuated the important lymphangiogenic marker LYVE-1 expression, tube formation, and migration of ALN derived primary cells. Nectin-4 stimulated the expressions of CXCR4 and CXCL12 under hypoxic conditions in ALN derived primary cells. Further, Nectin-4 augmented expressions of lymphatic metastatic markers (e.g. eNOS, TGF-ß, CD-105) and MMPs. Induced expressions of Nectin-4 along with other representative metastatic markers were noted in lymph and blood circulating tumor cells (LCTCs and BCTCs) of local and distant metastatic samples. Thus, Nectin-4 displayed a predominant role in promoting tumor-induced lymphangiogenesis and lymphatic metastasis by modulating CXCR4/CXCL12-LYVE-1- axis.


Asunto(s)
Neoplasias de la Mama , Moléculas de Adhesión Celular , Vasos Linfáticos , Nectinas , Receptores CXCR4 , Proteínas de Transporte Vesicular , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/metabolismo , Femenino , Humanos , Linfangiogénesis/fisiología , Metástasis Linfática/patología , Vasos Linfáticos/metabolismo , Nectinas/metabolismo , Receptores CXCR4/metabolismo , Proteínas de Transporte Vesicular/metabolismo
17.
Mol Pharm ; 17(7): 2463-2472, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32407635

RESUMEN

The presence of cancer stem cells (CSCs) in the tumor microenvironment is responsible for the development of chemoresistance and recurrence of cancer. Our previous investigation revealed the anticancer mechanism of quinacrine-based silver and gold hybrid nanoparticles (QAgNP and QAuNP) in oral cancer cells, but to avoid cancer recurrence, it is important to study the effect of these nanoparticles (NPs) on CSCs. Here, we developed an in vitro CSCs model using SCC-9 oral cancer cells and validated via FACS analysis. Then, 40-60% of cells were found to be CD44+/CD133+ and CD24-. QAuNP showed excellent anti-CSC growth potential against SCC-9-cancer stem like cells (IC50 = 0.4 µg/mL) with the down-regulation of representative CSC markers. Prolonged exposure of QAuNP induced the S-phase arrest and caused re-replication shown by the extended G2/M population and apoptosis to SCC-9-CSC like cells. Up-regulation of BAX, PARP cleavage, and simultaneous down-regulation of Bcl-xL in prolonged treatment to CSCs suggested that the majority of the cells have undergone apoptosis. QAuNP treatment also caused a loss in DNA repair in CSCs. Mostly, the base excision repair (BER) components (Fen-1, DNA ligase-1, Pol-ß, RPA, etc.) were significantly down-regulated after QAuNP treatment, which suggested its action against DNA repair machinery. The replication fork maintenance-related proteins, RAD 51 and BRCA-2, were also deregulated. Very surprisingly, depletion of WRN (an interacting partner for Pre-RC and Fen-1) and a significant increase in expression of fork-degrading nuclease MRE-11 in 96 h treated NPs were observed. Results suggest QAuNP treatment caused excessive DNA damage and re-replication mediated replication stress (RS) and stalling of the replication fork. Inhibition of BER components hinders the flap clearance activity of Fen-1, and it further caused RS and stopped DNA synthesis. Overall, QAuNP treatment led to irreparable replication fork movement, and the stalled replication fork might have degraded by MRE-11, which ultimately results in apoptosis and the death of the CSCs.


Asunto(s)
Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Oro/química , Nanopartículas del Metal/química , Células Madre Neoplásicas/efectos de los fármacos , Quinacrina/administración & dosificación , Plata/química , Neoplasias de la Lengua/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Neoplasias de la Lengua/patología , Microambiente Tumoral/efectos de los fármacos
18.
IET Nanobiotechnol ; 14(4): 253-260, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32463015

RESUMEN

The ever-promising opportunities and the uses of NP in our life are increasing but their present and future potential risks on the animals, plants and microorganisms are not well discussed elsewhere. In this review, the authors have systematically discussed the toxic effect of the uses of NP on animals, plants and microorganisms including human health. They have also discussed about the bioaccumulation of these NP in the food chain. Finally, they have provided some possible suggestions for the uses of NP to reduce the detrimental effect on the environment.


Asunto(s)
Nanopartículas , Animales , Bacterias/efectos de los fármacos , Bioacumulación , Línea Celular , Cadena Alimentaria , Humanos , Nanopartículas/análisis , Nanopartículas/química , Nanopartículas/toxicidad , Plantas/efectos de los fármacos , Reciclaje , Riesgo , Pruebas de Toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...