Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
IUBMB Life ; 73(1): 130-145, 2021 01.
Article in English | MEDLINE | ID: mdl-33205598

ABSTRACT

A little number of current autophagy inhibitors may have beneficial effects on the acute myeloid leukemia (AML) patients. However, there is a strong need to figure out which settings should be activated or inhibited in autophagy pathway to prevail drug resistance and also to improve current treatment options in leukemia. Therefore, this study aimed to compare the effects of well-known inhibitors of autophagy (as 3-MA, BafA1, and HCQ) in leukemia KG-1 and HL-60 cells exposed to arsenic trioxide (ATO) and/or all-trans retinoic acid (ATRA). Cell proliferation and cytotoxicity of cells were examined by MTT assay. Autophagy was studied by evaluating the development of acidic vesicular organelles, and the autophagosomes formation was investigated by acridine orange staining and transmission electron microscopy. Moreover, the gene and protein expressions levels of autophagy markers (ATGs, p62/SQSTM1, and LC-3B) were also performed by qPCR and western blotting, respectively. The rate of apoptosis and cell cycle were evaluated using flow cytometry. We compared the cytotoxic and apoptotic effects of ATO and/or ATRA in both cell lines and demonstrated that some autophagy markers upregulated in this context. Also, it was shown that autophagy blockers HCQ and/or BafA1 could potentiate the cytotoxic effects of ATO/ATRA, which were more pronounced in KG-1 cells compared to HL-60 cell line. This study showed the involvement of autophagy during the treatment of KG-1 and HL-60 cells by ATO/ATRA. This study proposed that therapy of ATO/ATRA in combination with HCQ can be considered as a more effective strategy for targeting leukemic KG-1 cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Apoptosis , Arsenic Trioxide/administration & dosage , Cell Proliferation , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Tretinoin/administration & dosage , Tumor Cells, Cultured
2.
J Cell Physiol ; 235(11): 8246-8259, 2020 11.
Article in English | MEDLINE | ID: mdl-31989650

ABSTRACT

The manipulation of autophagy provides a new opportunity for highly effective anticancer therapies. Recently, we showed that photodynamic therapy (PDT) with nitrogen-doped titanium dioxide (N-TiO2 ) nanoparticles (NPs) could promote the reactive oxygen species (ROS)-dependent autophagy in leukemia cells. However, the differential autophagic effects of N-TiO2 NPs in the dark and light conditions and the potential of N-TiO2- based PDT for the treatment of melanoma cells remain unknown. Here we show that depending on the visible-light condition, the autophagic response of human melanoma A375 cells to N-TiO2 NPs switches between two different statuses (ie., flux or blockade) with the opposite outcomes (ie., survival or death). Mechanistically, low doses of N-TiO2 NPs (1-100 µg/ml) stimulate a nontoxic autophagy flux response in A375 cells, whereas their photo-activation leads to the impairment of the autophagosome-lysosome fusion, the blockade of autophagy flux and consequently the induction of RIPK1-mediated necroptosis via ROS production. These results confirm that photo-controllable autophagic effects of N-TiO2 NPs can be utilized for the treatment of cancer, particularly melanoma.


Subject(s)
Autophagy/drug effects , Melanoma , Necroptosis/drug effects , Photosensitizing Agents/pharmacology , Titanium/pharmacology , Cell Line, Tumor , Humans , Light , Melanoma/pathology , Metal Nanoparticles/chemistry , Nitrogen/chemistry , Oxidative Stress/drug effects , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Reactive Oxygen Species , Titanium/chemistry , Titanium/radiation effects
3.
Mol Biol Rep ; 47(11): 9021-9032, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33034883

ABSTRACT

Despite the depth of knowledge concerning the pathogenesis of melanoma, the most aggressive type of skin cancer, the prognosis and survival of patients still remain a challenge. In addition, responses to chemotherapy and immunotherapy are still poor, which underscore an urgent need in the development of new therapeutic strategies for the treatment of melanoma. Recently, the dynamic role of autophagy has gained considerable interest in the pathogenesis and treatment of melanoma. Whereas a decrease in autophagy activity promotes melanoma formation by increasing oncogene-induced tumorigenesis and DNA damage accumulation, an enhanced level of autophagy sustains melanoma cell viability and contributes to drug resistance. Obviously, the understanding of autophagy regulation may lead to a better defining melanoma pathogenesis and the progression toward new treatment options. In this review, we present new insights into a dual role of autophagy during melanoma tumorigenesis. In addition to summarizing current therapeutics for treating melanoma, we discuss how autophagy manipulation may improve the patients' outcome. Finally, autophagy-modulating drugs and nanoparticles, alone or in combination with current therapeutics, are proposed for possible clinical use in melanoma management.


Subject(s)
Autophagy/physiology , Carcinogenesis/drug effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sorafenib/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Carcinogenesis/genetics , Gene Expression/drug effects , Humans , Melanoma/genetics , Melanoma/physiopathology , Skin Neoplasms/genetics , Skin Neoplasms/physiopathology
4.
Iran J Med Sci ; 49(1): 10-21, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38322164

ABSTRACT

Background: Three main cell signaling pathways including the endoplasmic reticulum stress (ERS) response, autophagy, and apoptosis play critical roles in both cell survival and death. They were found to crosstalk with one another during tumorigenesis and cancer progression. This study aimed to investigate the expression of the spliced form of X-box binding protein 1 (XBP1s), p62, and caspase-3, as the essential biomarkers of ERS, autophagy, and apoptosis in patients with colorectal cancer (CRC), as well as the correlation between their expression and clinicopathological data. Methods: This retrospective study was conducted on formalin-fixed paraffin-embedded (FFPE) blocks, which were collected from patients and their tumor margins, from the tumor bank of Imam Khomeini Hospital (Tehran, Iran) from 2017 to 2019. Tissue microarray (TMA) was used to measure the XBP1s, p62, and caspase-3 biomarkers. Data were analyzed using SPSS software version 20, and P≤0.05 was considered statistically significant. Results: Evaluating the total of 91 patients, a significant relationship was found between XBP1s expression and TNM stage (P=0.003), primary tumor (pT) (P=0.054), and the degree of differentiation (P=0.006); and between caspase-3 with pT (P=0.004), and lymphovascular invasion (P=0.02). However, no significant correlation was found between p62 and clinicopathological data. Furthermore, a positive relationship between XBP1s and p62 was confirmed (correlation coefficient: 22.2% and P=0.05). Conclusion: Our findings indicated that XBP1s could be considered as a target for therapy in personalized medicine.


Subject(s)
Caspase 3 , Colorectal Neoplasms , X-Box Binding Protein 1 , Humans , Biomarkers , Caspase 3/genetics , Clinical Relevance , Colorectal Neoplasms/genetics , Iran , Protein Serine-Threonine Kinases/metabolism , Retrospective Studies , X-Box Binding Protein 1/genetics
5.
Int J Pharm ; 649: 123622, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37989403

ABSTRACT

Photodynamic therapy (PDT) represents a non-invasive treatment strategy currently utilized in the clinical management of selected cancers and infections. This technique is predicated on the administration of a photosensitizer (PS) and subsequent irradiation with light of specific wavelengths, thereby generating reactive oxygen species (ROS) within targeted cells. The cellular effects of PDT are dependent on both the localization of the PS and the severity of ROS challenge, potentially leading to the stimulation of various cell death modalities. For many years, the concept of regulated cell death (RCD) triggered by photodynamic reactions predominantly encompassed apoptosis, necrosis, and autophagy. However, in recent decades, further explorations have unveiled additional cell death modalities, such as necroptosis, ferroptosis, cuproptosis, pyroptosis, parthanatos, and immunogenic cell death (ICD), which helps to achieve tumor cell elimination. Recently, nanoparticles (NPs) have demonstrated substantial advantages over traditional PSs and become important components of PDT, due to their improved physicochemical properties, such as enhanced solubility and superior specificity for targeted cells. This review aims to summarize recent advancements in the applications of different metal-based NPs as PSs or delivery systems for optimized PDT in cancer treatment. Furthermore, it mechanistically highlights the contribution of RCD pathways during PDT with metal NPs and how these forms of cell death can improve specific PDT regimens in cancer therapy.


Subject(s)
Metal Nanoparticles , Nanoparticles , Neoplasms , Photochemotherapy , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Photosensitizing Agents/chemistry , Nanoparticles/chemistry , Apoptosis , Cell Line, Tumor , Neoplasms/drug therapy
6.
Front Biosci (Landmark Ed) ; 29(1): 42, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38287839

ABSTRACT

Rheumatoid arthritis (RA) is a relatively common systemic autoimmune disease with an estimated prevalence of approximately 1% worldwide. Patients present predominantly with symmetrical small joint inflammatory arthritis, which involves dysregulated immune responses, leading to bone and cartilage deformities due to extensive erosive damage. The introduction of biological based therapies for the management of this life-altering condition, over the past three decades, has led to marked improvements in patients' quality of life. A wide range of both innate and adaptive immune cells are involved in the pathogenesis of RA, with a complex interplay of cytokines, T-cells, B-cells, and dendritic cells. Some of these cells have been successfully targeted in the treatment of RA by the use of biologics-based therapies. For example, rituximab therapy blocks B cell activation and abatacept effectively blocks T cell activation in patients with RA. Despite these advances, there remain some patients who are resistant to all current therapeutic options, which has encouraged further research into understanding the primary signal transduction pathways that mediate the disease. In this review we discuss the roles of the main signalling pathways, including metabolic reprogramming that have been implicated in RA disease progression, in order to develop a conceptual framework for more precise deployment of existing therapies, and to provide a rationale for producing molecular inhibitors of these pathways. Improved knowledge of the many intracellular signalling pathways in RA will complement current precision medicine strategies, particularly for the patients with difficult-to-treat RA, and especially in those with multidrug resistance disease.


Subject(s)
Arthritis, Rheumatoid , Quality of Life , Humans , Arthritis, Rheumatoid/drug therapy , Cytokines/physiology , Signal Transduction/physiology , Disease Progression
7.
Front Oncol ; 13: 1150492, 2023.
Article in English | MEDLINE | ID: mdl-37213283

ABSTRACT

Autophagy regenerates cellular nutrients, recycles metabolites, and maintains hemostasis through multistep signaling pathways, in conjunction with lysosomal degradation mechanisms. In tumor cells, autophagy has been shown to play a dual role as both tumor suppressor and tumor promoter, leading to the discovery of new therapeutic strategies for cancer. Therefore, regulation of autophagy is essential during cancer progression. In this regard, the use of nanoparticles (NPs) is a promising technique in the clinic to modulate autophagy pathways. Here, we summarized the importance of breast cancer worldwide, and we discussed its classification, current treatment strategies, and the strengths and weaknesses of available treatments. We have also described the application of NPs and nanocarriers (NCs) in breast cancer treatment and their capability to modulate autophagy. Then the advantages and disadvantaged of NPs in cancer therapy along with future applications will be disscussed. The purpose of this review is to provide up-to-date information on NPs used in breast cancer treatment and their impacts on autophagy pathways for researchers.

8.
Med Oncol ; 40(9): 247, 2023 Jul 22.
Article in English | MEDLINE | ID: mdl-37480455

ABSTRACT

Drug resistance is one of the clinical challenges that limits the effectiveness of chemotherapy. Recent reports suggest that the unfolded protein response (UPR) and endoplasmic reticulum stress-adaptation signalling pathway, along with increased activation of its inositol-requiring enzyme 1α (IRE1α) arm, may be contributors to the pathogenesis of colorectal cancer (CRC). Here, we aimed to target the IRE1α/XBP1 pathway in order to sensitise CRC cells to the effects of chemotherapy. The CT26 colorectal cell line was treated with tunicamycin, and then was exposed to different concentrations of 5-fluorouracil (5-FU), either alone and/or in combination with the IRE1α inhibitor, 4µ8C. An MTT assay, flow cytometry and RT-PCR were performed to determine cell growth, apoptosis and IRE1α activity, respectively. In vivo BALB/c syngeneic colorectal mice received chemotherapeutic drugs. Treatment responses, tumour sizes and cytotoxicity were assessed via a range of pathological tests. 4µ8C was found to inhibit the growth of CRC, at a concentration of 10 µg/ml, without detectable cytotoxic effects and also significantly enhanced the cytotoxic potential of 5-FU, in CRC cells. In vivo experiments revealed that 4µ8C, at a concentration of 50 µM/kg prevented tumour growth without any cytotoxic or metastatic effects. Interestingly, the combination of 4µ8C with 5-FU remarkably enhanced drug responses, up to 40-60% and also lead to significantly greater inhibition of tumour growth, in comparison to monotherapy, in CRC mice. Targeting the IRE1α/XBP1 axis of the UPR could enhance the effectiveness of chemotherapy in both in vitro and in vivo models of CRC.


Subject(s)
Colorectal Neoplasms , Endoribonucleases , Animals , Mice , Protein Serine-Threonine Kinases , Fluorouracil/pharmacology , Apoptosis , Mice, Inbred BALB C , Colorectal Neoplasms/drug therapy
9.
Biochim Biophys Acta Mol Basis Dis ; 1868(10): 166484, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35811032

ABSTRACT

Exosomal release pathway and autophagy together maintain homeostasis and survival of cells under stressful conditions. Autophagy is a catabolic process through which cell entities, such as malformed biomacromolecules and damaged organelles, are degraded and recycled via the lysosomal-dependent pathway. Exosomes, a sub-type of extracellular vesicles (EVs) formed by the inward budding of multivesicular bodies (MVBs), are mostly involved in mediating communication between cells. The unfolded protein response (UPR) is an adaptive response that is activated to sustain survival in the cells faced with the endoplasmic reticulum (ER) stress through a complex network that involves protein synthesis, exosomes secretion and autophagy. Disruption of the critical crosstalk between EVs, UPR and autophagy may be implicated in various human diseases, including cancers and neurodegenerative diseases, yet the molecular mechanism(s) behind the coordination of these communication pathways remains obscure. Here, we review the available information on the mechanisms that control autophagy, ER stress and EV pathways, with the view that a better understanding of their crosstalk and balance may improve our knowledge on the pathogenesis and treatment of human diseases, where these pathways are dysregulated.


Subject(s)
Exosomes , Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Humans , Lysosomes , Unfolded Protein Response
10.
Cancers (Basel) ; 13(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638414

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma in children, and is associated with a poor prognosis in patients presenting with recurrent or metastatic disease. The unfolded protein response (UPR) plays pivotal roles in tumor development and resistance to therapy, including RMS. METHODS: In this study, we used immunohistochemistry and a tissue microarray (TMA) on human RMS and normal skeletal muscle to evaluate the expression of key UPR proteins (GRP78/BiP, IRE1α and cytosolic/nuclear XBP1 (spliced XBP1-sXBP1)) in the four main RMS subtypes: alveolar (ARMS), embryonal (ERMS), pleomorphic (PRMS) and sclerosing/spindle cell (SRMS) RMS. We also investigated the correlation of these proteins with the risk of RMS and several clinicopathological indices, such as lymph node involvement, distant metastasis, tumor stage and tumor scores. RESULTS: Our results revealed that the expression of BiP, sXBP1, and IRE1α, but not cytosolic XBP1, are significantly associated with RMS (BiP and sXBP1 p-value = 0.0001, IRE1 p-value = 0.001) in all of the studied types of RMS tumors (n = 192) compared to normal skeletal muscle tissues (n = 16). In addition, significant correlations of BiP with the lymph node score (p = 0.05), and of IRE1α (p value = 0.004), cytosolic XBP1 (p = 0.001) and sXBP1 (p value = 0.001) with the stage score were observed. At the subtype level, BiP and sXBP1 expression were significantly associated with all subtypes of RMS, whereas IRE1α was associated with ARMS, PRMS and ERMS, and cytosolic XBP1 expression was associated with ARMS and SRMS. Importantly, the expression levels of IRE1α and sXBP1 were more pronounced in ARMS than in any of the other subtypes. The results also showed correlations of BiP with the lymph node score in ARMS (p value = 0.05), and of sXBP1 with the tumor score in PRMS (p value = 0.002). CONCLUSIONS: In summary, this study demonstrates that the overall UPR is upregulated and, more specifically, that the IRE1/sXBP1 axis is active in RMS. The subtype and stage-specific dependency on the UPR machinery in RMS may open new avenues for the development of novel targeted therapeutic strategies and the identification of specific tumor markers in this rare but deadly childhood and young-adult disease.

11.
Life Sci ; 248: 117466, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32101760

ABSTRACT

AIMS: Nanoparticles (NPs)-based drugs have been recently introduced to improve the efficacy of current therapeutic strategies for the treatment of cancer; however, the molecular mechanisms by which a NP interacts with cellular systems still need to be delineated. Here, we utilize the autophagic potential of TiO2 NPs for improving chemotherapeutic effects of 5-fluorouracil (5-FU) in human AGS gastric cells. MATERIALS AND METHODS: Cell growth and viability were determined by trypan blue exclusion test and MTT assay, respectively. Vesicular organelles formation was evaluated by acridine orange staining of cells. Cell cycle and apoptosis were monitored by flow cytometry. Reactive oxygen species (ROS) level were measured by DCHF-DA staining. Autophagy was examined by q-PCR and western blotting. Molecular docking was used for studying NP interaction with autophagic proteins. KEY FINDINGS: TiO2 NPs increase ROS production, impair lysosomal function and subsequently block autophagy flux in AGS cells. In addition, the autophagy blockade induced by non-toxic concentrations of TiO2 NPs (1 µg/ml) can promote cytotoxic and apoptotic effects of 5-FU in AGS cells. SIGNIFICANCE: These results confirm the beneficial effects of TiO2 NPs in combination with chemotherapy in in vitro model of gastric cancer, which may pave the way to develop a possible solution to circumvent chemoresistance in cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Autophagy/drug effects , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Nanoparticles/chemistry , Titanium/pharmacology , Antimetabolites, Antineoplastic/chemistry , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fluorouracil/chemistry , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Docking Simulation , Nanoparticles/ultrastructure , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Titanium/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Adv Pharm Bull ; 10(2): 323-328, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32373503

ABSTRACT

Purpose: Triple-negative breast cancer (TNBC) is specified by high vascularity and repetitious metastasis. Although several studies have indicated that angiogenesis has an important role in invasive breast cancer, a suitable model of TNBC that can show the exact onset of angiogenesis factors still needs to be developed. The purpose of this study is to determine the expression level of angiogenesis factors in different clinical stages of the 4T1 tumor as TNBC mouse model. Methods: Twenty mice were injected by the 4T1 cell line, and four mice selected as healthy controls. Following by tumor induction, the mice were randomly put into four groups, each contains four mice. Once the tumor volume reached to the early stage (<100 mm3), intermediate stage (100-300 mm3), advanced stage (300-500 mm3), and end stage (>500 mm3), they were removed by surgery. Then, the expression levels of Hif1α, VEGFR1, and VEGFR2 genes, as well as tumor markers of VEGF, bFGF and CD31, were evaluated by qPCR and immunohistochemistry (IHC) respectively. The statistical analysis was done by SPSS version 16. Results: TNBC tumors were confirmed and multi-foci metastasis in the lung were seen. The mRNA and protein expression levels of the angiogenesis factors increased in the early stage and as the tumor grew, their expression level enhanced dramatically. Conclusion: The 4T1 syngeneic mouse tumor may serve as an appropriate TNBC model for further investigation of the angiogenesis and therapies. Moreover, angiogenesis factors are induced before the advanced stage, and anti-angiogenesis therapy is necessary to be considered at the first line of treatment in TBNC.

13.
Adv Pharm Bull ; 9(3): 505-509, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31592115

ABSTRACT

Purpose: Despite recent advances in the diagnosis and treatment of rheumatoid arthritis (RA), this inflammatory disease remains a challenge to patients and physicians. Recent evidence highlights the contribution of endoplasmic reticulum (ER) stress in the pathogenesis and treatment of RA. Herein, we study the expression of the ER stress sensor inositol-requiring enzyme 1α (IRE1α), as well as XBP1 splicing and the regulated IRE1-dependent decay (RIDD), in peripheral blood mononuclear cells (PBMCs) from patients with RA compared with healthy controls. Methods: The PBMCs from blood samples of RA patients and healthy volunteers were isolated by a density gradient centrifugation method using Ficoll. The gene expression levels of GRP78/ Bip, IRE1, XBP1s, micro-RNAs (miRNAs) were evaluated by real-time PCR. Results: The expression of GRP78, IRE1, and XBP1s were increased in PBMCs of RA patients compared with healthy controls. We further show that the RIDD targets (miRNA-17, -34a, -96, and -125b) were downregulated in RA samples. Conclusion: This study can expand our knowledge on the importance of RNase activity of IRE1α in RA and may offer new potentials for developing novel diagnostic and/or therapeutic biomarkers.

14.
Autophagy ; 15(1): 4-33, 2019 01.
Article in English | MEDLINE | ID: mdl-30160607

ABSTRACT

Nanomaterials have gained a rapid increase in use in a variety of applications that pertain to many aspects of human life. The majority of these innovations are centered on medical applications and a range of industrial and environmental uses ranging from electronics to environmental remediation. Despite the advantages of NPs, the knowledge of their toxicological behavior and their interactions with the cellular machinery that determines cell fate is extremely limited. This review is an attempt to summarize and increase our understanding of the mechanistic basis of nanomaterial interactions with the cellular machinery that governs cell fate and activity. We review the mechanisms of NP-induced necrosis, apoptosis and autophagy and potential implications of these pathways in nanomaterial-induced outcomes. Abbreviations: Ag, silver; CdTe, cadmium telluride; CNTs, carbon nanotubes; EC, endothelial cell; GFP, green fluorescent protein; GO, graphene oxide; GSH, glutathione; HUVECs, human umbilical vein endothelial cells; NP, nanoparticle; PEI, polyethylenimine; PVP, polyvinylpyrrolidone; QD, quantum dot; ROS, reactive oxygen species; SiO2, silicon dioxide; SPIONs, superparamagnetic iron oxide nanoparticles; SWCNT, single-walled carbon nanotubes; TiO2, titanium dioxide; USPION, ultra-small super paramagnetic iron oxide; ZnO, zinc oxide.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Metal Nanoparticles/toxicity , Apoptosis/genetics , Autophagy/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Lysosomes/drug effects , Metal Nanoparticles/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Necroptosis/drug effects , Necroptosis/genetics , Necrosis , Particle Size , Quantum Dots/chemistry , Quantum Dots/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Trends Pharmacol Sci ; 39(7): 610-623, 2018 07.
Article in English | MEDLINE | ID: mdl-29691058

ABSTRACT

Diverse physiological and pathological conditions that impact on protein folding of the endoplasmic reticulum (ER) cause ER stress. The unfolded protein response (UPR) and the ER-associated degradation (ERAD) pathway are activated to cope with ER stress. In rheumatoid arthritis (RA), inflammation and ER stress work in parallel by driving inflammatory cells to release cytokines that induce chronic ER stress pathways. This chronic ER stress may contribute to the pathogenesis of RA through synoviocyte proliferation and proinflammatory cytokine production. Therefore, ER stress pathways and their constituent elements are attractive targets for RA drug development. In this review, we integrate current knowledge of the contribution of ER stress to the overall pathogenesis of RA, and suggest some therapeutic implications of these discoveries.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum-Associated Degradation/drug effects , Animals , Arthritis, Rheumatoid/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans
16.
Nanomaterials (Basel) ; 8(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134524

ABSTRACT

Nanoparticles (NPs) are currently used in diagnosis and treatment of many human diseases, including autoimmune diseases and cancer. However, cytotoxic effects of NPs on normal cells and living organs is a severe limiting factor that hinders their use in clinic. In addition, diversity of NPs and their physico-chemical properties, including particle size, shape, surface area, dispersity and protein corona effects are considered as key factors that have a crucial impact on their safe or toxicological behaviors. Current studies on toxic effects of NPs are aimed to identify the targets and mechanisms of their side effects, with a focus on elucidating the patterns of NP transport, accumulation, degradation, and elimination, in both in vitro and in vitro models. NPs can enter the body through inhalation, skin and digestive routes. Consequently, there is a need for reliable information about effects of NPs on various organs in order to reveal their efficacy and impact on health. This review covers the existing knowledge base on the subject that hopefully prepares us better to address these challenges.

17.
Cancer Lett ; 424: 46-69, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29474859

ABSTRACT

The dysregulation of autophagy is involved in the pathogenesis of a broad range of diseases, and accordingly universal research efforts have focused on exploring novel compounds with autophagy-modulating properties. While a number of synthetic autophagy modulators have been identified as promising cancer therapy candidates, autophagy-modulating phytochemicals have also attracted attention as potential treatments with minimal side effects. In this review, we firstly highlight the importance of autophagy and its relevance in the pathogenesis and treatment of cancer. Subsequently, we present the data on common phytochemicals and their mechanism of action as autophagy modulators. Finally, we discuss the challenges associated with harnessing the autophagic potential of phytochemicals for cancer therapy.


Subject(s)
Autophagy/drug effects , Neoplasms/drug therapy , Phytochemicals/pharmacology , Autophagy-Related Proteins/metabolism , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Structure , Neoplasms/metabolism , Phytochemicals/chemistry , Phytochemicals/therapeutic use
18.
Leuk Res ; 63: 15-21, 2017 12.
Article in English | MEDLINE | ID: mdl-29096331

ABSTRACT

Here, we report that targeting Nucleostemin (NS), a recently discovered stem cells-enriched gene, by a specific small interference RNA (siNS), decreases the rate of proliferation of acute promyelocytic leukemia (APL) NB4 cells and induces differentiation and autophagy. In addition, NS silencing promotes the effects of all-trans-retinoic acid (ATRA)-based differentiation therapy in NB4 cells. Autophagy inhibitors 3-methyladenine and bafilomycin block the effect of NS targeting on differentiation, indicating a new functional link between NS and autophagy as an important regulator of differentiation in NB4 cells. The capability of NS in modulating autophagy and differentiation, alone or in combination with ATRA, may help to broaden the range of treatment options available to treat leukemia.


Subject(s)
Autophagy/drug effects , Cell Differentiation/drug effects , GTP-Binding Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Leukemia, Promyelocytic, Acute/pathology , Nuclear Proteins/antagonists & inhibitors , Tretinoin/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , GTP-Binding Proteins/genetics , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Tumor Cells, Cultured
19.
Sci Rep ; 6: 34413, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27698385

ABSTRACT

In this study, we used nitrogen-doped titanium dioxide (N-TiO2) NPs in conjugation with visible light, and show that both reactive oxygen species (ROS) and autophagy are induced by this novel NP-based photodynamic therapy (PDT) system. While well-dispersed N-TiO2 NPs (≤100 µg/ml) were inert, their photo-activation with visible light led to ROS-mediated autophagy in leukemia K562 cells and normal peripheral lymphocytes, and this increased in parallel with increasing NP concentrations and light doses. At a constant light energy (12 J/cm2), increasing N-TiO2 NP concentrations increased ROS levels to trigger autophagy-dependent megakaryocytic terminal differentiation in K562 cells. By contrast, an ROS challenge induced by high N-TiO2 NP concentrations led to autophagy-associated apoptotic cell death. Using chemical autophagy inhibitors (3-methyladenine and Bafilomycin A1), we confirmed that autophagy is required for both terminal differentiation and apoptosis induced by photo-activated N-TiO2. Pre-incubation of leukemic cells with ROS scavengers muted the effect of N-TiO2 NP-based PDT on cell fate, highlighting the upstream role of ROS in our system. In summary, PDT using N-TiO2 NPs provides an effective method of priming autophagy by ROS induction. The capability of photo-activated N-TiO2 NPs in obtaining desirable cellular outcomes represents a novel therapeutic strategy of cancer cells.


Subject(s)
Autophagy/drug effects , Cell Differentiation/drug effects , Leukemia/drug therapy , Nanoparticles , Nitrogen , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Titanium , Humans , K562 Cells , Leukemia/metabolism , Leukemia/pathology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Nitrogen/chemistry , Nitrogen/pharmacology , Titanium/chemistry , Titanium/pharmacology
20.
Pathol Oncol Res ; 21(2): 293-300, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25417742

ABSTRACT

Breast cancer is the most frequent cause of mortality in women all around the world; therefore, study on molecular aspects of breast cancer is necessary for finding new biomarkers. Recent studies have shown that WW Binding Protein 2 (WBP2) is an important protein for the oncogenic property of cancer. We have previously evaluated the WW Binding Protein 2 N-Terminal Like (WBP2NL) gene expression in cancerous cell line and breast tumor tissues, and reported changes in expression, which could increase tumorigenic cell growth. However, the molecular mechanisms of WBP2NL and its clinical relevance have not been investigated. In this study, the expression of WBP2NL-related genes in the invasive breast carcinoma and normal breast tissues was evaluated for the first time. Analysis of WBP2NL-related genes expression was performed with reverse transcription-PCR and real time-PCR detection method. The target genes studied were as follow: WW domain containing E3 ubiquitin protein ligase 1(WWP1), membrane associated guanylatekinase containing WW and PDZ domain-1 (MAGI1), neural precursor cell expressed developmentally down-regulated 4 (NEDD4), formin binding protein-4 (FNBP4), BCL2-associated athanogene-3 (BAG3), WW domain-containing oxidoreductase (WWOX), yes-associated protein-1 (YAP1), WW domain containing transcription regulator (WWTR1), member RAS oncogene family (RAB2A), and small G protein signaling modulator 3 (SGSM3). The expression of WWP1, BAG3, and WWTR1 was significantly increased in breast cancer. In contrast, the expression of WWOX, YAP1, RAB2A, and SGSM3 was significantly decreased. The MAGI1 and NEDD4 expression was increased, while the expression of FNBP4 was unchanged. These findings lead us to suggest that WBP2NL might play roles as an anti-apoptotic factor or co-activator to promote breast cancer cell survival and proliferation.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Carcinoma, Ductal, Breast/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Seminal Plasma Proteins/metabolism , Up-Regulation/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carrier Proteins/genetics , Case-Control Studies , Cell Adhesion Molecules , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation/genetics , Cell Proliferation/physiology , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Guanylate Kinases , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Middle Aged , Nedd4 Ubiquitin Protein Ligases , Seminal Plasma Proteins/genetics , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL