Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Mol Ther ; 32(1): 13-31, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37919901

ABSTRACT

mRNA vaccines have evolved as promising cancer therapies. These vaccines can encode tumor-allied antigens, thus enabling personalized treatment approaches. They can also target cancer-specific mutations and overcome immune evasion mechanisms. They manipulate the body's cellular functions to produce antigens, elicit immune responses, and suppress tumors by overcoming limitations associated with specific histocompatibility leukocyte antigen molecules. However, successfully delivering mRNA into target cells destroys a crucial challenge. Viral and nonviral vectors (lipid nanoparticles and cationic liposomes) have shown great capacity in protecting mRNA from deterioration and assisting in cellular uptake. Cell-penetrating peptides, hydrogels, polymer-based nanoparticles, and dendrimers have been investigated to increase the delivery efficacy and immunogenicity of mRNA. This comprehensive review explores the landscape of mRNA vaccines and their delivery platforms for cancer, addressing design considerations, diverse delivery strategies, and recent advancements. Overall, this review contributes to the progress of mRNA vaccines as an innovative strategy for effective cancer treatment.


Subject(s)
Cancer Vaccines , Communicable Diseases , Nanoparticles , Neoplasms , Vaccines , Humans , mRNA Vaccines , Neoplasms/genetics , Neoplasms/therapy , Antigens, Neoplasm , Nanoparticles/chemistry , RNA, Messenger/genetics , Cancer Vaccines/genetics
2.
Semin Cancer Biol ; 97: 12-20, 2023 12.
Article in English | MEDLINE | ID: mdl-37926347

ABSTRACT

Obesity is a prominent health issue worldwide and directly impacts pancreatic health, with obese individuals exhibiting a significant risk for increasing pancreatic ductal adenocarcinoma (PDAC). Several factors potentially explain the increased risk for the development of PDAC, including obesity-induced chronic inflammation within and outside of the pancreas, development of insulin resistance and metabolic dysfunction, promotion of immune suppression within the pancreas during inflammation, pre- and malignant stages, variations in hormones levels (adiponectin, ghrelin, and leptin) produced from the adipose tissue, and acquisition of somatic mutations in tumor once- and suppressor proteins critical for pancreatic tumorigenesis. In this manuscript, we will explore the broad impact of these obesity-induced risk factors on the development and progression of PDAC, focusing on changes within the tumor microenvironment (TME) as they pertain to prevention, current therapeutic strategies, and future directions for targeting obesity management as they relate to the prevention of pancreatic tumorigenesis.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/therapy , Obesity/complications , Obesity/metabolism , Inflammation/complications , Carcinogenesis , Tumor Microenvironment
3.
Semin Cancer Biol ; 88: 67-80, 2023 01.
Article in English | MEDLINE | ID: mdl-36535506

ABSTRACT

Pancreatic cancer is one of the most aggressive cancers worldwide due to the resistances to conventional therapies and early metastasis. Recent research has shown that cancer stem cell populations modulate invasiveness, recurrence, and drug resistance in various cancers, including pancreatic cancer. Pancreatic cancer stem cells (PaCSCs) are characterized by their high plasticity and self-renewal capacities that endow them with unique metabolic, metastatic, and chemoresistant properties. Understanding the exact molecular and signaling mechanisms that underlay malignant processes in PaCSCs is instrumental for developing novel therapeutic modalities that overcome the limitations of current therapeutic regimens. In this paper, we provide an updated review of the latest research in the field and summarize the current knowledge of PaCSCs characteristics, cellular metabolism, stemness, and drug resistance. We explore how the crosstalk between the TME and PaCSCs influences stemness. We also highlight some of the key signalling pathways involved in PaCSCs stemness and drug evasion. The aim of this review is to explore how PaCSCs develop, maintain their properties, and drive tumor relapse in PC. The last section explores some of the latest therapeutic strategies aimed at targeting PaCSCs.


Subject(s)
Drug Resistance, Neoplasm , Pancreatic Neoplasms , Humans , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms
4.
Semin Cancer Biol ; 86(Pt 3): 616-621, 2022 11.
Article in English | MEDLINE | ID: mdl-34774995

ABSTRACT

Pancreatic cancer (PC) is a major gastrointestinal cancer in terms of worldwide incidence and mortality. Despite advances in diagnostic and treatment modalities, the mortality of PC is still a serious concern in both sexes. Immune therapy using inhibitors of immune checkpoints, especially inhibitors of programmed cell death protein 1/programmed cell death ligand-1(PD-1/PD-L1), offer huge benefits to cancer patients. This review describes an up-to-date information on the role of PD-1 and PD-L1 in the development of immune tolerance in PC alongside the current clinical trials and the known outcomes citing the available literature. We also included the details on PD-1/PD-L1-mediated signalling in maintenance of PC stem cells and metastasis. We reviewed the critical information on safety, tolerance, and efficacy of clinically important regimens of PD-1/PD-L1 blocking agents and targeted therapeutics. This review elucidates the underlying mechanisms of PD-1/PD-L1 alliance in tolerance of the immune system, maintenance of stem cells, and metastasis promotion as well as design regimens with high safety and excellent tolerability and efficacy for management of PC in advanced stages.


Subject(s)
B7-H1 Antigen , Pancreatic Neoplasms , Humans , Programmed Cell Death 1 Receptor , Immunotherapy , Pancreatic Neoplasms/therapy , Immunologic Factors , Pancreatic Neoplasms
5.
Semin Cancer Biol ; 86(Pt 3): 622-632, 2022 11.
Article in English | MEDLINE | ID: mdl-34324953

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and has a high fatality rate. Genetic and epigenetic aberrations are commonly observed in HCC. The epigenetic processes include chromatin remodelling, histone alterations, DNA methylation, and noncoding RNA (ncRNA) expression and are connected with the progression and metastasis of HCC. Due to their potential reversibility, these epigenetic alterations are widely targeted for the development of biomarkers. In-depth understanding of the epigenetics of HCC is critical for developing rational clinical strategies that can provide a meaningful improvement in overall survival and prediction of therapeutic outcomes. In this article, we have summarised the epigenetic modifications involved in HCC progression and highlighted the potential biomarkers for diagnosis and drug development.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Epigenesis, Genetic , DNA Methylation , Epigenomics
6.
Semin Cancer Biol ; 86(Pt 2): 1155-1162, 2022 11.
Article in English | MEDLINE | ID: mdl-34147639

ABSTRACT

Despite advanced therapeutic strategies, the mortality and morbidity of pancreatic cancer (PC) have been increasing. This is due to the anomalous proliferation activity of stromal cells, like cancer-associated fibroblasts (CAFs), in the tumor microenvironment (TME). These cells develop resistance in the tumor cells, blocking the drug from entering the target tumor site, ultimately resulting in tumor metastasis. Additionally, the current conventional adjuvant techniques, including chemo and radiotherapy, carry higher risk due to their excess toxicity against normal healthy cells. Phytochemicals including curcumin, irinotecan and paclitaxel are anti-oxidants, less toxic, and have anti-cancerous properties; however, the use of phytochemicals is limited due to their less solubility and bioavailability. Nanotechnology offers the resources to directly target the drug to the tumor site, thereby enhancing the therapeutic efficacy of the current treatment modalities. This review focuses on the importance of nanotechnology for pancreatic ductal adenocarcinoma (PDAC) therapy and on delivering the nano-formulated phytochemicals to the target site.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tumor Microenvironment , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms
7.
Mol Cancer ; 22(1): 65, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997931

ABSTRACT

HOX transcript antisense intergenic RNA (HOTAIR) is an oncogenic non-coding RNA whose expression is strongly correlated with the tumor grade and prognosis of a variety of carcinomas including breast cancer (BC). HOTAIR regulates various target genes via sponging and epigenetic mechanisms and controls various oncogenic cellular and signaling mechanisms including metastasis and drug resistance. In BC cells, HOTAIR expression is regulated by a variety of transcriptional and epigenetic mechanisms. In this review, we describe the regulatory mechanisms that govern HOTAIR expression during cancer development and explore how HOTAIR drives BC development, metastasis, and drug resistance. In the final section of this review, we focus on the role of HOTAIR in BC management, therapeutic treatment, and prognosis, highlighting its potential therapeutic applications.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Prognosis , RNA, Long Noncoding/genetics
8.
Semin Cancer Biol ; 69: 69-76, 2021 02.
Article in English | MEDLINE | ID: mdl-31954835

ABSTRACT

Gastric cancer represents a deadly malignancy worldwide, yet current therapeutic regimens remain ineffective. Nanoparticle (NP) -based solutions could allow the design of novel therapeutic methods to eliminate this fatal disease. NPs typically carry out a significant role in multifunctional, multimodal imaging, and drug delivery carriers. In the recent decade, they have emerged as candidate approaches for the design of novel treatment strategies. Tumor nanotherapeutics characteristically possess various distinct advantages compared to conventional anti-cancer medications, which suffer from nonspecific bio-distribution, low solubility, and poor bioavailability. In this review, we will discuss the application of NPs in diagnosis and controlled drug delivery in gastric cancer (GC). We will focus on various NPs-based strategies employed against GC.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Multimodal Imaging/methods , Nanoparticles/administration & dosage , Stomach Neoplasms/drug therapy , Animals , Biological Availability , Humans , Nanoparticles/chemistry , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology
9.
Semin Cancer Biol ; 69: 178-189, 2021 02.
Article in English | MEDLINE | ID: mdl-31419527

ABSTRACT

Cancer is an outrageous disease with uncontrolled differentiation, growth, and migration to the other parts of the body. It is the second-most common cause of death both in the U.S. and worldwide. Current conventional therapies, though much improved and with better prognosis, have several limitations. Chemotherapeutic agents, for instance, are cytotoxic to both tumor and healthy cells, and the non-specific distribution of drugs at tumor sites limits the dose administered. Nanotechnology, which evolved from the coalescence and union of varied scientific disciplines, is a novel science that has been the focus of much research. This technology is generating more effective cancer therapies to overcome biomedical and biophysical barriers against standard interventions in the body; its unique magnetic, electrical, and structural properties make it a promising tool. This article reviews endogenous- and exogenous-based stimulus-responsive drug delivery systems designed to overcome the limitations of conventional therapies. The article also summarizes the study of nanomaterials, including polymeric, gold, silver, magnetic, and quantum dot nanoparticles. Though an array of drug delivery systems has so far been proposed, there remain many challenges and concerns that should be addressed in order to fill the gaps in the field. Prominence is given to drug delivery systems that employ external- and internal-based stimuli and that are emerging as promising tools for cancer therapeutics in clinical settings.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Nanomedicine , Nanoparticles/administration & dosage , Nanostructures/chemistry , Neoplasms/drug therapy , Animals , Humans , Nanoparticles/chemistry , Neoplasms/pathology
10.
Semin Cancer Biol ; 69: 376-390, 2021 02.
Article in English | MEDLINE | ID: mdl-31301361

ABSTRACT

Female-specific cancers are the most common cancers in women worldwide. Early detection methods remain unavailable for most of these cancers, signifying that most of them are diagnosed at later stages. Furthermore, current treatment options for most female-specific cancers are surgery, radiation and chemotherapy. Although important milestones in molecularly targeted approaches have been achieved lately, current therapeutic strategies for female-specific cancers remain limited, ineffective and plagued by the emergence of chemoresistance, which aggravates prognosis. Recently, the application of nanotechnology to the medical field has allowed the development of novel nano-based approaches for the management and treatment of cancers, including female-specific cancers. These approaches promise to improve patient survival rates by reducing side effects, enabling selective delivery of drugs to tumor tissues and enhancing the uptake of therapeutic compounds, thus increasing anti-tumor activity. In this review, we focus on the application of nano-based technologies to the design of novel and innovative diagnostic and therapeutic strategies in the context of female-specific cancers, highlighting their potential uses and limitations.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Genital Neoplasms, Female/drug therapy , Nanomedicine , Nanoparticles/administration & dosage , Animals , Female , Genital Neoplasms, Female/pathology , Humans , Nanoparticles/chemistry
11.
Semin Cancer Biol ; 69: 293-306, 2021 02.
Article in English | MEDLINE | ID: mdl-31260733

ABSTRACT

Colorectal cancer (CRC) is one of the deadliest diseases worldwide due to a lack of early detection methods and appropriate drug delivery strategies. Conventional imaging techniques cannot accurately distinguish benign from malignant tissue, leading to frequent misdiagnosis or diagnosis at late stages of the disease. Novel screening tools with improved accuracy and diagnostic precision are thus required to reduce the mortality burden of this malignancy. Additionally, current therapeutic strategies, including radio- and chemotherapies carry adverse side effects and are limited by the development of drug resistance. Recent advances in nanotechnology have rendered it an attractive approach for designing novel clinical solutions for CRC. Nanoparticle-based formulations could assist early tumor detection and help to overcome the limitations of conventional therapies including poor aqueous solubility, nonspecific biodistribution and limited bioavailability. In this review, we shed light on various types of nanoparticles used for diagnosis and drug delivery in CRC. In addition, we will explore how these nanoparticles can improve diagnostic accuracy and promote selective drug targeting to tumor sites with increased efficiency and reduced cytotoxicity against healthy colon tissue.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Multimodal Imaging/methods , Nanoparticles/administration & dosage , Nanotechnology/methods , Animals , Colorectal Neoplasms/diagnostic imaging , Humans , Nanoparticles/chemistry
12.
J Cell Physiol ; 237(9): 3496-3504, 2022 09.
Article in English | MEDLINE | ID: mdl-35853181

ABSTRACT

The evolutionarily conserved signaling intermediate in toll pathway (ECSIT) is a cytosolic adaptor protein associated with the toll-like receptor pathway. It has a distinct N-terminal mitochondrial targeting sequence, pentatricopeptide repeat motif, and a C-terminal pleckstrin homology domain. ECSIT regulates many biological processes like embryonic development, inflammation, cardiac function, and assembly of mitochondrial complex I. Besides, ECSIT also interacts with multiple signaling intermediates like tumor necrosis receptor associated factor 6 and retinoic acid inducible gene 1 as well as regulates various pathways in the microcellular environment. However, molecular details of ECSIT functions in pathophysiology remain elusive. This review summarizes the diverse functions of ECSIT and its involvement in pathophysiological conditions such as Alzheimer's, oxidative stress, and infection.


Subject(s)
Adaptor Proteins, Signal Transducing , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Electron Transport Complex I/metabolism , Mitochondria/genetics , Mitochondria/metabolism
13.
Cancer ; 128(6): 1171-1183, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34990009

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive and immunogenic subtype of breast cancer. This tumorigenicity is independent of hormonal or HER2 pathways because of a lack of respective receptor expression. TNBC is extremely prone to drug resistance and early recurrence because of T-regulatory cell (Treg) infiltration into the tumor microenvironment (TME) in addition to other mechanisms like genomic instability. Tumor-infiltrating Tregs interact with both tumor and stromal cells as well as extracellular matrix components in the TME and induce an immune-suppressive phenotype. Hence, treatment of TNBC with conventional therapies remains challenging. Understanding the protective mechanism of Tregs in shielding TNBC from antitumor immune responses in the TME will pave the way for developing novel, immune-based therapeutics. The current review focuses on the role of tumor-infiltrating Tregs in tumor progression and metabolic reprogramming of the TME. The authors have extended their focus to oncotargeting Treg-mediated immune suppression in breast cancer. Because of its potential role in the TME, modulating Treg activity may provide a novel strategic intervention to combat TNBC. Both under laboratory conditions and in clinical trials, currently available anticancer drugs and natural therapeutics as potential agents for targeting Tregs are explored.


Subject(s)
Triple Negative Breast Neoplasms , Humans , T-Lymphocytes, Regulatory , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
14.
Pharmacol Res ; 178: 106197, 2022 04.
Article in English | MEDLINE | ID: mdl-35358681

ABSTRACT

Colorectal cancer (CRC) is classified as the third leading cancer globally and one of the major causes of morbidity and mortality around the world. The interaction of the immune system with the cancer cells plays a vital role in CRC progression. Regulatory T cells (Tregs) are a form of T cells, which regulate and suppress unwanted activation of the immune system and play a major role in preventing autoimmune diseases. Tregs exhibit a significant role in immune modulation during CRC progression through accumulation in the tumor microenvironment (TME) and suppression of immunity against tumor specific antigens promoting tumor progression. The role of Tregs in CRC progression and its interaction with other immune cells within CRC TME and current clinical trials were reviewed in this paper.


Subject(s)
Autoimmune Diseases , Colorectal Neoplasms , Antigens, Neoplasm , Colorectal Neoplasms/pathology , Humans , T-Lymphocytes, Regulatory , Tumor Microenvironment
15.
J Nanobiotechnology ; 20(1): 274, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35701781

ABSTRACT

The tumor microenvironment (TME) plays a key role in cancer development and emergence of drug resistance. TME modulation has recently garnered attention as a potential approach for reprogramming the TME and resensitizing resistant neoplastic niches to existing cancer therapies such as immunotherapy or chemotherapy. Nano-based solutions have important advantages over traditional platform and can be specifically targeted and delivered to desired sites. This review explores novel nano-based approaches aimed at targeting and reprogramming aberrant TME components such as macrophages, fibroblasts, tumor vasculature, hypoxia and ROS pathways. We also discuss how nanoplatforms can be combined with existing anti-tumor regimens such as radiotherapy, immunotherapy, phototherapy or chemotherapy to enhance clinical outcomes in solid tumors.


Subject(s)
Nanoparticles , Neoplasms , Humans , Immunologic Factors , Immunotherapy , Macrophages , Neoplasms/drug therapy , Tumor Microenvironment
16.
Exp Cell Res ; 394(1): 112126, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32485183

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) has been recognized as one of the essential regulators that is expressed in greater levels in pancreatic cancer (PC) and is connected with poor prognosis. Resveratrol was identified as a natural compound with many biological functions, with anti-inflammatory, antioxidant, and anticancer effects that inhibit the proliferation and progression of PC cells caused by HIF-1α. The current investigation explored the binding affinity and ligand efficacy of resveratrol against HIF-1α using an in silico approach, and the execution of molecular dynamics simulation (MDS) increased the prediction precision of these outcomes. This is the first study that provides an in silico characterization of the interaction between resveratrol and HIF-1α and its spatial structural arrangements in pancreatic cancer therapy, providing an in-depth analysis of their drug target interactions.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Resveratrol/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Stilbenes/pharmacology , Pancreatic Neoplasms
17.
Semin Cancer Biol ; 56: 149-167, 2019 06.
Article in English | MEDLINE | ID: mdl-30314681

ABSTRACT

Pancreatic cancer (PC) is an aggressive carcinoma and the fourth cause of cancer deaths in Western countries. Although surgery is the most effective therapeutic option for PC, the management of unresectable, locally advanced disease is highly challenging. Our improved understanding of pancreatic tumor biology and associated pathways has led to the development of various treatment modalities that can control the metastatic spread of PC. This review intends to present trials of small molecule tyrosine kinase inhibitors (TKIs) in PC management and the troubles encountered due to inevitable acquired resistance to TKIs.


Subject(s)
Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Clinical Trials as Topic , Drug Resistance, Neoplasm/genetics , Humans , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Retreatment , Signal Transduction/drug effects , Standard of Care , Treatment Outcome
18.
Cancer ; 126(14): 3360-3371, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32383803

ABSTRACT

BACKGROUND: The induction of reactive oxygen species (ROS) represents a viable strategy for enhancing the activity of radiotherapy. The authors hypothesized that napabucasin would increase ROS via its ability to inhibit NAD(P)H:quinone oxidoreductase 1 and potentiate the response to chemoradiotherapy in rectal cancer via distinct mechanisms. METHOD: Proliferation studies, colony formation assays, and ROS levels were measured in HCT116 and HT29 cell lines treated with napabucasin, chemoradiation, or their combination. DNA damage (pγH2AX), activation of STAT, and downstream angiogenesis were evaluated in both untreated and treated cell lines. Finally, the effects of napabucasin, chemoradiotherapy, and their combination were assessed in vivo with subcutaneous mouse xenograft models. RESULTS: Napabucasin significantly potentiated the growth inhibition of chemoradiation in both cell lines. Napabucasin increased ROS generation. Inhibition of ROS by N-acetylcysteine decreased the growth inhibitory effect of napabucasin alone and in combination with chemoradiotherapy. Napabucasin significantly increased pγH2AX in comparison with chemoradiotherapy alone. Napabucasin reduced the levels of pSTAT3 and VEGF and inhibited angiogenesis through an ROS-mediated effect. Napabucasin significantly potentiated the inhibition of growth and blood vessel formation by chemoradiotherapy in mouse xenografts. CONCLUSION: Napabucasin is a radiosensitizer with a novel mechanism of action: increasing ROS production and inhibiting angiogenesis. Clinical trials testing the addition of napabucasin to chemoradiotherapy in rectal cancer are needed.


Subject(s)
Benzofurans/administration & dosage , Chemoradiotherapy/methods , Naphthoquinones/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Rectal Neoplasms/metabolism , Rectal Neoplasms/therapy , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Reactive Oxygen Species/metabolism , Rectal Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/radiation effects , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
Amino Acids ; 52(9): 1299-1306, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32918615

ABSTRACT

Raf-1 kinase inhibitory protein (RKIP) acts as a tumor cell metastasis suppressor and prognostic indicator for survival in various cancers. Its use is predicted to improve therapy for various malignancies, including colorectal cancer (CRC). RKIP, frequently denoted as phosphatidylethanolamine-binding protein 1, is expressed in all normal mammalian tissues. RKIP functions as an inhibitor of the Raf-1, PI-3K, and MAP kinase (MAPK) pathways. In this study, we found that resveratrol induced the expression of RKIP at protein levels. To elucidate the structural basis of the interaction between resveratrol and RKIP, we performed computational studies that explore the binding affinity and ligand efficacy of resveratrol against RKIP. This study reveals the prognostic significance of RKIP metastasis suppressor activity against CRC and its structural arrangements during drug-target interactions.


Subject(s)
Antioxidants/pharmacology , Colorectal Neoplasms/drug therapy , Phosphatidylethanolamine Binding Protein/metabolism , Resveratrol/pharmacology , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Phosphatidylethanolamine Binding Protein/chemistry , Protein Conformation , Tumor Cells, Cultured
20.
Mol Cell Biochem ; 472(1-2): 95-103, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32562168

ABSTRACT

Protein-protein interactions (PPIs) are important biochemical processes that represent a major challenge in modern biology. Current approaches, which include high-throughput screening and computer aided ligand design, have limitations regarding the identification of hit matter. This current investigation focuses on computational study for protein-protein docking of hypoxia inducible factor-1α (HIF-1α), a tumor inducible factor, and Raf-1 kinase inhibitory protein (RKIP), a tumor metastasis suppressor. These are individually crystallized structures of interacting proteins, which interact to generate a conformational space. HIF activity in pancreatic tumors is determined by hypoxia and HIF-1α subunit availability. RKIP can be used as a prognostic indicator in a number of tumors. The interaction of RKIP with HIF-1α protects against pancreatic cancer (PC) metastasis by inhibiting its hypoxia function. We have explored the binding affinity between both the proteins with the HADDOCK (high ambiguity driven protein-protein docking) server, which determined that 158 structures in 11 clusters represent 79.0% of water-refined models. Of the best 10 clusters, the structures of cluster 2 were found to be better, as they had the lowest Z-score. Further supporting HIF-1α-RKIP interaction, pulldown assay has shown dissociation of RKIP from HIF-1α after CoCl2 treatment in both PC cell lines.


Subject(s)
Computational Biology/methods , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Docking Simulation , Pancreatic Neoplasms/pathology , Phosphatidylethanolamine Binding Protein/chemistry , Phosphatidylethanolamine Binding Protein/metabolism , Protein Interaction Domains and Motifs , Crystallography, X-Ray , Humans , Pancreatic Neoplasms/metabolism , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL