Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26590477

ABSTRACT

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Subject(s)
Genetic Heterogeneity , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction , Tumor Microenvironment/genetics
2.
Semin Cancer Biol ; 35 Suppl: S151-S184, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25951989

ABSTRACT

Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.


Subject(s)
Antineoplastic Agents/therapeutic use , Inflammation/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Cell Transformation, Neoplastic/drug effects , Genetic Heterogeneity/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/drug effects
3.
Semin Cancer Biol ; 35 Suppl: S78-S103, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25936818

ABSTRACT

Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.


Subject(s)
Apoptosis/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25865775

ABSTRACT

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Subject(s)
Carcinogenesis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/genetics , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Cell Proliferation/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Signal Transduction , Tumor Microenvironment/drug effects
5.
Semin Cancer Biol ; 35 Suppl: S25-S54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25892662

ABSTRACT

Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.


Subject(s)
Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Neoplasms/pathology , Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects
6.
Semin Cancer Biol ; 35 Suppl: S104-S128, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25869441

ABSTRACT

One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.


Subject(s)
Cell Proliferation/genetics , Cellular Senescence/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Genomic Instability/drug effects , Humans , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Telomerase/drug effects , Telomerase/genetics , Tumor Suppressor Protein p53/genetics
7.
Semin Cancer Biol ; 35 Suppl: S5-S24, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25869442

ABSTRACT

Genomic instability can initiate cancer, augment progression, and influence the overall prognosis of the affected patient. Genomic instability arises from many different pathways, such as telomere damage, centrosome amplification, epigenetic modifications, and DNA damage from endogenous and exogenous sources, and can be perpetuating, or limiting, through the induction of mutations or aneuploidy, both enabling and catastrophic. Many cancer treatments induce DNA damage to impair cell division on a global scale but it is accepted that personalized treatments, those that are tailored to the particular patient and type of cancer, must also be developed. In this review, we detail the mechanisms from which genomic instability arises and can lead to cancer, as well as treatments and measures that prevent genomic instability or take advantage of the cellular defects caused by genomic instability. In particular, we identify and discuss five priority targets against genomic instability: (1) prevention of DNA damage; (2) enhancement of DNA repair; (3) targeting deficient DNA repair; (4) impairing centrosome clustering; and, (5) inhibition of telomerase activity. Moreover, we highlight vitamin D and B, selenium, carotenoids, PARP inhibitors, resveratrol, and isothiocyanates as priority approaches against genomic instability. The prioritized target sites and approaches were cross validated to identify potential synergistic effects on a number of important areas of cancer biology.


Subject(s)
Genomic Instability/drug effects , Neoplasms/diet therapy , Neoplasms/genetics , Centrosome/metabolism , DNA Damage/genetics , DNA Repair/genetics , Diet , Genomic Instability/genetics , Humans , Neoplasms/pathology , Prognosis , Telomerase/antagonists & inhibitors , Telomerase/genetics
8.
Semin Cancer Biol ; 35 Suppl: S185-S198, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25818339

ABSTRACT

Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.


Subject(s)
Carcinogenesis/immunology , Immune Evasion , Neoplasms/immunology , Neoplasms/therapy , Antigen Presentation/immunology , Carcinogenesis/drug effects , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Neoplasms/pathology , Phytochemicals/therapeutic use , T-Lymphocytes, Regulatory/immunology , Tumor Escape/drug effects , Tumor Escape/immunology
9.
Semin Cancer Biol ; 35 Suppl: S55-S77, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25749195

ABSTRACT

The evasion of anti-growth signaling is an important characteristic of cancer cells. In order to continue to proliferate, cancer cells must somehow uncouple themselves from the many signals that exist to slow down cell growth. Here, we define the anti-growth signaling process, and review several important pathways involved in growth signaling: p53, phosphatase and tensin homolog (PTEN), retinoblastoma protein (Rb), Hippo, growth differentiation factor 15 (GDF15), AT-rich interactive domain 1A (ARID1A), Notch, insulin-like growth factor (IGF), and Krüppel-like factor 5 (KLF5) pathways. Aberrations in these processes in cancer cells involve mutations and thus the suppression of genes that prevent growth, as well as mutation and activation of genes involved in driving cell growth. Using these pathways as examples, we prioritize molecular targets that might be leveraged to promote anti-growth signaling in cancer cells. Interestingly, naturally occurring phytochemicals found in human diets (either singly or as mixtures) may promote anti-growth signaling, and do so without the potentially adverse effects associated with synthetic chemicals. We review examples of naturally occurring phytochemicals that may be applied to prevent cancer by antagonizing growth signaling, and propose one phytochemical for each pathway. These are: epigallocatechin-3-gallate (EGCG) for the Rb pathway, luteolin for p53, curcumin for PTEN, porphyrins for Hippo, genistein for GDF15, resveratrol for ARID1A, withaferin A for Notch and diguelin for the IGF1-receptor pathway. The coordination of anti-growth signaling and natural compound studies will provide insight into the future application of these compounds in the clinical setting.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Neoplasms/genetics , Neoplasms/therapy , Signal Transduction , DNA-Binding Proteins , Growth Differentiation Factor 15/genetics , Hippo Signaling Pathway , Humans , Kruppel-Like Transcription Factors/genetics , Molecular Targeted Therapy , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/genetics , Somatomedins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
10.
Semin Cancer Biol ; 35 Suppl: S224-S243, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25600295

ABSTRACT

Deregulation of angiogenesis--the growth of new blood vessels from an existing vasculature--is a main driving force in many severe human diseases including cancer. As such, tumor angiogenesis is important for delivering oxygen and nutrients to growing tumors, and therefore considered an essential pathologic feature of cancer, while also playing a key role in enabling other aspects of tumor pathology such as metabolic deregulation and tumor dissemination/metastasis. Recently, inhibition of tumor angiogenesis has become a clinical anti-cancer strategy in line with chemotherapy, radiotherapy and surgery, which underscore the critical importance of the angiogenic switch during early tumor development. Unfortunately the clinically approved anti-angiogenic drugs in use today are only effective in a subset of the patients, and many who initially respond develop resistance over time. Also, some of the anti-angiogenic drugs are toxic and it would be of great importance to identify alternative compounds, which could overcome these drawbacks and limitations of the currently available therapy. Finding "the most important target" may, however, prove a very challenging approach as the tumor environment is highly diverse, consisting of many different cell types, all of which may contribute to tumor angiogenesis. Furthermore, the tumor cells themselves are genetically unstable, leading to a progressive increase in the number of different angiogenic factors produced as the cancer progresses to advanced stages. As an alternative approach to targeted therapy, options to broadly interfere with angiogenic signals by a mixture of non-toxic natural compound with pleiotropic actions were viewed by this team as an opportunity to develop a complementary anti-angiogenesis treatment option. As a part of the "Halifax Project" within the "Getting to know cancer" framework, we have here, based on a thorough review of the literature, identified 10 important aspects of tumor angiogenesis and the pathological tumor vasculature which would be well suited as targets for anti-angiogenic therapy: (1) endothelial cell migration/tip cell formation, (2) structural abnormalities of tumor vessels, (3) hypoxia, (4) lymphangiogenesis, (5) elevated interstitial fluid pressure, (6) poor perfusion, (7) disrupted circadian rhythms, (8) tumor promoting inflammation, (9) tumor promoting fibroblasts and (10) tumor cell metabolism/acidosis. Following this analysis, we scrutinized the available literature on broadly acting anti-angiogenic natural products, with a focus on finding qualitative information on phytochemicals which could inhibit these targets and came up with 10 prototypical phytochemical compounds: (1) oleanolic acid, (2) tripterine, (3) silibinin, (4) curcumin, (5) epigallocatechin-gallate, (6) kaempferol, (7) melatonin, (8) enterolactone, (9) withaferin A and (10) resveratrol. We suggest that these plant-derived compounds could be combined to constitute a broader acting and more effective inhibitory cocktail at doses that would not be likely to cause excessive toxicity. All the targets and phytochemical approaches were further cross-validated against their effects on other essential tumorigenic pathways (based on the "hallmarks" of cancer) in order to discover possible synergies or potentially harmful interactions, and were found to generally also have positive involvement in/effects on these other aspects of tumor biology. The aim is that this discussion could lead to the selection of combinations of such anti-angiogenic compounds which could be used in potent anti-tumor cocktails, for enhanced therapeutic efficacy, reduced toxicity and circumvention of single-agent anti-angiogenic resistance, as well as for possible use in primary or secondary cancer prevention strategies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Blood Vessels/drug effects , Blood Vessels/growth & development , Blood Vessels/pathology , Cell Proliferation/drug effects , Humans , Immunotherapy , Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control
11.
Interdiscip Sci ; 5(1): 53-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23605640

ABSTRACT

The interactions between the molecules and DNA shape up an avenue for DNA targeted therapeutics. For the first time, brazilin, a major component of Caesalpinia sappan L., has been investigated for its interaction with natural and synthetic DNA. Detailed analyses of the binding property of brazilin dye with DNA by UV-vis, FTIR and Circular Dichroism were carried out. In addition, in silico studies have been conducted via tools of energy minimization and ligand optimization using Yasara and Argus Lab softwares along with the molecular docking server integrating Auto Dock, Mavin and Mopac. Results show that brazilin dye has commendable proficiency in being moulded as a binder with DNA. The specificity of the dye to stain nuclei in tissue sections positively indicates its interaction with nucleic acid. As the intracellular target for the majority of anticancer and antibiotic drugs is DNA, the study on the interaction between molecules like brazilin and DNA has great significance and implications in several biological applications.


Subject(s)
Benzopyrans/metabolism , Caesalpinia/chemistry , Coloring Agents/metabolism , DNA/metabolism , Drug Discovery/methods , Models, Molecular , Benzopyrans/analysis , Benzopyrans/chemistry , Circular Dichroism , Coloring Agents/analysis , Coloring Agents/chemistry , Molecular Structure , Protein Binding , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
12.
Appl Biochem Biotechnol ; 167(4): 885-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22639367

ABSTRACT

For the first time, interaction between non-toxic anthraquinone morindone with both natural and synthetic DNA duplexes has been demonstrated in this paper. Detailed analyses of the binding of morindone with DNA via UV-vis, FTIR, and circular dichroism spectroscopies were carried out. In addition, bioinformatics tools have been employed to scrutinize the binding of the dye with DNA in silico. Results represent morindone to be a better binder with a score of -5.79 compared to EtBr (known mutagenic intercalator) recorded at -5.02. Further interaction is accentuated by the microscope-assisted evidence of nuclear specific staining of tissues by morindone. The electrophoretic analysis reveals the efficacy endowed within morindone dye in rendering protection to DNA exposed to H(2)O(2) damage and thereby conferring it safe to the nucleic acid. As DNA is often the target for majority of anticancer and antibiotic drugs, study on the interaction between molecules like morindone and DNA has relevance and implications in several biological applications including cancer therapy. Thus, we propose that morindone can also be harnessed as a diagnostic probe for DNA structure in addition to DNA-directed therapeutics.


Subject(s)
Anthraquinones/pharmacology , Coloring Agents/pharmacology , DNA/chemistry , Intercalating Agents/pharmacology , Models, Molecular , Animals , Anthraquinones/chemistry , Anthraquinones/metabolism , Anthraquinones/toxicity , Cattle , Coloring Agents/chemistry , Coloring Agents/metabolism , Coloring Agents/toxicity , DNA/metabolism , Electrophoresis , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Intercalating Agents/toxicity , Nucleic Acid Conformation , Safety , Spectrum Analysis
13.
Food Chem Toxicol ; 50(6): 2062-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22487463

ABSTRACT

Plant dyes have been in use for coloring and varied purposes since prehistoric times. A red dye found in the roots of plants belonging to genus Morinda is a well recognized coloring ingredient. The dye fraction obtained from the methanolic extract of the roots of Morinda tinctoria was explored for its role in attenuating damages caused by H(2)O(2)-induced oxidative stress. The antioxidant potential of the dye fraction was assessed through DPPH radical scavenging, deoxyribose degradation and inhibition of lipid peroxidation in mice liver. It was subsequently screened for its efficiency in extenuating damage incurred to biomembrane (using erythrocytes and their ghost membranes) and macromolecules (pBR322 DNA, lipids and proteins) from exposure to hydrogen peroxide. In addition, the non-toxic nature of the dye was supported by the histological evaluation conducted on the tissue sections from the major organs of Swiss Albino mice as well as effect on Hep3B cell line (human hepatic carcinoma). The LC-MS confirms the dye fraction to be morindone. Our study strongly suggests that morindone present in the root extracts of M. tinctoria, in addition to being a colorant, definitely holds promise in the pharmaceutical industry.


Subject(s)
Coloring Agents/pharmacology , Membranes/metabolism , Morinda/chemistry , Oxidative Stress/drug effects , Animals , Biphenyl Compounds/chemistry , Chromatography, High Pressure Liquid , Coloring Agents/isolation & purification , DNA/drug effects , Deoxyribose/chemistry , Erythrocyte Membrane/drug effects , Erythrocytes/drug effects , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Hemolysis/drug effects , Humans , Lipid Peroxidation/drug effects , Lipids/chemistry , Liver/drug effects , Liver/metabolism , Mice , Picrates/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Proteins/chemistry , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...