Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 324(1): G24-G37, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36410023

RESUMO

Single immunoglobulin interleukin-1-related receptor (SIGIRR), toll-interacting protein (TOLLIP), and A20 are major inhibitors of toll-like receptor (TLR) signaling induced postnatally in the neonatal intestine. Short-chain fatty acids (SCFAs), fermentation products of indigestible carbohydrates produced by symbiotic bacteria, inhibit intestinal inflammation. Herein, we investigated the mechanisms by which SCFAs regulate SIGIRR, A20, and TOLLIP expression and mitigate experimental necrotizing enterocolitis (NEC). Butyrate induced NOTCH activation by repressing sirtuin 1 (SIRT1)-mediated deacetylation of the Notch intracellular domain (NICD) in human intestinal epithelial cells (HIECs). Overexpression of NICD induced SIGIRR, A20, and TOLLIP expression. Chromatin immunoprecipitation revealed that butyrate-induced NICD binds to the SIGIRR, A20, and TOLLIP gene promoters. Notch1-shRNA suppressed butyrate-induced SIGIRR/A20 upregulation in mouse enteroids and HIEC. Flagellin (TLR5 agonist)-induced inflammation in HIEC was inhibited by butyrate in a SIGIRR-dependent manner. Neonatal mice fed butyrate had increased NICD, A20, SIGIRR, and TOLLIP expression in the ileal epithelium. Butyrate inhibited experimental NEC-induced intestinal apoptosis, cytokine expression, and histological injury. Our data suggest that SCFAs can regulate the expression of the major negative regulators of TLR signaling in the neonatal intestine through Notch1 and ameliorate experimental NEC. Enteral SCFAs supplementation in preterm infants provides a promising bacteria-free, therapeutic option for NEC.NEW & NOTEWORTHY Short-chain fatty acids (SCFAs), such as propionate and butyrate, metabolites produced by symbiotic gut bacteria are known to be anti-inflammatory, but the mechanisms by which they protect against NEC are not fully understood. In this study, we reveal that SCFAs regulate intestinal inflammation by inducing the key TLR and IL1R inhibitors, SIGIRR and A20, through activation of the pluripotent transcriptional factor NOTCH1. Butyrate-mediated SIGIRR and A20 induction represses experimental NEC in the neonatal intestine.


Assuntos
Enterocolite Necrosante , Recém-Nascido , Animais , Camundongos , Humanos , Enterocolite Necrosante/tratamento farmacológico , Enterocolite Necrosante/prevenção & controle , Enterocolite Necrosante/genética , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Recém-Nascido Prematuro , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Ácidos Graxos Voláteis/farmacologia , Ácidos Graxos Voláteis/metabolismo , Butiratos/metabolismo , Imunoglobulinas/metabolismo , Interleucina-1/metabolismo , Receptor Notch1/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
2.
Molecules ; 27(4)2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35209196

RESUMO

Traditionally, medicinal plants have long been used as a natural therapy. Plant-derived extracts or phytochemicals have been exploited as food additives and for curing many health-related ailments. The secondary metabolites produced by many plants have become an integral part of human health and have strengthened the value of plant extracts as herbal medicines. To fulfil the demand of health care systems, food and pharmaceutical industries, interest in the cultivation of precious medicinal plants to harvest bio-active compounds has increased considerably worldwide. To achieve maximum biomass and yield, growers generally apply chemical fertilizers which have detrimental impacts on the growth, development and phytoconstituents of such therapeutically important plants. Application of beneficial rhizosphere microbiota is an alternative strategy to enhance the production of valuable medicinal plants under both conventional and stressed conditions due to its low cost, environmentally friendly behaviour and non-destructive impact on fertility of soil, plants and human health. The microbiological approach improves plant growth by various direct and indirect mechanisms involving the abatement of various abiotic stresses. Given the negative impacts of fertilizers and multiple benefits of microbiological resources, the role of plant growth promoting rhizobacteria (PGPR) in the production of biomass and their impact on the quality of bio-active compounds (phytochemicals) and mitigation of abiotic stress to herbal plants have been described in this review. The PGPR based enhancement in the herbal products has potential for use as a low cost phytomedicine which can be used to improve health care systems.


Assuntos
Bactérias/crescimento & desenvolvimento , Bioprospecção , Produtos Agrícolas , Compostos Fitoquímicos , Plantas Medicinais , Rizosfera , Microbiologia do Solo , Produtos Agrícolas/química , Produtos Agrícolas/crescimento & desenvolvimento , Produtos Agrícolas/microbiologia , Humanos , Compostos Fitoquímicos/química , Compostos Fitoquímicos/uso terapêutico , Plantas Medicinais/química , Plantas Medicinais/crescimento & desenvolvimento , Plantas Medicinais/microbiologia
3.
Cells ; 10(7)2021 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-34206989

RESUMO

Honokiol (HNK) is a biphenolic compound that has been used in traditional medicine for treating various ailments, including cancers. In this study, we determined the effect of HNK on colon cancer cells in culture and in a colitis-associated cancer model. HNK treatment inhibited proliferation and colony formation while inducing apoptosis. In addition, HNK suppressed colonosphere formation. Molecular docking suggests that HNK interacts with reserve stem cell marker protein DCLK1, with a binding energy of -7.0 Kcal/mol. In vitro kinase assays demonstrated that HNK suppressed the DCLK1 kinase activity. HNK also suppressed the expression of additional cancer stem cell marker proteins LGR5 and CD44. The Hippo signaling pathway is active in intestinal stem cells. In the canonical pathway, YAP1 is phosphorylated at Ser127 by upstream Mst1/2 and Lats1/2. This results in the sequestration of YAP1 in the cytoplasm, thereby not allowing YAP1 to translocate to the nucleus and interact with TEAD1-4 transcription factors to induce gene expression. However, HNK suppressed Ser127 phosphorylation in YAP1, but the protein remains sequestered in the cytoplasm. We further determined that this occurs by YAP1 interacting with PUMA. To determine if this also occurs in vivo, we performed studies in an AOM/DSS induced colitis-associated cancer model. HNK administered by oral gavage at a dose of 5mg/kg bw for 24 weeks demonstrated a significant reduction in the expression of YAP1 and TEAD1 and in the stem marker proteins. Together, these data suggest that HNK prevents colon tumorigenesis in part by inducing PUMA-YAP1 interaction and cytoplasmic sequestration, thereby suppressing the oncogenic YAP1 activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Compostos de Bifenilo/farmacologia , Carcinogênese/patologia , Neoplasias do Colo/patologia , Lignanas/farmacologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colite/complicações , Quinases Semelhantes a Duplacortina , Regulação para Baixo/efeitos dos fármacos , Via de Sinalização Hippo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Endogâmicos ICR , Modelos Biológicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Proteínas de Sinalização YAP
4.
J Immunol Res ; 2021: 8121407, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34046506

RESUMO

To mimic Alzheimer's disease, transgenic mice overexpressing the amyloid precursor protein (APP) were used in this study. We hypothesize that the neuroprotective effects of ETAS®50, a standardized extract of Asparagus officinalis stem produced by Amino Up Co., Ltd. (Sapporo, Japan), are linked to the inhibition of the apoptosis cascade through an enhancement of the stress-response proteins: heat shock proteins (HSPs). APP-overexpressing mice (double-transgenic APP and PS1 mouse strains with a 129s6 background), ages 6-8 weeks old, and weighing 20-24 grams were successfully bred in our laboratory. The animals were divided into 5 groups. APP-overexpressing mice and wild-type (WT) mice were pretreated with ETAS®50 powder (50% elemental ETAS and 50% destrin) at 200 mg/kg and 1000 mg/kg body weight. Saline, the vehicle for ETAS®50, was administered in APP-overexpressing mice and WT mice. ETAS®50 and saline were administered by gavage daily for 1 month. Cognitive assessments, using the Morris Water Maze, demonstrated that memory was recovered following ETAS®50 treatment as compared to nontreated APP mice. At euthanization, the brain was removed and HSPs, amyloid ß, tau proteins, and caspase-3 were evaluated through immunofluorescence staining with the appropriate antibodies. Our data indicate that APP mice have cognitive impairment along with elevated amyloid ß, tau proteins, and caspase-3. ETAS®50 restored cognitive function in these transgenic mice, increased both HSP70 and HSP27, and attenuated pathogenic level of amyloid ß, tau proteins, and caspsase-3 leading to neuroprotection. Our results were confirmed with a significant increase in HSP70 gene expression in the hippocampus.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Asparagus/química , Fármacos Neuroprotetores/administração & dosagem , Extratos Vegetais/administração & dosagem , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Proteínas de Choque Térmico HSP27/análise , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP70/análise , Proteínas de Choque Térmico HSP70/metabolismo , Hipocampo/patologia , Humanos , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Teste do Labirinto Aquático de Morris/efeitos dos fármacos , Presenilina-1/genética
5.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008767

RESUMO

Decreases in short-chain-fatty-acids (SCFAs) are linked to inflammatory bowel disease (IBD). Yet, the mechanisms through which SCFAs promote wound healing, orchestrated by intestinal stem cells, are poorly understood. We discovered that, in mice with Citrobacter rodentium (CR)-induced infectious colitis, treatment with Pectin and Tributyrin diets reduced the severity of colitis by restoring Firmicutes and Bacteroidetes and by increasing mucus production. RNA-seq in young adult mouse colon (YAMC) cells identified higher expression of Lgr4, Lgr6, DCLK1, Muc2, and SIGGIR after Butyrate treatment. Lineage tracing in CR-infected Lgr5-EGFP-IRES-CreERT2/ROSA26-LacZ (Lgr5-R) mice also revealed an expansion of LacZ-labeled Lgr5(+) stem cells in the colons of both Pectin and Tributyrin-treated mice compared to control. Interestingly, gut microbiota was required for Pectin but not Tributyrin-induced Lgr5(+) stem cell expansion. YAMC cells treated with sodium butyrate exhibited increased Lgr5 promoter reporter activity due to direct Butyrate binding with Lgr5 at -4.0 Kcal/mol, leading to thermal stabilization. Upon ChIP-seq, H3K4me3 increased near Lgr5 transcription start site that contained the consensus binding motif for a transcriptional activator of Lgr5 (SPIB). Thus, a multitude of effects on gut microbiome, differential gene expression, and/or expansion of Lgr5(+) stem cells seem to underlie amelioration of colitis following dietary intervention.


Assuntos
Colite/microbiologia , Colite/patologia , Dieta , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Microbiota , Células-Tronco/patologia , Animais , Biodiversidade , Butiratos/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Citrobacter rodentium/fisiologia , Epitélio/patologia , Fermentação , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Mucina-2/metabolismo , Pectinas/farmacologia , Regiões Promotoras Genéticas/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regeneração/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Triglicerídeos/farmacologia
6.
Pediatr Res ; 88(4): 546-555, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32053825

RESUMO

BACKGROUND: Exaggerated Toll-like receptor (TLR) signaling and intestinal dysbiosis are key contributors to necrotizing enterocolitis (NEC). Lactobacillus rhamnosus GG (LGG) decreases NEC in preterm infants, but underlying mechanisms of protection remain poorly understood. We hypothesized that LGG alleviates dysbiosis and upregulates TLR inhibitors to protect against TLR-mediated gut injury. METHODS: Effects of LGG (low- and high-dose) on intestinal pro-inflammatory TLR signaling and injury in neonatal mice subjected to formula feeding (FF) and NEC were determined. 16S sequencing of stool and expression of anti-TLR mediators SIGIRR (single immunoglobulin interleukin-1-related receptor) and A20 were analyzed. RESULTS: FF induced mild intestinal injury with increased expression of interleukin-1ß (IL-1ß) and Kupffer cell (KC) (mouse homolog of IL-8) compared to controls. LGG decreased IL-1ß and KC in association with attenuated TLR signaling and increased SIGIRR and A20 expression in a dose-dependent manner. Low- and high-dose LGG had varying effects on gut microbiome despite both doses providing gut protection. Subsequent experiments of LGG on NEC revealed that pro-inflammatory TLR signaling and intestinal injury were also decreased, and SIGIRR and A20 expression increased, in a dose-dependent manner with LGG pre-treatment. CONCLUSIONS: LGG protects against intestinal TLR-mediated injury by upregulating TLR inhibitors without major changes in gut microbiome composition.


Assuntos
Enterocolite Necrosante/metabolismo , Intestinos/lesões , Lacticaseibacillus rhamnosus/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Citocinas/metabolismo , Suplementos Nutricionais , Microbioma Gastrointestinal , Íleo/patologia , Fórmulas Infantis , Inflamação , Mucosa Intestinal/metabolismo , Células de Kupffer/citologia , Camundongos , Camundongos Endogâmicos C57BL , Probióticos , RNA Ribossômico 16S/metabolismo , Transdução de Sinais
7.
PLoS One ; 10(8): e0135561, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26270561

RESUMO

Gastrointestinal (GI) mucosal damage is a devastating adverse effect of radiation therapy. We have recently reported that expression of Dclk1, a Tuft cell and tumor stem cell (TSC) marker, 24h after high dose total-body gamma-IR (TBI) can be used as a surrogate marker for crypt survival. Dietary pectin has been demonstrated to possess chemopreventive properties, whereas its radioprotective property has not been studied. The aim of this study was to determine the effects of dietary pectin on ionizing radiation (IR)-induced intestinal stem cell (ISC) deletion, crypt and overall survival following lethal TBI. C57BL/6 mice received a 6% pectin diet and 0.5% pectin drinking water (pre-IR mice received pectin one week before TBI until death; post-IR mice received pectin after TBI until death). Animals were exposed to TBI (14 Gy) and euthanized at 24 and 84h post-IR to assess ISC deletion and crypt survival respectively. Animals were also subjected to overall survival studies following TBI. In pre-IR treatment group, we observed a three-fold increase in ISC/crypt survival, a two-fold increase in Dclk1+ stem cells, increased overall survival (median 10d vs. 7d), and increased expression of Dclk1, Msi1, Lgr5, Bmi1, and Notch1 (in small intestine) post-TBI in pectin treated mice compared to controls. We also observed increased survival of mice treated with pectin (post-IR) compared to controls. Dietary pectin is a radioprotective agent; prevents IR-induced deletion of potential reserve ISCs; facilitates crypt regeneration; and ultimately promotes overall survival. Given the anti-cancer activity of pectin, our data support a potential role for dietary pectin as an agent that can be administered to patients receiving radiation therapy to protect against radiation-induces mucositis.


Assuntos
Mucosite/prevenção & controle , Pectinas/administração & dosagem , Lesões Experimentais por Radiação/prevenção & controle , Protetores contra Radiação/administração & dosagem , Células-Tronco/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Suplementos Nutricionais/análise , Quinases Semelhantes a Duplacortina , Feminino , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mucosite/dietoterapia , Mucosite/etiologia , Mucosite/patologia , Pectinas/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Lesões Experimentais por Radiação/dietoterapia , Lesões Experimentais por Radiação/patologia , Protetores contra Radiação/farmacologia , Células-Tronco/metabolismo , Células-Tronco/efeitos da radiação , Análise de Sobrevida , Irradiação Corporal Total
8.
Nat Prod Res ; 28(23): 2081-101, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24912126

RESUMO

Andrographispaniculata, referred to as 'Kalmegh' in ancient texts, is regarded as a nostrum of the modern world. Oral administration of the leaves of this plant is effective in the treatment of upper respiratory tract infections, liver toxicity and a variety of other ailments. The practices of adulteration in the original material of this drug have made it imperative to develop techniques to verify the authenticity of the products of this plant for quality assurance. Andrographolide, a bitter diterpenoid, is the major active principle present in the plant, and therefore its determination and quantification in the plant extracts are important. This article provides a detailed account of various extraction and chromatographic techniques used for the identification, isolation and quantification of the various active principles of A. paniculata. Pharmacological properties and phytochemistry of this plant and the physico-chemical properties of these bio-compounds have also been outlined.


Assuntos
Andrographis/química , Diterpenos , Extratos Vegetais/química , Plantas Medicinais/química , Diterpenos/análise , Diterpenos/química , Diterpenos/isolamento & purificação , Diterpenos/farmacologia , Estrutura Molecular
9.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G1-15, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24789206

RESUMO

The human intestinal tract harbors a complex ecosystem of commensal bacteria that play a fundamental role in the well-being of their host. There is a general consensus that diet rich in plant-based foods has many advantages in relation to the health and well-being of an individual. In adults, diets that have a high proportion of fruit and vegetables and a low consumption of meat are associated with a highly diverse microbiota and are defined by a greater abundance of Prevotella compared with Bacteroides, whereas the reverse is associated with a diet that contains a low proportion of plant-based foods. In a philosophical term, our consumption of processed foods, widespread use of antibiotics and disinfectants, and our modern lifestyle may have forever altered our ancient gut microbiome. We may never be able to identify or restore our microbiomes to their ancestral state, but dietary modulation to manipulate specific gut microbial species or groups of species may offer new therapeutic approaches to conditions that are prevalent in modern society, such as functional gastrointestinal disorders, obesity, and age-related nutritional deficiency. We believe that this will become an increasingly important area of health research.


Assuntos
Quimioprevenção/métodos , Dieta , Gastroenteropatias/prevenção & controle , Trato Gastrointestinal/microbiologia , Estilo de Vida , Microbiota , Compostos Fitoquímicos/uso terapêutico , Comportamento de Redução do Risco , Animais , Antibacterianos/efeitos adversos , Dieta/efeitos adversos , Desinfetantes/efeitos adversos , Disbiose , Frutas , Gastroenteropatias/epidemiologia , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Carne , Microbiota/efeitos dos fármacos , Fatores de Risco , Verduras
10.
Artigo em Inglês | MEDLINE | ID: mdl-23533514

RESUMO

Bitter melon fruit is recommended in ancient Indian and Chinese medicine for prevention/treatment of diabetes. However its effects on cancer progression are not well understood. Here, we have determined the efficacy of methanolic extracts of bitter melon on colon cancer stem and progenitor cells. Both, whole fruit (BMW) and skin (BMSk) extracts showed significant inhibition of cell proliferation and colony formation, with BMW showing greater efficacy. In addition, the cells were arrested at the S phase of cell cycle. Moreover, BMW induced the cleavage of LC3B but not caspase 3/7, suggesting that the cells were undergoing autophagy and not apoptosis. Further confirmation of autophagy was obtained when western blots showed reduced Bcl-2 and increased Beclin-1, Atg 7 and 12 upon BMW treatment. BMW reduced cellular ATP levels coupled with activation of AMP activated protein kinase; on the other hand, exogenous additions of ATP lead to revival of cell proliferation. Finally, BMW treatment results in a dose-dependent reduction in the number and size of colonospheres. The extracts also decreased the expression of DCLK1 and Lgr5, markers of quiescent, and activated stem cells. Taken together, these results suggest that the extracts of bitter melon can be an effective preventive/therapeutic agent for colon cancer.

11.
Environ Sci Pollut Res Int ; 20(9): 6039-49, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23529401

RESUMO

In a hydroponic culture, experiments were performed to study the influence of potassium (K) supplementation (0, 20, 40, 60, 80, and 100 mg L(-1)) on the arsenic (As; 0, 8, and 10 mg L(-1))-accrued changes in growth traits (plant biomass, root-shoot length) and the contents of lepidine, As and K, in garden cress (Lepidium sativum Linn.) at 10 days after treatment. The changes in these traits were correlated with shoot proline content, protein profile, and the activities of antioxidant enzymes namely superoxide dismutase (SOD, EC 1.15.1.1), catalase (CAT, EC 1.11.1.6), glutathione reductase (GR, EC 1.8.1.7), and ascorbate peroxidase (APX, EC 1.11.1.11). In general, As-alone treatments significantly decreased the growth traits but lead to significant enhancements in shoot proline and enzyme activities. K-supplementation to As-treated L. sativum seedlings decreased shoot-As content, reduced As-induced decreases in growth traits but enhanced the content of shoot proline, and the activities of the studied enzymes maximally with K100 + As8 and As10 mg L(-1). Both 8 and 10 mg L(-1) of As drastically downregulated the shoot proteins ranging from 43-65 kDa. With As10 mg L(-1), there was a total depletion of protein bands below 23 kDa; however, K80 mg L(-1) maximally recovered and upregulated the protein bands. Additionally, protein bands were downregulated (at par with As-alone treatment) above K80 mg L(-1) level. Interestingly, As-stress increased lepidine content in a dose-dependent manner which was further augmented with the K-supplementation. It is suggested that K protects L. sativum against As-toxicity by decreasing its accumulation and strengthening antioxidant defense system and protein stability.


Assuntos
Arsênio/toxicidade , Lepidium sativum/efeitos dos fármacos , Potássio/metabolismo , Poluentes do Solo/toxicidade , Arsênio/química , Relação Dose-Resposta a Droga , Lepidium sativum/crescimento & desenvolvimento , Lepidium sativum/metabolismo , Proteínas de Plantas/metabolismo , Potássio/administração & dosagem , Prolina/química , Poluentes do Solo/química
12.
Mol Cancer Ther ; 11(4): 963-72, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22319203

RESUMO

Cancer stem cells are implicated in resistance to ionizing radiation (IR) and chemotherapy. Honokiol, a biphenolic compound has been used in traditional Chinese medicine for treating various ailments. In this study, we determined the ability of honokiol to enhance the sensitivity of colon cancer stem cells to IR. The combination of honokiol and IR suppressed proliferation and colony formation while inducing apoptosis of colon cancer cells in culture. There were also reduced numbers and size of spheroids, which was coupled with reduced expression of cancer stem cell marker protein DCLK1. Flow cytometry studies confirmed that the honokiol-IR combination reduced the number of DCLK1+ cells. In addition, there were reduced levels of activated Notch-1, its ligand Jagged-1, and the downstream target gene Hes-1. Furthermore, expression of components of the Notch-1 activating γ-secretase complex, presenilin 1, nicastrin, Pen2, and APH-1 was also suppressed. On the other hand, the honokiol effects were mitigated when the Notch intracellular domain was expressed. To determine the effect of honokiol-IR combination on tumor growth in vivo, nude mice tumor xenografts were administered honokiol intraperitoneally and exposed to IR. The honokiol-IR combination significantly inhibited tumor xenograft growth. In addition, there were reduced levels of DCLK1 and the Notch signaling-related proteins in the xenograft tissues. Together, these data suggest that honokiol is a potent inhibitor of colon cancer growth that targets the stem cells by inhibiting the γ-secretase complex and the Notch signaling pathway. These studies warrant further clinical evaluation for the combination of honokiol and IR for treating colon cancers.


Assuntos
Compostos de Bifenilo/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/radioterapia , Medicamentos de Ervas Chinesas/farmacologia , Lignanas/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Terapia Combinada , Regulação para Baixo , Células HCT116 , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Óxido Nítrico Sintase/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Am J Physiol Gastrointest Liver Physiol ; 301(5): G929-37, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21836060

RESUMO

DNA mismatch repair is required for correcting any mismatches that are created during replication and recombination, and a defective mismatch repair system contributes to DNA damage-induced growth arrest. The colorectal cancer cell line HCT116 is known to have a mutation in the hMLH1 mismatch repair gene resulting in microsatellite instability and defective mismatch repair. Honokiol is a biphenolic compound that has been used in traditional Chinese medicine for treating various ailments including cancer. This study was designed to test the hypothesis that honokiol enhances the radiosensitivity of cancer cells with mismatch repair defect (HCT116) compared with those that are mismatch repair proficient (HCT116-CH3). We first determined that the combination of honokiol and γ-irradiation treatment resulted in dose-dependent inhibition of proliferation and colony formation in both cell lines. However, the effects were more pronounced in HCT116 cells. Similarly, the combination induced higher levels of apoptosis (caspase 3 activation, Bax to Bcl2 ratio) in the HCT116 cells compared with HCT116-CH3 cells. Cell cycle analyses revealed higher levels of dead cells in HCT116 cells. The combination treatment reduced expression of cyclin A1 and D1 and increased phosphorylated p53 in both cell lines, although there were significantly lower amounts of phosphorylated p53 in the HCT116-CH3 cells, suggesting that high levels of hMLH1 reduce radiosensitivity. These data demonstrate that honokiol is highly effective in radiosensitizing colorectal cancer cells, especially those with a mismatch repair defect.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Compostos de Bifenilo/farmacologia , Proliferação de Células/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Lignanas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Células HCT116 , Humanos , Tolerância a Radiação/genética , Células Tumorais Cultivadas
14.
Am J Physiol Gastrointest Liver Physiol ; 291(2): G318-30, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16574993

RESUMO

We investigated the effects of PKC-stimulating 12-deoxyphorbol 13-phenylacetate 20-acetate (DOPPA) and phorbol 12-myristate 13-acetate (PMA) phorbol esters on cAMP-dependent, forskolin (FSK)-stimulated, short-circuit Cl- current (ISC-cAMP) generation by colonocyte monolayers. These agonists elicited different actions depending on their dose and incubation time; PMA effects at the onset (<5 min) were independent of cAMP agonist and were characterized by transient anion-dependent transcellular and apical membrane ISC generation. DOPPA failed to elicit similar responses. Whereas chronic (24 h) exposure to both agents inhibited FSK-stimulated transcellular and apical membrane ISC-cAMP, the effects of DOPPA were more complex: this conventional PKC-beta-specific agonist also stimulated Ba2+-sensitive basolateral membrane-dependent facilitation of transcellular ISC-cAMP. PMA did not elicit a similar phenomenon. Prolonged exposure to high-dose PMA but not DOPPA led to apical membrane ISC-cAMP recovery. Changes in PKC alpha-, beta1-, gamma-, and epsilon-isoform membrane partitioning and expression correlated with these findings. PMA-induced transcellular ISC correlated with PKC-alpha membrane association, whereas low doses of both agents inhibited transcellular and apical membrane ISC-cAMP, increased PKC-beta1, decreased PKC-beta2 membrane association, and caused reciprocal changes in isoform mass. During the apical membrane ISC-cAMP recovery after prolonged high-dose PMA exposure, an almost-complete depletion of cellular PKC-beta1 and a significant reduction in PKC-epsilon mass occurred. Thus activated PKC-beta1 and/or PKC-epsilon prevented, whereas activated PKC-alpha facilitated, apical membrane ISC-cAMP. PKC-beta-dependent augmentation of transcellular ISC-cAMP at the level of the basolateral membrane demonstrated that transport events with geographically distinct subcellular membranes can be independently regulated by the PKC beta-isoform.


Assuntos
Colo/fisiologia , AMP Cíclico/metabolismo , Potenciais da Membrana/fisiologia , Proteína Quinase C/metabolismo , Ativação Enzimática , Células HT29 , Humanos , Ativação do Canal Iônico/fisiologia , Proteína Quinase C beta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA