Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Appl Microbiol ; 133(5): 2941-2953, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35938351

RESUMEN

AIMS: The aim of this work was to assess the effects of a probiotic diet on well-being of healthy seniors living in boarding and private homes in Marche Region, Italy. In particular, we focused on the modulation of high-sensitivity C-reactive protein (HsCRP), intestinal microbiota and short-chain fatty acids (SCFAs). METHODS AND RESULTS: Ninety-seven healthy seniors took part in a double-blind, placebo-controlled feeding study (59 fed probiotics, 38 fed placebo) for 6 months. Each volunteer ingested daily one food product or a dietary supplement enriched with Synbio® blend (Synbiotec Srl, Camerino, Italy) or the placebo (control group). Blood and faecal samples were collected before and at the end of the intervention period to perform biochemical and microbiological analyses. The serum HsCRP difference value after 6 months of treatment was significantly higher in the probiotic group than placebo (p < 0.05). After the intervention, a significant increase in faecal lactobacilli and a bifidobacteria increase in more participants were observed in the probiotic group. The 16S NGS analysis on the probiotic group showed a decreasing trend of Proteobacteria at the end of the treatment and conversely, an increasing trend of Actinobacteria and Verrucomicrobia phyla, to which the increase of Akkermansiaceae and Bifidobacteriaceae contributes at the family level. Finally, total short-chain fatty acids (SCFAs) and butyric acid were significantly higher in the probiotic group at the end of the treatment respect to the beginning. CONCLUSIONS: Overall, this study emphasizes the beneficial anti-inflammageing effect of a prolonged diet based on functional foods enriched with Synbio® through the modulation of the intestinal microbiota and the consequent increase in the SCFA production. SIGNIFICANCE AND IMPACT OF THE STUDY: Synbio® integration in elderly daily diet may be a preventive strategy to support healthy ageing.


Asunto(s)
Proteína C-Reactiva , Probióticos , Humanos , Anciano , Heces/microbiología , Ácidos Grasos Volátiles , Dieta , Ácido Butírico , Método Doble Ciego
2.
J Appl Microbiol ; 133(3): 1956-1968, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35796632

RESUMEN

AIMS: Probiotic supplementation approach offers the possibility to shape the gut microbiota (GM), enabling the development of innovative formulations able to improve intestinal well-being and consequently the related body weight modulation and energy metabolism. In the present clinical study, a new potential probiotic supplement based on Lactiplantibacillus plantarum IMC 510 was studied for weight management. METHODS AND RESULTS: Quantitative characterization by qPCR of representative bacterial groups of GM was used to determine the microbiota modulation at different supplementation periods. Furthermore, measurement of the endpoints linked to weight control (body mass index, body weight, waist circumference) was assessed. Specific questionnaires to evaluate the impact on psychological and physiological point of view were performed. Results showed that after 90 days, Lact. plantarum IMC 510 supplementation brought an improvement in endpoints linked to weight control and healthy status, although no significant changes in the microbiota composition were reported for analysed bacterial groups, except for Lactobacillus spp. and Bifidobacterium spp. CONCLUSIONS: We concluded that Lact. plantarum IMC 510 supplementation could be an interesting tool for weight management. More studies are needed to understand the impact on GM, for example, evaluating the production of short-chain fatty acids, since their important role in dietary metabolism. Further research is necessary to better elucidate the relationship between GM and overweight and the mechanism of action by which Lact. plantarum IMC 510 modifies body weight. SIGNIFICANCE AND IMPACT OF THE STUDY: However, these promising outcomes represent a clear advantage of probiotic supplementation and identify a new potential probiotic as a novel and safe therapeutic approach in the obesity prevention and management.


Asunto(s)
Lactobacillus plantarum , Probióticos , Bacterias , Peso Corporal , Suplementos Dietéticos , Humanos , Lactobacillus plantarum/fisiología , Obesidad , Sobrepeso , Probióticos/farmacología
3.
Genes (Basel) ; 14(1)2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36672830

RESUMEN

(1) Background: Gut microbiota (GM) is the set of microorganisms inhabiting the gastroenteric tract that seems to have a role in the pathogenesis of rheumatic diseases. Recently, many authors proved that GM may influence pharmacodynamics and pharmacokinetics of several drugs with complex interactions that are studied by the growing field of pharmacomicrobiomics. The aim of this review is to highlight current evidence on pharmacomicrobiomics applied to the main treatments of Rheumatoid Arthritis and Spondyloarthritis in order to maximize therapeutic success, in the framework of Personalized Medicine. (2) Methods: We performed a narrative review concerning pharmacomicrobiomics in inflammatory arthritides. We evaluated the influence of gut microbiota on treatment response of conventional Disease Modifying Anti-Rheumatic drugs (cDMARDs) (Methotrexate and Leflunomide) and biological Disease Modifying Anti-Rheumatic drugs (bDMARDs) (Tumor necrosis factor inhibitors, Interleukin-17 inhibitors, Interleukin 12/23 inhibitors, Abatacept, Janus Kinase inhibitors and Rituximab). (3) Results: We found a great amount of studies concerning Methotrexate and Tumor Necrosis Inhibitors (TNFi). Conversely, fewer data were available about Interleukin-17 inhibitors (IL-17i) and Interleukin 12/23 inhibitors (IL-12/23i), while none was identified for Janus Kinase Inhibitors (JAKi), Tocilizumab, Abatacept and Rituximab. We observed that microbiota and drugs are influenced in a mutual and reciprocal way. Indeed, microbiota seems to influence therapeutic response and efficacy, whereas in the other hand, drugs may restore healthy microbiota. (4) Conclusions: Future improvement in pharmacomicrobiomics could help to detect an effective biomarker able to guide treatment choice and optimize management of inflammatory arthritides.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Inhibidores de las Cinasas Janus , Humanos , Metotrexato/farmacología , Metotrexato/uso terapéutico , Abatacept/uso terapéutico , Rituximab/uso terapéutico , Inhibidores de las Cinasas Janus/uso terapéutico , Interleucina-17 , Yin-Yang , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Interleucina-12/uso terapéutico
4.
Int J Mol Sci ; 22(20)2021 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-34681831

RESUMEN

Changes in functionality and composition of gut microbiota (GM) have been associated and may contribute to the development and maintenance of obesity and related diseases. The aim of our study was to investigate for the first time the impact of Lactiplantibacillus (L.) plantarum IMC 510 in a rat model of diet-induced obesity, specifically in the cafeteria (CAF) diet. This diet provides a strong motivation to voluntary overeat, due to the palatability and variety of selected energy-dense foods. The oral administration for 84 days of this probiotic strain, added to the CAF diet, decreased food intake and body weight gain. Accordingly, it ameliorated body mass index, liver and white adipose tissue weight, hepatic lipid accumulation, adipocyte size, serum parameters, including glycemia and low-density lipoprotein levels, in CAF fed rats, potentially through leptin control. In this scenario, L. plantarum IMC 510 showed also beneficial effects on GM, limiting the microbial imbalance established by long exposure to CAF diet and preserving the proportion of different bacterial taxa. Further research is necessary to better elucidate the relationship between GM and overweight and then the mechanism of action by which L. plantarum IMC 510 modifies weight. However, these promising results prompt a clear advantage of probiotic supplementation and identify a new potential probiotic as a novel and safe therapeutic approach in obesity prevention and management.


Asunto(s)
Biodiversidad , Suplementos Dietéticos/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/microbiología , Probióticos/administración & dosificación , Aumento de Peso/efectos de los fármacos , Adipocitos/citología , Tejido Adiposo Blanco/efectos de los fármacos , Alimentación Animal/microbiología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , ADN Bacteriano , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/genética , Leptina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas LDL/efectos de los fármacos , Lipoproteínas LDL/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Obesidad/inducido químicamente , ARN Ribosómico 16S , Ratas , Ratas Sprague-Dawley
5.
Int J Mol Sci ; 19(12)2018 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-30486338

RESUMEN

In recent years, the human microbiota has gained increasing relevance both in research and clinical fields. Increasing studies seem to suggest the centrality of the microbiota and its composition both in the development and maintenance of what we call "health" and in generating and/or favoring (those cases in which the microbiota's complex relational architecture is dysregulated) the onset of pathological conditions. The complex relationships between the microbiota and human beings, which invest core notions of biomedicine such as "health" and "individual," do concern not only problems of an empirical nature but seem to require the need to adopt new concepts and new perspectives in order to be properly analysed and utilized, especially for their therapeutic implementation. In this contribution we report and discuss some of the theoretical proposals and innovations (from the ecological component to the notion of polygenomic organism) aimed at producing this change of perspective. In conclusion, we summarily analyze what impact and what new challenges these new approaches might have on personalized/person centred/precision medicine.


Asunto(s)
Microbioma Gastrointestinal , Medicina de Precisión , Animales , Biodiversidad , Terapia Biológica , Susceptibilidad a Enfermedades , Trasplante de Microbiota Fecal , Heces/microbiología , Genómica/métodos , Humanos , Metagenómica/métodos , Microbiota , Especificidad de Órganos , Medicina de Precisión/métodos , Investigación , Simbiosis
6.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26590477

RESUMEN

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.


Asunto(s)
Heterogeneidad Genética , Terapia Molecular Dirigida , Neoplasias/terapia , Medicina de Precisión , Antineoplásicos Fitogénicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Neoplasias/prevención & control , Transducción de Señal , Microambiente Tumoral/genética
7.
Semin Cancer Biol ; 35 Suppl: S151-S184, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25951989

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Inflamación/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Transformación Celular Neoplásica/efectos de los fármacos , Heterogeneidad Genética/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de los fármacos
8.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25865775

RESUMEN

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.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Microambiente Tumoral/genética , Antineoplásicos/uso terapéutico , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/prevención & control , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos
9.
Semin Cancer Biol ; 35 Suppl: S25-S54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25892662

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Neoplasias/patología , Neoplasias/terapia , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/biosíntesis , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Transducción de Señal/efectos de los fármacos
10.
Semin Cancer Biol ; 35 Suppl: S5-S24, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25869442

RESUMEN

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.


Asunto(s)
Inestabilidad Genómica/efectos de los fármacos , Neoplasias/dietoterapia , Neoplasias/genética , Centrosoma/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Dieta , Inestabilidad Genómica/genética , Humanos , Neoplasias/patología , Pronóstico , Telomerasa/antagonistas & inhibidores , Telomerasa/genética
11.
Semin Cancer Biol ; 35 Suppl: S55-S77, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25749195

RESUMEN

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.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Neoplasias/genética , Neoplasias/terapia , Transducción de Señal , Proteínas de Unión al ADN , Factor 15 de Diferenciación de Crecimiento/genética , Vía de Señalización Hippo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Terapia Molecular Dirigida , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteína de Retinoblastoma/genética , Somatomedinas/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
12.
Semin Cancer Biol ; 35 Suppl: S224-S243, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25600295

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/terapia , Neovascularización Patológica/terapia , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/patología , Proliferación Celular/efectos de los fármacos , Humanos , Inmunoterapia , Neoplasias/prevención & control , Neovascularización Patológica/prevención & control
13.
Expert Opin Ther Pat ; 19(5): 623-41, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19441938

RESUMEN

BACKGROUND: Cancer remains one of the leading causes of death. Over the past decade, discovery of tumor antigens, as well as new findings in basic immunology, have led to novel opportunities for developing active immunotherapeutical approaches for prevention and treatment of cancer. OBJECTIVE/METHODS: This is a review of the literature and patents on the therapeutic potential of immune-based cell cancer therapies. RESULTS/CONCLUSION: In this article, we discuss the different approaches at present used for immune-based cell cancer therapies, and the results obtained both in preclinical models and in clinical trials of hematological malignancies and solid tumors.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/farmacología , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata/inmunología , Neoplasias/inmunología , Neoplasias/prevención & control , Patentes como Asunto
14.
Mol Cell Biol ; 28(16): 5043-60, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18559421

RESUMEN

A common feature of tumor cells is the aberrant expression of ion channels on their plasma membrane. The molecular mechanisms regulating ion channel expression in cancer cells are still poorly known. K(+) channels that belong to the human ether-a-go-go-related gene 1 (herg1) family are frequently misexpressed in cancer cells compared to their healthy counterparts. We describe here a posttranslational mechanism for the regulation of hERG1 channel surface expression in cancer cells. This mechanism is based on the activity of hERG1 isoforms containing the USO exon. These isoforms (i) are frequently overexpressed in human cancers, (ii) are retained in the endoplasmic reticulum, and (iii) form heterotetramers with different proteins of the hERG family. (iv) The USO-containing heterotetramers are retained intracellularly and undergo ubiquitin-dependent degradation. This process results in decreased hERG1 current (I(hERG1)) density. We detailed such a mechanism in heterologous systems and confirmed its functioning in tumor cells that endogenously express hERG1 proteins. The silencing of USO-containing hERG1 isoforms induces a higher I(hERG1) density in tumors, an effect that apparently regulates neurite outgrowth in neuroblastoma cells and apoptosis in leukemia cells.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico , Procesamiento Proteico-Postraduccional , Empalme Alternativo/genética , Apoptosis , Línea Celular Tumoral , Membrana Celular/metabolismo , Clonación Molecular , ADN Complementario/genética , Canales de Potasio Éter-A-Go-Go/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Salud , Humanos , Espacio Intracelular/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Neuritas/metabolismo , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección
15.
Blood ; 106(2): 626-34, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15817684

RESUMEN

Common variable immunodeficiency (CVID) is a primary immune disorder characterized by impaired antibody production, which is in many instances secondary to defective T-cell function (T-CVID). We have previously identified a subset of patients with T-CVID characterized by defective T-cell receptor (TCR)-dependent protein tyrosine phosphorylation. In these patients, ZAP-70 fails to be recruited to the TCR as the result of impaired CD3zeta phosphorylation, which is, however, not dependent on defective Lck expression or activity. Here we show that neither Fyn nor CD45 is affected in these patients. On the other hand, T-CVID T cells show dramatic defects in the Vav/Rac pathway controlling F-actin dynamics. A significant deficiency in Vav protein was indeed observed; in 3 of 4 patients with T-CVID, it was associated with reduced VAV1 mRNA levels. The impairment in Vav expression correlated with defective F-actin reorganization in response to TCR/CD28 co-engagement. Furthermore, TCR/CD28-dependent up-regulation of lipid rafts at the cell surface, which requires F-actin dynamics, was impaired in these patients. The actin cytoskeleton defect could be reversed by reconstitution of Vav1 expression in the patients' T cells. Results demonstrate an essential role of Vav in human T cells and strongly suggest Vav insufficiency in T-CVID.


Asunto(s)
Actinas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Complejo CD3/metabolismo , Señalización del Calcio , Inmunodeficiencia Variable Común/inmunología , ADN Complementario/genética , Gangliósido G(M1)/metabolismo , Expresión Génica , Humanos , Técnicas In Vitro , Antígenos Comunes de Leucocito/genética , Microdominios de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Proteínas Proto-Oncogénicas c-vav , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína Tirosina Quinasa ZAP-70 , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA