Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1155468, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37266436

RESUMEN

Recent discoveries shed light on molecular mechanisms responsible for classical Hodgkin lymphoma (HL) development and progression, along with features of Hodgkin - Reed and Sternberg cells (HRS). Here, we summarize current knowledge on characteristic molecular alterations in HL, as well as existing targeted therapies and potential novel treatments for this disease. We discuss the importance of cluster of differentiation molecule 30 (CD30) and the programmed cell death-1 protein (PD-1) and ligands (PD-L1/2), and other molecules involved in immune modulation in HL. We highlight emerging evidence indicating that the altered function of SWI/SNF-type chromatin remodeling complexes, PRC2, and other epigenetic modifiers, contribute to variations in chromatin status, which are typical for HL. We postulate that despite of the existence of plentiful molecular data, the understanding of HL development remains incomplete. We therefore propose research directions involving analysis of reverse signaling in the PD-1/PD-L1 mechanism, chromatin remodeling, and epigenetics-related alterations, in order to identify HL features at the molecular level. Such attempts may lead to the identification of new molecular targets, and thus will likely substantially contribute to the future development of more effective targeted therapies.


Asunto(s)
Enfermedad de Hodgkin , Células de Reed-Sternberg , Humanos , Células de Reed-Sternberg/metabolismo , Células de Reed-Sternberg/patología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Enfermedad de Hodgkin/genética , Transducción de Señal
2.
Cancers (Basel) ; 13(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34439305

RESUMEN

Growing tumors avoid recognition and destruction by the immune system. During continuous stimulation of tumor-infiltrating lymphocytes (TILs) by tumors, TILs become functionally exhausted; thus, they become unable to kill tumor cells and to produce certain cytokines and lose their ability to proliferate. This collectively results in the immune escape of cancer cells. Here, we show that breast cancer cells expressing PD-L1 can accelerate exhaustion of persistently activated human effector CD4+ T cells, manifesting in high PD-1 and PD-L1 expression level son T cell surfaces, decreased glucose metabolism genes, strong downregulation of SWI/SNF chromatin remodeling complex subunits, and p21 cell cycle inhibitor upregulation. This results in inhibition of T cell proliferation and reduction of T cell numbers. The RNAseq analysis on exhausted CD4+ T cells indicated strong overexpression of IDO1 and genes encoding pro-inflammatory cytokines and chemokines. Some interleukins were also detected in media from CD4+ T cells co-cultured with cancer cells. The PD-L1 overexpression was also observed in CD4+ T cells after co-cultivation with other cell lines overexpressing PD-L1, which suggested the existence of a general mechanism of CD4+ T cell exhaustion induced by cancer cells. The ChIP analysis on the PD-L1 promoter region indicated that the BRM recruitment in control CD4+ T cells was replaced by BRG1 and EZH2 in CD4+ T cells strongly exhausted by cancer cells. These findings suggest that epi-drugs such as EZH2 inhibitors may be used as immunomodulators in cancer treatment.

3.
Am J Cancer Res ; 11(12): 5965-5978, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35018236

RESUMEN

About 40% of clear cell renal cell carcinoma (ccRCC) cases carry the pbrm1 mutation inactivating BAF180 subunit of the SWI/SNF chromatin remodeling complex (CRC). Here we show that the majority of transcriptomic changes appear at the stage I of ccRCC development. By contrast, the stage II ccRCC exhibits hyperactivation of DNA replication demonstrated by the overexpression of several genes, e.g., RRM1 and RRM2 genes encoding subunits of ribonucleotide reductase (RNR) complex. We found that the degree of RRM1 and RRM2 upregulation in ccRCC patients depends on pbrm1 mutation. We show that the BAF180 protein product of the PBRM1 gene directly binds to RRM1 and RRM2 loci. The BAF180 binding regions are targeted by regulatory proteins previously reported as SWI/SNF CRC interacting partners. BAF180 binding to RRMs loci correlates with enrichment of H3K27me3 in case of RRM1 and H3K14Ac on RRM2, indicating the existence of differential regulatory mechanism controlling expression of these genes. We found that the strong overexpression of RRM2 in ccRCC patient samples correlates with T cell infiltration. Surprisingly, the majority of tumor infiltrating lymphocytes (TILs) consisted of CD4+ T cells. Furthermore, we show that exhausted CD4+ T cells induced the expression of the RRM2 gene in the primary ccRCC cell line. Collectively, our results provide the link between PBRM1 loss, RRM2 expression and T cell infiltration, which may lead to the establishment of new treatment of this disease.

4.
J Steroid Biochem Mol Biol ; 189: 48-62, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30772447

RESUMEN

Conventional cytotoxic drugs preferentially eliminate differentiated cancer cells but spare relatively more resistant stem-like cancer cells capable to initiate recurrence. Due to cancer cell plasticity, the stem-like phenotype can be also acquired by cancer cells refractory to treatment with cytotoxic drugs. We investigated whether drugs inhibiting receptor tyrosine kinases could be used to target human colon cancer cells initiating cancer regrowth following conventional cytotoxic treatment. The moderately differentiated cell line HT-29 and poorly differentiated cell line HCT-116 were exposed to 5-fluorouracil (5-FU). Cells that resisted the exposure to 5-FU were subsequently treated with imatinib or sunitinib. Both drugs reduced clonogenicity of 5-FU-refractory cells under normoxic and hypoxic culture conditions. The expression of numerous stemness-related genes was upregulated in cancer cells following the exposure to 5-FU, and remained at a high level in 5-FU-refractory cells undergoing renewal under normoxia, but decreased spontaneously under hypoxia. Imatinib downregulated the expression of stemness-related genes in cells undergoing renewal under normoxia. A combination of imatinib with PRI-2191, an analogue of 1,25-dihydroxyvitamin D3, downregulated stemness-related genes in HCT-116/5-FU cells more efficiently than imatinib alone. A synthetic analogue of 1,25-dihydroxyvitamin D2 (PRI-1906) abolished the effect of imatinib on gene expression in HCT-116/5-FU cells undergoing renewal under normoxia. Sunitinib promoted shift of phenotype of HT-29/5-FU cells undergoing renewal toward stem-like one. It suggests that the phenotype shift toward stemness induced by sequential sunitinib treatment following 5-FU treatment could increase a risk of cancer recurrence. In contrast to sunitinib, imatinib could be used both to interfere with cancer regrowth after conventional chemotherapy and to downregulate the expression of stemness-related genes in residual colon cancer cells capable to initiate cancer recurrence. The findings suggest that imatinib could also be combined with vitamin D analogue PRI-2191 to prevent recurrence more efficiently than imatinib alone and to compensate for vitamin D deficiency resulting from imatinib treatment.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Dihidroxicolecalciferoles/farmacología , Fluorouracilo/farmacología , Mesilato de Imatinib/farmacología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos
5.
Arch Immunol Ther Exp (Warsz) ; 65(1): 51-67, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26956644

RESUMEN

Conventional anti-cancer drugs preferentially eliminate differentiated cancer cells but those cells that are spared (i.e. cancer stem cells: CSC), initiate recurrence. We tested whether drugs that target receptor tyrosine kinases (RTKs) involved in developmental signaling cascades and activated in CSC, could be used to silence and/or to eliminate colorectal cancer cells refractory to conventional treatment with cytoreductive drugs. A sequential treatment model was thereby developed with doxorubicin (DOX) and imatinib. CT-26 mouse colon carcinoma cells were pre-treated with DOX to select DOX-refractory cells with CSC properties, which were then subsequently treated with RTK inhibitor imatinib, where their regrowth was found to be inhibited. Under both normoxic and hypoxic conditions, imatinib potently inhibited clonogenicity of DOX-refractory CT-26 cells. Treatment with DOX did not eliminate tumorigenic CT-26 cells, since CT-26 cells pre-exposed to DOX in vitro, when inoculated subcutaneously, induced tumors in 90 % of mice, as opposed to a 100 % rate in the case of chemonaive CT-26 cells. In mice inoculated with chemonaive CT-26 cells, tumor formation was not prevented by imatinib. However, imatinib prevented tumor formation in 50 % of mice inoculated with CT-26 cells pre-exposed to DOX in vitro, with the remaining 50 % mice showing delayed tumor formation. These results suggest that the sequential use of the drug imatinib, as a drug targeting cancer cells expressing stem cell features after conventional cytoreductive treatment, is a promising future strategy for preventing tumor recurrence.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Mesilato de Imatinib/uso terapéutico , Antígeno AC133/química , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Citometría de Flujo , Hipoxia , Ratones , Ratones Endogámicos BALB C , Recurrencia Local de Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Oxígeno/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
6.
Clin Exp Med ; 17(2): 193-207, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-26961690

RESUMEN

We have previously found that ex vivo expanded human CD4+CD25+Treg cells suppress proliferation of lymphoma B-cell lines. Here we demonstrate that the immunomodulatory drug lenalidomide potentiates suppression of lymphoma B-cell proliferation by freshly isolated CD4+CD25+Tregs, as well as suppression by Tregs expanded polyclonally in the presence of rapamycin from CD4+CD25+T cells or CD4+CD25+CD127loT cells. The regulation of lymphoma cell proliferation by Tregs pre-expanded with "third-party" allogeneic MoDCs in the presence of rapamycin was also potentiated by lenalidomide. Lenalidomide contributed to the suppression exerted by Tregs despite concomitant downregulation of Treg proliferation. Lenalidomide did not reduce the suppression of conventional T cells by expanded Tregs. The exposure of polyclonally expanded Tregs to lenalidomide did not significantly alter their phenotype. There was no uniform pattern of lenalidomide effect on Treg-mediated regulation of lymphoma B cells freshly isolated from patients. Freshly isolated lymphoma cells activated with multimeric CD40L and IL-4 to support their survival in vitro varied in their sensitivity to lenalidomide, and the regulatory effect of Tregs on such lymphoma cells ranged from suppression to help in individual patients. Lenalidomide potentiated or attenuated Treg effects on the survival of freshly isolated lymphoma cells. A combination of lenalidomide treatment with adoptive transfer of CD4+CD25+Tregs or CD4+CD25+CD127loTregs expanded ex vivo could be used to suppress proliferation of residual lymphoma in select patients with lymphoma responsive to the regulation by Tregs and sensitive to lenalidomide.


Asunto(s)
Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Proliferación Celular , Factores Inmunológicos/metabolismo , Linfoma/inmunología , Linfocitos T Reguladores/inmunología , Talidomida/análogos & derivados , Adulto , Anciano , Linfocitos B/fisiología , Donantes de Sangre , Antígenos CD4/análisis , Células Cultivadas , Femenino , Humanos , Subunidad alfa del Receptor de Interleucina-2/análisis , Lenalidomida , Linfoma/patología , Masculino , Persona de Mediana Edad , Sirolimus/metabolismo , Linfocitos T Reguladores/química , Talidomida/metabolismo
7.
Int J Mol Sci ; 17(6)2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27314328

RESUMEN

This study aimed to evaluate the capacity of hypocalcemic analogues of 1α,25-dihydroxyvitamin D2 (1,25D2) and 1α,25-dihydroxyvitamin D3 (1,25D3) to inhibit regrowth and regulate the stemness-related gene expression in colon cancer cells undergoing renewal after exposure to 5-fluorouracil (5-FU). All of the tested analogues of 1,25D2 equally potently decreased the clonogenicity and the proliferative activity of HT-29 cells which survived the exposure to 5-FU, but differently regulated gene expression of these cells during their renewal. 1,25D2 and analogues (PRI-1907 and PRI-1917), as well as 1,25D3 and analogue PRI-2191, decreased the relative expression level of several stemness-related genes, such as NANOG, OCT3/4, PROM1, SOX2, ALDHA1, CXCR4, in HT-29/5-FU cells during their renewal, in comparison to untreated HT-29/5-FU cells. The other 1,25D2 analogues (PRI-1906 and PRI-1916) were not capable of downregulating the expression of these stemness-related genes as the analogues PRI-1907 and PRI-1917 did. All of the tested vitamin D analogues upregulated CDH1, the gene encoding E-cadherin associated with epithelial phenotype. Out of the series of analogues studied, side-chain branched analogues of 1,25D2 (PRI-1907, PRI-1917) and the analogue of 1,25D3 (PRI-2191) might be used to target cancer cells with stem-like phenotypes that survive conventional chemotherapy.


Asunto(s)
Calcitriol/farmacología , Vitaminas/farmacología , Antineoplásicos/toxicidad , Calcitriol/análogos & derivados , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos , Fluorouracilo/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos
8.
PLoS One ; 11(5): e0155893, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27196001

RESUMEN

Resistance to glucocorticosteroids (GCs) is a major adverse prognostic factor in B-ALL, but the molecular mechanisms leading to GC resistance are not completely understood. Herein, we sought to elucidate the molecular background of GC resistance in B-ALL and characterize the therapeutic potential of targeted intervention in these mechanisms. Using exploratory bioinformatic approaches, we found that resistant cells exhibited significantly higher expression of MEK/ERK (MAPK) pathway components. We found that GC-resistant ALL cell lines had markedly higher baseline activity of MEK and small-molecule MEK1/2 inhibitor selumetinib increased GCs-induced cell death. MEK inhibitor similarly increased in vitro dexamethasone activity in primary ALL blasts from 19 of 22 tested patients. To further confirm these observations, we overexpressed a constitutively active MEK mutant in GC-sensitive cells and found that forced MEK activity induced resistance to dexamethasone. Since recent studies highlight the role GC-induced autophagy upstream of apoptotic cell death, we assessed LC3 processing, MDC staining and GFP-LC3 relocalization in cells incubated with either DEX, SEL or combination of drugs. Unlike either drug alone, only their combination markedly increased these markers of autophagy. These changes were associated with decreased mTOR activity and blocked 4E-BP1 phosphorylation. In cells with silenced beclin-1 (BCN1), required for autophagosome formation, the synergy of DEX and SEL was markedly reduced. Taken together, we show that MEK inhibitor selumetinib enhances dexamethasone toxicity in GC-resistant B-ALL cells. The underlying mechanism of this interaction involves inhibition of mTOR signaling pathway and modulation of autophagy markers, likely reflecting induction of this process and required for cell death. Thus, our data demonstrate that modulation of MEK/ERK pathway is an attractive therapeutic strategy overcoming GC resistance in B-ALL patients.


Asunto(s)
Autofagia , Dexametasona/farmacología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Serina-Treonina Quinasas TOR/metabolismo , Apoptosis , Bencimidazoles/farmacología , Muerte Celular , Línea Celular Tumoral , Biología Computacional , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Microscopía Fluorescente , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , ARN Interferente Pequeño/metabolismo
9.
Leuk Lymphoma ; 57(8): 1903-20, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26758248

RESUMEN

Activated regulatory T cells (Tregs) suppress proliferation and differentiation of normal B cells. In our study, allogeneic polyclonal CD4 (+) CD25 (+) Tregs and CD4 (+) CD25 (+) CD127(lo)Tregs expanded in vitro in the presence of rapamycin and low dose IL-2 suppressed proliferation of 11 out of 12 established lymphoma B-cell lines. The effect of expanded CD4 (+) CD25 (+) Tregs on survival of freshly isolated lymphoma B cells maintained in culture with soluble multimeric CD40L and IL-4 was variable across lymphoma entities. The survival of freshly isolated follicular lymphoma cells usually decreased in cocultures with CD4 (+) CD25 (+) Tregs. Treg effect on chronic lymphocytic leukemia/small lymphocytic lymphoma cells ranged from suppression to help in individual patients. CD4 (+) CD25 (+) Tregs or CD4 (+) CD25 (+) CD127(lo)Tregs expanded ex vivo with rapamycin could be used to suppress regrowth of residual lymphoma after autologous hematopoietic cell transplantation (HCT), and to counteract both graft-versus-host disease and lymphoma re-growth after allogeneic HCT in select patients with lymphoma susceptible to the regulation by Tregs.


Asunto(s)
Linfocitos B/patología , Proliferación Celular , Leucemia Linfocítica Crónica de Células B/inmunología , Linfoma de Células B/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/efectos de los fármacos , Biopsia con Aguja Fina , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Cocultivo , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead , Humanos , Interleucina-2/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ganglios Linfáticos/patología , Activación de Linfocitos , Linfoma de Células B/patología , Masculino , Persona de Mediana Edad , Cultivo Primario de Células , Sirolimus/farmacología
10.
Blood ; 127(6): 739-48, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26585955

RESUMEN

Inhibition of spleen tyrosine kinase (SYK) in tonic B-cell receptor (BCR) signal-dependent diffuse large B-cell lymphomas (DLBCLs) inhibits cellular proliferation, decreases cholesterol biosynthesis, and triggers apoptosis, at least in part via a mechanism involving decreased activity of phosphatidylinositol 3-kinase/AKT axis. Because forkhead box O1 (FOXO1) is a major effector of this pathway, we investigated the role of FOXO1 in toxicity of BCR pathway inhibition. Inhibition of SYK in DLBCL cells with tonic BCR signaling decreased phospho-AKT and phospho-FOXO1 levels and triggered FOXO1-driven gene expression. Introduction of constitutively active FOXO1 mutant triggered cell cycle arrest and apoptosis, indicating that increased FOXO1 activity is toxic to these DLBCL cells. Depletion of FOXO1 with short hairpin RNA led to almost complete resistance to chemical SYK inhibitor R406, demonstrating that FOXO1 is also required for R406-induced cell death. FOXO1 in these cells is also involved in regulation of expression of the critical master regulator of cholesterol biosynthesis, SREBP1. Because HRK is the key effector of SYK inhibition, we characterized a mechanism linking FOXO1 activation and HRK induction that involves caspase-dependent cleavage of HRK's transcriptional repressor DREAM. Because AKT in lymphoma cells can be regulated by other signals than BCR, we assessed the combined effects of the AKT inhibitor MK-2206 with R406 and found markedly synergistic FOXO1-dependent toxicity. In primary DLBCLs, FOXO1 expression was present in 80% of tumors, correlated with SYK activity, and was associated with longer overall survival. These results demonstrate that FOXO1 is required for SYK and AKT inhibitor-induced toxicity.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos B/genética , Apoptosis/genética , Ciclo Celular/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/patología , Análisis por Micromatrices , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/genética , Quinasa Syk , Activación Transcripcional , Células Tumorales Cultivadas
11.
Tumour Biol ; 37(4): 4699-709, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26511971

RESUMEN

This study was aimed to determine whether hypocalcemic analogs of active forms of vitamins D modulate expression of genes related to stem-like phenotype in colon cancer cell lines HT-29 and HCT-116 undergoing renewal after the treatment with 5-fluorouracil (5-FU). Both lines express vitamin D receptor, but differ in differentiation stage and vitamin D sensitivity. Cells that resisted the 5-FU exposure were treated with synthetic analog of 1,25-dihydroxyvitamin D2 (PRI-1906) and analogs of 1,25-dihydroxyvitamin D3 (PRI-2191 and PRI-2205). Proliferative activity was more profoundly affected by vitamin D analogs in HT-29/5-FU than in HCT-116/5-FU cells. In HT-29/5-FU cells, analogs PRI-1906 and PRI-2191 downregulated the expression of genes related to survival, re-growth, and invasiveness during renewal, while PRI-2205 increased expression of genes related to differentiation only. In HCT-116/5-FU cells, PRI-2191 decreased the expression of stemness- and angiogenesis-related genes, whereas PRI-1906 augmented their expression. The effects in HCT-116/5-FU cells were observed at higher concentrations of the analogs than those used for HT-29/5-FU cells. Out of the series of analogs studied, PRI-2191 might be used to counteract the renewal of both moderately and poorly differentiated cancer cells following conventional treatment.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Calcitriol/análogos & derivados , Dihidroxicolecalciferoles/farmacología , Resistencia a Antineoplásicos , Ergocalciferoles/farmacología , Fluorouracilo/farmacología , Calcitriol/farmacología , Autorrenovación de las Células/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal , Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/fisiología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
12.
PLoS One ; 9(6): e98073, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24915193

RESUMEN

The primary function of hair and fur covering mammalian skin is to provide mechanical and thermal protection for the body. The proteins that constitute hair are extremely resistant to degradation by environmental factors. However, even durable materials can be slowly broken down by mechanical stresses, biodegradation mediated by endogenous enzymes in the skin or host microbes. We hypothesised that the biodegradation products of hair may possess bioprotective properties, which supplement their physical protective properties. Although evolutionary processes have led to a reduction in the amount of hair on the human body, it is possible that the bioprotective properties of hair biodegradation products have persisted. The human skin is exposed to various environmental carcinogenic factors. Therefore, we hypothesised that the potential bioprotective mechanisms of hair degradation products affect melanoma growth. We used pepsin to partially digest hair enzymatically, and this process produced a water-soluble lysate containing a mixture of peptides, including fragments of keratin and keratin-associated proteins. We found out that the mixtures of soluble peptides obtained from human hair inhibited the proliferation of human melanoma cells in vitro. Moreover, the hair-derived peptide mixtures also inhibited the proliferation of B lymphoma cells and urinary bladder cancer cells. Normal human cells varied in their susceptibility to the effects of the lysate; the hair-derived peptide mixtures modulated the proliferation of normal human fibroblasts but did not inhibit the proliferation of human mesenchymal cells derived from umbilical cord stromal cells. These results suggest that hair-derived peptides may represent a new class of anti-proliferative factors derived from basically structural proteins. Identification of active regulatory compounds and recognition of the mechanism of their action might pave the way to elaboration of new anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Queratinas Específicas del Pelo/química , Fragmentos de Péptidos/farmacología , Hidrolisados de Proteína/farmacología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Queratinocitos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos
13.
Folia Neuropathol ; 51(2): 132-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23821385

RESUMEN

Opiate analgesics like morphine or fentanyl are the most widely used medicines for relieving severe acute or chronic pain, including cancer pain. Unfortunately, chronic pain treatment is associated with fast development of tolerance that creates the need to escalate the treatment doses. In addition, opiates may stimulate progression of cancer. Therefore, a new type of effective analgesic especially designed for chronic cancer pain treatment is needed. In this paper, a new opioid peptide analogue has been described as a new analgesic. The compound is characterized by very high agonist affinities to MOR and also high, but ten times lower affinity to DOR. Affinity to hNK1 as an antagonist is on the level of C-terminal hexapeptide fragment analogue of Substance P. The compound expressed reasonable antiproliferative properties toward various cancer cells. Interestingly, the peptide did not interfere with the proliferation of fibro-blasts. Therefore, the compound should be considered as a new analgesic for treatment of cancer-related pains with adjuvant anticancer properties which may support cancer treatments.


Asunto(s)
Analgésicos Opioides/farmacología , Antineoplásicos/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides/agonistas , Taquicininas/antagonistas & inhibidores , Adyuvantes Farmacéuticos/síntesis química , Adyuvantes Farmacéuticos/metabolismo , Adyuvantes Farmacéuticos/farmacología , Analgésicos Opioides/síntesis química , Analgésicos Opioides/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Adyuvante/métodos , Cricetinae , Cricetulus , Humanos , Antagonistas de Narcóticos/síntesis química , Antagonistas de Narcóticos/metabolismo , Unión Proteica/fisiología , Ratas Wistar , Receptores de Neuroquinina-1/fisiología , Receptores Opioides/metabolismo , Taquicininas/fisiología
14.
Med Oncol ; 29(4): 2966-77, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22302285

RESUMEN

Dendritic cells may be successfully used to induce in vivo-specific anti-tumor responses when combined with the appropriate antigen in the appropriate context. The purpose of this study was to evaluate efficacy of peptide-loaded DC vaccine in high-risk stage III melanoma patients after lymph node dissection (LND). HLA-A2+, -A1+, or -A3+ melanoma patients (N=22), stage III, N1b-N3, received 5­16 (median: 11) DC vaccines loaded with MHC class-I-restricted melanoma peptides respective to the patient's haplotype, and with autologous tumor lysate, if available. Vaccinated patients were matched to unvaccinated stage III controls (22 of 869) by sex, number of metastatic lymph nodes, extracapsular involvement, LND type, Breslow stage, and ulceration. Vaccination elicited cutaneous delayed-type hypersensitivity (DTH) or/and IFN-γ-producing CD8+ cell response to melanoma peptides in 15 of 22 patients. Three-year overall survival (OS) rate was 68.2% in the vaccinated group versus 25.7% in the control group, P value accounting for matching: 0.0290. In a Cox regression model, hazard ratio (HR) for death of vaccinated patients was 0.31 [95% confidence interval (CI): 0.10­0.94]. The corresponding values for 3-year disease-free survival rate were 40.9 versus 14.5%, P=0.1083; HR of recurrence for vaccinated, 0.46 (95% CI: 0.18­1.22). There was no grade>1 toxicity. The DC/peptide vaccine was well tolerated and elicited immune responses to melanoma antigens. Vaccinated patients had significantly longer OS after LND than the matched controls, but a significant improvement in the primary endpoint DFS was not achieved.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Melanoma/terapia , Vacunación , Adulto , Anciano , Vacunas contra el Cáncer/efectos adversos , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Antígenos HLA/genética , Haplotipos , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Vacunación/efectos adversos
15.
J Proteomics ; 74(12): 2596-616, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21310271

RESUMEN

The success of genome projects has provided us with a vast amount of information on genes of many pathogenic species and has raised hopes for rapid progress in combating infectious diseases, both by construction of new effective vaccines and by creating a new generation of therapeutic drugs. Proteomics, a strategy complementary to the genomic-based approach, when combined with immunomics (looking for immunogenic proteins) and vaccinomics (characterization of host response to immunization), delivers valuable information on pathogen-host cell interaction. It also speeds the identification and detailed characterization of new antigens, which are potential candidates for vaccine development. This review begins with an overview of the global status of vaccinology based on WHO data. The main part of this review describes the impact of proteomic strategies on advancements in constructing effective antibacterial, antiviral and anticancer vaccines. Diverse aspects of disease mechanisms and disease preventions have been investigated by proteomics.


Asunto(s)
Proteómica/métodos , Vacunas/inmunología , Vacunas/farmacología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Humanos , Micosis/inmunología , Micosis/prevención & control , Neoplasias/inmunología , Neoplasias/prevención & control , Vacunación/métodos , Virosis/inmunología , Virosis/prevención & control
16.
Acta Pol Pharm ; 65(6): 625-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19172843

RESUMEN

This review presents the latest achievements in basic studies on stem-cell biology and on approaches aimed to design therapies based on stem cells and dendritic cells. Studies on stem-cell homeostasis are aimed to delineate the accessibility of these cells for therapeutic purposes. Hematopoietic stem cell transplantation has become a routine application of stem cells in the treatment of patients with cancer and with hematologic disorders. Perspectives of application of the mesenchymal stem cells for regenerative medicine and for tolerance induction following allogeneic transplantation are being extensively explored. Reprogramming of adult somatic cells to an undifferentiated pluripotent state in vitro by a transduction with just four genes encoding transcription factors opened the way for the generation of patient-specific pluripotent stem cells. Such induced pluripotent stem cells hold a great promise for replacement therapies of various so far incurable disorders. However, because of the tumorigenic potential of the retroviral vector transduction-induced pluripotent stem cells, before these cells become useful for clinical application, appropriate, safe methods of stem cell generation, selection, proliferation and differentiation need to be elaborated. There is a great potential for further developments of cancer immunotherapies and for controlling of post-transplantational reactions, if current basic studies on stem cells and on immunostimulatory and tolerogenic dendritic cells would be successfully translated to the clinic.


Asunto(s)
Células Dendríticas/trasplante , Trasplante de Células Madre , Animales , Enfermedades Hematológicas/terapia , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos , Células Madre Mesenquimatosas/metabolismo , Neoplasias/terapia , Células Madre Pluripotentes/metabolismo
17.
Eur J Cancer ; 42(16): 2671-4, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16978860

RESUMEN

Marker genes, commonly used to detect circulating tumour cells in RT-PCR-based tests: squamous-cell carcinoma antigen, epidermal growth factor receptor, mammaglobin, small breast epithelial mucin, but not carbonic anhydrase 9, were shown to be expressed in normal, mitogen-stimulated peripheral blood mononuclear cells (PBMNC). Thus, considering the inflammatory reactions often accompanying cancer development, to reduce false-positive results of the metastatic tumour cell tests, molecular markers should be validated not against normal peripheral blood, but against activated lymphoid cells, such as in vitro mitogen-stimulated PBMNC.


Asunto(s)
Biomarcadores de Tumor/análisis , Leucocitos Mononucleares/patología , Activación de Linfocitos/fisiología , Células Neoplásicas Circulantes/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas , Línea Celular Tumoral , Humanos
18.
Acta Biochim Pol ; 53(1): 203-12, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16404477

RESUMEN

Dendritic cells (DC) generated from human umbilical cord blood might replace patients' DC in attempts to elicit tumor-specific immune response in cancer patients. We studied the efficiency of transfection of human cord blood DC with plasmid DNA carrying the enhanced version of green fluorescent protein (EGFP) as a reporter gene, to test if nonviral gene transfer would be a method to load DC with protein antigens for immunotherapy purposes. Cord blood mononuclear cells were cultured in serum-free medium in the presence of granulocyte-monocyte colony stimulating factor (GM-CSF), stem cell factor (SCF) and Flt-3 ligand (FL), to generate DC from their precursors, and thereafter transfected by electroporation. Maturation of DC was induced by stimulation with GM-CSF, SCF, FL and phorbol myristate acetate (PMA). Transfected DC strongly expressed EGFP, but transfection efficiency of DC, defined as HLA-DR(+) cells lacking lineage-specific markers, did not exceed 2.5%. Expression of the reporter gene was also demonstrated in the DC generated from transfected, purified CD34(+) cord blood cells, by stimulation with GM-CSF, SCF, FL, and tumor necrosis factor alpha (TNF-alpha). Transfection of CD34(+) cells was very efficient, but proliferation of the transfected cells was much reduced as compared to the untransfected cells. Therefore, the yield of transgene-expressing DC was relatively low. In conclusion, nonviral transfection of cord blood DC proved feasible, but considering the requirements for immunotherapy in cancer patients, transfection of differentiated DC or generation of DC from transfected hematopoietic stem cells provide only a limited number of DC expressing the transgene.


Asunto(s)
Células Dendríticas/citología , Sangre Fetal/citología , Inmunoterapia/métodos , Neoplasias/terapia , Transgenes , Venas Umbilicales/citología , Antígenos CD34/biosíntesis , Medio de Cultivo Libre de Suero/metabolismo , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Leucocitos Mononucleares/citología , Proteínas de la Membrana/metabolismo , Factor de Células Madre/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...