Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Eur J Pharm Sci ; 173: 106167, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35304859

RESUMO

Bladder diseases affect millions of patients worldwide and compromise their quality of life with a substantial economic impact. The not fully understood aetiologies of bladder diseases limit the current diagnosis and therapeutic options to primarily symptomatic treatment. In addition, bladder targeted drug delivery is challenging due to its unique anatomical features and its natural physiological function of urine storage and frequent voiding. Therefore, current treatment options often fail to provide a highly effective, precisely targeted and long-lasting treatment. With the growing maturity of gene therapy, comprehensive studies are needed to provide a better understanding of the molecular mechanisms underpinning bladder diseases and help to identify novel gene therapeutic targets and biomarkers for treating bladder diseases. In this review, molecular mechanisms involved in pathology of bladder cancer, interstitial cystitis and overactive bladder syndrome are reviewed, with focus on establishing potential novel treatment options. Proposed novel therapies, including gene therapy combined with nanotechnology, localised drug delivery by nanoparticles, and probiotics, are discussed in regard to their safety profiles, efficacy, treatment lenght, precise targeting, and in comparison to conventional treatment methods.


Assuntos
Cistite Intersticial , Nanopartículas , Biomarcadores , Feminino , Humanos , Masculino , Qualidade de Vida , Bexiga Urinária
2.
Cancer Res ; 79(9): 2285-2297, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-30622116

RESUMO

Metabolic adjustments are necessary for the initiation, proliferation, and spread of cancer cells. Although mitochondria have been shown to move to cancer cells from their microenvironment, the metabolic consequences of this phenomenon have yet to be fully elucidated. Here, we report that multiple myeloma cells use mitochondrial-based metabolism as well as glycolysis when located within the bone marrow microenvironment. The reliance of multiple myeloma cells on oxidative phosphorylation was caused by intercellular mitochondrial transfer to multiple myeloma cells from neighboring nonmalignant bone marrow stromal cells. This mitochondrial transfer occurred through tumor-derived tunneling nanotubes (TNT). Moreover, shRNA-mediated knockdown of CD38 inhibits mitochondrial transfer and TNT formation in vitro and blocks mitochondrial transfer and improves animal survival in vivo. This study describes a potential treatment strategy to inhibit mitochondrial transfer for clinical benefit and scientifically expands the understanding of the functional effects of mitochondrial transfer on tumor metabolism. SIGNIFICANCE: Multiple myeloma relies on both oxidative phosphorylation and glycolysis following acquisition of mitochondria from its bone marrow microenvironment.See related commentary by Boise and Shanmugam, p. 2102.


Assuntos
Mieloma Múltiplo , Animais , Metabolismo Energético , Glicólise , Mitocôndrias , Fosforilação Oxidativa , Microambiente Tumoral
4.
Blood ; 130(14): 1649-1660, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28733324

RESUMO

Improvements in the understanding of the metabolic cross-talk between cancer and its microenvironment are expected to lead to novel therapeutic approaches. Acute myeloid leukemia (AML) cells have increased mitochondria compared with nonmalignant CD34+ hematopoietic progenitor cells. Furthermore, contrary to the Warburg hypothesis, AML relies on oxidative phosphorylation to generate adenosine triphosphate. Here we report that in human AML, NOX2 generates superoxide, which stimulates bone marrow stromal cells (BMSC) to AML blast transfer of mitochondria through AML-derived tunneling nanotubes. Moreover, inhibition of NOX2 was able to prevent mitochondrial transfer, increase AML apoptosis, and improve NSG AML mouse survival. Although mitochondrial transfer from BMSC to nonmalignant CD34+ cells occurs in response to oxidative stress, NOX2 inhibition had no detectable effect on nonmalignant CD34+ cell survival. Taken together, we identify tumor-specific dependence on NOX2-driven mitochondrial transfer as a novel therapeutic strategy in AML.


Assuntos
Leucemia Mieloide Aguda/patologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/patologia , Mitocôndrias/patologia , NADPH Oxidases/metabolismo , Superóxidos/metabolismo , Animais , Antígenos CD34/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Mitocôndrias/metabolismo , NADPH Oxidase 2 , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
5.
Blood ; 129(10): 1320-1332, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28049638

RESUMO

Despite currently available therapies, most patients diagnosed with acute myeloid leukemia (AML) die of their disease. Tumor-host interactions are critical for the survival and proliferation of cancer cells; accordingly, we hypothesize that specific targeting of the tumor microenvironment may constitute an alternative or additional strategy to conventional tumor-directed chemotherapy. Because adipocytes have been shown to promote breast and prostate cancer proliferation, and because the bone marrow adipose tissue accounts for up to 70% of bone marrow volume in adult humans, we examined the adipocyte-leukemia cell interactions to determine if they are essential for the growth and survival of AML. Using in vivo and in vitro models of AML, we show that bone marrow adipocytes from the tumor microenvironment support the survival and proliferation of malignant cells from patients with AML. We show that AML blasts alter metabolic processes in adipocytes to induce phosphorylation of hormone-sensitive lipase and consequently activate lipolysis, which then enables the transfer of fatty acids from adipocytes to AML blasts. In addition, we report that fatty acid binding protein-4 (FABP4) messenger RNA is upregulated in adipocytes and AML when in coculture. FABP4 inhibition using FABP4 short hairpin RNA knockdown or a small molecule inhibitor prevents AML proliferation on adipocytes. Moreover, knockdown of FABP4 increases survival in Hoxa9/Meis1-driven AML model. Finally, knockdown of carnitine palmitoyltransferase IA in an AML patient-derived xenograft model improves survival. Here, we report the first description of AML programming bone marrow adipocytes to generate a protumoral microenvironment.


Assuntos
Adipócitos/patologia , Células da Medula Óssea/patologia , Leucemia Mieloide Aguda/patologia , Microambiente Tumoral/fisiologia , Adipócitos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Técnicas de Cocultura , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Citometria de Fluxo , Xenoenxertos , Humanos , Imuno-Histoquímica , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Oncotarget ; 7(26): 39784-39795, 2016 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-27174919

RESUMO

Phosphoinositide-3-kinase (PI3K) is an enzyme group, known to regulate key survival pathways in acute myeloid leukaemia (AML). It generates phosphatidylinositol-3,4,5-triphosphate, which provides a membrane docking site for protein kinaseB activation. PI3K catalytic p110 subunits are divided into 4 isoforms; α,ß,δ and γ. The PI3Kδ isoform is always expressed in AML cells, whereas the frequency of PI3Kγ expression is highly variable. The functions of these individual catalytic enzymes have not been fully resolved in AML, therefore using the PI3K p110δ and p110γ-targeted inhibitor IPI-145 (duvelisib) and specific p110δ and p110γ shRNA, we analysed the role of these two p110 subunits in human AML blast survival. The results show that PI3Kδ and PI3Kγ inhibition with IPI-145 has anti-proliferative activity in primary AML cells by inhibiting the activity of AKT and MAPK. Pre-treatment of AML cells with IPI-145 inhibits both adhesion and migration of AML blasts to bone marrow stromal cells. Using shRNA targeted to the individual isoforms we demonstrated that p110δ-knockdown had a more significant anti-proliferative effect on AML cells, whereas targeting p110γ-knockdown significantly inhibited AML migration. The results demonstrate that targeting both PI3Kδ and PI3Kγ to inhibit AML-BMSC interactions provides a biologic rationale for the pre-clinical evaluation of IPI-145 in AML.


Assuntos
Células da Medula Óssea/citologia , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Mesenquimais/citologia , Adesão Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Impressões Digitais de DNA , Regulação Leucêmica da Expressão Gênica , Humanos , Isoquinolinas/farmacologia , Leucemia Mieloide Aguda/genética , Fosforilação , Purinas/farmacologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Resultado do Tratamento
8.
Lancet Haematol ; 2(5): e204-11, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26688095

RESUMO

BACKGROUND: Roughly 80% of patients with acute myeloid leukaemia have high activity of Bruton's tyrosine-kinase (BTK) in their blast cells compared with normal haemopoietic cells, rendering the cells sensitive to the oral BTK inhibitor ibrutinib in vitro. We aimed to develop the biological understanding of the BTK pathway in acute myeloid leukaemia to identify clinically relevant diagnostic information that might define a subset of patients that should respond to ibrutinib treatment. METHODS: We obtained acute myeloid leukaemia blast cells from unselected patients attending our UK hospital between Feb 19, 2010, and Jan 20, 2014. We isolated primary acute myeloid leukaemia blast cells from heparinised blood and human peripheral blood mononuclear cells to establish the activity of BTK in response to CD117 activation. Furthermore, we investigated the effects of ibrutinib on CD117-induced BTK activation, downstream signalling, adhesion to primary bone-marrow mesenchymal stromal cells, and proliferation of primary acute myeloid leukaemia blast cells. We used the Mann-Whitney U test to compare results between groups. FINDINGS: We obtained acute myeloid leukaemia blast cells from 29 patients. Ibrutinib significantly inhibited CD117-mediated proliferation of primary acute myeloid leukaemia blast cells (p=0·028). CD117 activation increased BTK activity by inducing phosphorylated BTK in patients with CD117-positive acute myeloid leukaemia. Furthermore, ibrutinib inhibited CD117-induced activity of BTK and downstream kinases at a concentration of 100 nM or more. CD117-mediated adhesion of CD117-expressing blast cells to bone-marrow stromal cells was significantly inhibited by Ibrutinib at 500 nM (p=0·028) INTERPRETATION: As first-in-man clinical trials of ibrutinib in patients with acute myeloid leukaemia commence, the data suggest not all patients will respond. Our findings show that BTK has specific pro-tumoural biological actions downstream of surface CD117 activation, which are inhibited by ibrutinib. Accordingly, we propose that patients with acute myeloid leukaemia whose blast cells express CD117 should be considered for forthcoming clinical trials of ibrutinib. FUNDING: Worldwide Cancer Research, The Big C, UK National Institutes for Health Research, the Humane Research Trust, the Department of Higher Education and Research of the Libyan Government, and Norwich Research Park.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Transdução de Sinais , Adenina/análogos & derivados , Adulto , Tirosina Quinase da Agamaglobulinemia , Idoso , Idoso de 80 Anos ou mais , Linfócitos B/citologia , Feminino , Humanos , Leucócitos Mononucleares/citologia , Masculino , Pessoa de Meia-Idade , Piperidinas , Proteínas Proto-Oncogênicas c-kit
9.
Sci Rep ; 5: 12949, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26292723

RESUMO

Approximately 20% of patients with acute myeloid leukaemia (AML) have a mutation in FMS-like-tyrosine-kinase-3 (FLT3). FLT3 is a trans-membrane receptor with a tyrosine kinase domain which, when activated, initiates a cascade of phosphorylated proteins including the SRC family of kinases. Recently our group and others have shown that pharmacologic inhibition and genetic knockdown of Bruton's tyrosine kinase (BTK) blocks AML blast proliferation, leukaemic cell adhesion to bone marrow stromal cells as well as migration of AML blasts. The anti-proliferative effects of BTK inhibition in human AML are mediated via inhibition of downstream NF-κB pro-survival signalling however the upstream drivers of BTK activation in human AML have yet to be fully characterised. Here we place the FLT3-ITD upstream of BTK in AML and show that the BTK inhibitor ibrutinib inhibits the survival and proliferation of FLT3-ITD primary AML blasts and AML cell lines. Furthermore ibrutinib inhibits the activation of downstream kinases including MAPK, AKT and STAT5. In addition we show that BTK RNAi inhibits proliferation of FLT3-ITD AML cells. Finally we report that ibrutinib reverses the cyto-protective role of BMSC on FLT3-ITD AML survival. These results argue for the evaluation of ibrutinib in patients with FLT3-ITD mutated AML.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Proteínas Tirosina Quinases/metabolismo , Tirosina Quinase 3 Semelhante a fms/química , Tirosina Quinase 3 Semelhante a fms/metabolismo , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia , Apoptose/efeitos dos fármacos , Crise Blástica/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Daunorrubicina/farmacologia , Humanos , Mutação/genética , Piperidinas , Estrutura Terciária de Proteína , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Cell Cycle ; 14(14): 2367-75, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25565020

RESUMO

Multiple Myeloma (MM) is a haematologic malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. Over the last 10-15 y the introduction of the proteasome-inhibitor bortezomib has improved MM prognosis, however relapse due to bortezomib-resistance is inevitable and the disease, at present, remains incurable. To model bortezomib-resistant MM we generated bortezomib-resistant MM cell lines (n = 4 ) and utilised primary malignant plasma cells from patients relapsing after bortezomib treatment (n = 6 ). We identified enhanced Bruton's tyrosine kinase (BTK) activity in bortezomib-resistant MM cells and found that inhibition of BTK, either pharmacologically with ibrutinib (0.5 µM) or via lenti-viral miRNA-targeted BTK interference, re-sensitized previously bortezomib-resistant MM cells to further bortezomib therapy at a physiologically relevant concentration (5 nM). Further analysis of pro-survival signaling revealed a role for the NF-κB p65 subunit in MM bortezomib-resistance, thus a combination of BTK and NF-κB p65 inhibition, either pharmacologically or via further lenti-viral miRNA NF-κB p65 interference, also restored sensitivity to bortezomib, significantly reducing cell viability (37.5 ± 6 .9 %, ANOVA P ≤ 0 .001). Accordingly, we propose the clinical evaluation of a bortezomib/ibrutinib combination therapy, including in patients resistant to single-agent bortezomib.


Assuntos
Bortezomib/toxicidade , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Proteassoma/toxicidade , Proteínas Tirosina Quinases/metabolismo , Pirazóis/toxicidade , Pirimidinas/toxicidade , Fator de Transcrição RelA/metabolismo , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia , Bortezomib/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , MicroRNAs/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Recidiva Local de Neoplasia , Piperidinas , Inibidores de Proteassoma/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Células Tumorais Cultivadas
11.
Oncotarget ; 5(20): 9930-8, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25294819

RESUMO

Pharmacological targeting of BTK using ibrutinib has recently shown encouraging clinical activity in a range of lymphoid malignancies. Recently we reported that ibrutinib inhibits human acute myeloid leukemia (AML) blast proliferation and leukemic cell adhesion to the surrounding bone marrow stroma cells. Here we report that in human AML ibrutinib, in addition, functions to inhibit SDF1/CXCR4-mediated AML migration at concentrations achievable in vivo. It has previously been shown that SDF1/CXCR4-induced migration is dependent on activation of downstream BTK in chronic lymphocytic leukaemia (CLL) and multiple myeloma. Here we show that SDF-1 induces BTK phosphorylation and downstream MAPK signalling in primary AML blast. Furthermore, we show that ibrutinib can inhibit SDF1-induced AKT and MAPK activation. These results reported here provide a molecular mechanistic rationale for clinically evaluating BTK inhibition in AML patients and suggests that in some AML patients the blasts count may initially rise in response to ibrutinib therapy, analgous to similar clinical observations in CLL.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Adenina/análogos & derivados , Adulto , Tirosina Quinase da Agamaglobulinemia , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Piperidinas , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo
12.
Biochem Soc Trans ; 42(4): 747-51, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25109952

RESUMO

Human leukaemia cells have an often unique ability to either undergo apoptotic cell death mechanisms or, at other times, undergo proliferative expansion, sometimes to the same stimulus such as the pluripotent cytokine TNFα (tumour necrosis factor α). This potential for life/death switching helps us to understand the molecular signalling machinery that underlies these cellular processes. Furthermore, looking at the involvement of these switching signalling pathways that may be aberrant in leukaemia informs us of their importance in cancer tumorigenesis and how they may be targeted pharmacologically to treat various types of human leukaemias. Furthermore, these important pathways may play a crucial role in acquired chemotherapy resistance and should be studied further to overcome in the clinic many drug-resistant forms of blood cancers. In the present article, we uncover the relationship that exists in human leukaemia life/death switching between the anti-apoptotic pro-inflammatory transcription factor NF-κB (nuclear factor κB) and the cytoprotective antioxidant-responsive transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2). We also discuss recent findings that reveal a major role for Btk (Bruton's tyrosine kinase) in both lymphocytic and myeloid forms of human leukaemias and lymphomas.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Tirosina Quinase da Agamaglobulinemia , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição/metabolismo
13.
Blood ; 123(8): 1229-38, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24307721

RESUMO

Bruton's tyrosine kinase (BTK) is a cytoplasmic protein found in all hematopoietic cell lineages except for T cells. BTK mediates signaling downstream of a number of receptors. Pharmacologic targeting of BTK using ibrutinib (previously PCI-32765) has recently shown encouraging clinical activity in a range of lymphoid malignancies. This study reports for the first time that ibrutinib inhibits blast proliferation from human acute myeloid leukemia (AML) and that treatment with ibrutinib significantly augmented cytotoxic activities of standard AML chemotherapy cytarabine or daunorubicin. Here we describe that BTK is constitutively phosphorylated in the majority of AML samples tested, with BTK phosphorylation correlating highly with the cell's cytotoxic sensitivity toward ibrutinib. BTK-targeted RNAi knockdown reduced colony-forming capacity of primary AML blasts and proliferation of AML cell lines. We showed that ibrutinib binds at nanomolar range to BTK. Furthermore, we showed ibrutinib's antiproliferative effects in AML are mediated via an inhibitory effect on downstream nuclear factor-κB survival pathways. Moreover, ibrutinib inhibited AML cell adhesion to bone marrow stroma. Furthermore, these effects of ibrutinib in AML were seen at comparable concentrations efficacious in chronic lymphocytic leukemia. These results provide a biological rationale for clinical evaluation of BTK inhibition in AML patients.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Adenina/análogos & derivados , Adulto , Tirosina Quinase da Agamaglobulinemia , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Fosforilação/fisiologia , Piperidinas , Proteínas Tirosina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Cell Cycle ; 12(13): 2144-53, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23759586

RESUMO

Dexamethasone is a key front-line chemotherapeutic for B-cell malignant multiple myeloma (MM). Dexamethasone modulates MM cell survival signaling but fails to induce marked cytotoxicity when used as a monotherapy. We demonstrate here the mechanism behind this insufficient responsiveness of MM cells toward dexamethasone, revealing in MM a dramatic anti-apoptotic role for microRNA (miRNA)-125b in the insensitivity toward dexamethasone-induced apoptosis. MM cells responding to dexamethasone exhibited enhanced expression of oncogenic miR-125b. Dexamethasone also induced expression of miR-34a, which acts to suppress SIRT1 deacetylase, and thus allows maintained acetylation and inactivation of p53. p53 mRNA is also suppressed by miR-125b targeting. Reporter assays showed that both these dexamethasone-induced miRNAs act downstream of their target genes to prevent p53 tumor suppressor actions and, ultimately, resist cytotoxic responses in MM. Use of antisense miR-125b transcripts enhanced expression of pro-apoptotic p53, repressed expression of anti-apoptotic SIRT1 and, importantly, significantly enhanced dexamethasone-induced cell death responses in MM. Pharmacological manipulations showed that the key regulation enabling complete dexamethasone sensitivity in MM cells lies with miR-125b. In summary, dexamethasone-induced miR-125b induces cell death resistance mechanisms in MM cells via the p53/miR-34a/SIRT1 signaling network and provides these cells with an enhanced level of resistance to cytotoxic chemotherapeutics. Clearly, such anti-apoptotic mechanisms will need to be overcome to more effectively treat nascent, refractory and relapsed MM patients. These mechanisms provide insight into the role of miRNA regulation of apoptosis and their promotion of MM cell proliferative mechanisms.


Assuntos
Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , MicroRNAs/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , MicroRNAs/metabolismo , Mieloma Múltiplo , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Ativação Transcricional/efeitos dos fármacos , Transcriptoma , Proteína Supressora de Tumor p53/metabolismo
15.
Cell Signal ; 25(1): 106-12, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22975686

RESUMO

Ibrutinib (previously known as PCI-32765) has recently shown encouraging clinical activity in chronic lymphocytic leukaemia (CLL) effecting cell death through inhibition of Bruton's tyrosine kinase (BTK). In this study we report for the first time that ibrutinib is cytotoxic to malignant plasma cells from patients with multiple myeloma (MM) and furthermore that treatment with ibrutinib significantly augments the cytotoxic activity of bortezomib and lenalidomide chemotherapies. We describe that the cytotoxicity of ibrutinib in MM is mediated via an inhibitory effect on the nuclear factor-κB (NF-κB) pathway. Specifically, ibrutinib blocks the phosphorylation of serine-536 of the p65 subunit of NF-κB, preventing its nuclear translocation, resulting in down-regulation of anti-apoptotic proteins Bcl-xL, FLIP(L) and survivin and culminating in caspase-mediated apoptosis within the malignant plasma cells. Taken together these data provide a platform for clinical trials of ibrutinib in myeloma and a rationale for its use in combination therapy, particularly with bortezomib.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirazinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Talidomida/análogos & derivados , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia , Amidas/farmacologia , Bortezomib , Caspases/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Lenalidomida , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Inibidor de NF-kappaB alfa , Nitrilas/farmacologia , Fosforilação , Piperidinas , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Talidomida/farmacologia , Células Tumorais Cultivadas
16.
Blood ; 120(26): 5188-98, 2012 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23077289

RESUMO

NF-E2-related factor 2 (Nrf2) transcription factor regulates a range of cytoprotective transcriptional responses, preventing further cellular injury by removing biochemical damage and renewing tissue. Here we show that acute myeloid leukemia (AML) cells possess greater constitutive nuclear levels of Nrf2 than normal control CD34(+) cells because of an imbalance between mRNA expression levels of Nrf2 and its inhibitor Keap1 but not through their somatic mutation. Elevated Nrf2 was reduced by NF-κB inhibitors. Using promoter assays, ChIP and siRNA knockdown, we demonstrated NF-κB subunits p50 and p65 induce transcription of Nrf2 in AML cells at a specific promoter κB-site and that long-term lentiviral miRNA-knockdown of Nrf2 significantly reduced clonogenicity of AML patient cells and improved their chemotherapeutic responsiveness. Normal physiologic Nrf2 protects cells from damage, but here we have exposed aberrant continuous nuclear activation of Nrf2 in AML that allows cell survival, even against cytotoxic chemotherapeutics. We show for the first time that Nrf2, an important regulator of several biologic processes involved in the progression of cancer, has abnormal NF-κB-driven constitutive expression in AML. Such a mechanism allows for a greater cytoprotective response in human AML cells and encourages their evasion of chemotherapy-induced cytotoxicity, which is necessary for improved clinical outcomes.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Leucemia Mieloide Aguda/genética , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Regulação Leucêmica da Expressão Gênica/genética , Células HL-60 , Humanos , Masculino , Pessoa de Meia-Idade , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Células Tumorais Cultivadas , Células U937 , Regulação para Cima/genética
17.
Am J Cancer Res ; 2(1): 65-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22206046

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by binding to complementary sequences in mRNAs encoding downstream target genes. A large variety of cellular processes, including differentiation, development, apoptosis and cell cycle progression, are dependent on miRNA-mediated suppression of gene expression for their regulation. As such, it is unsurprising that these small RNA molecules are associated with signaling networks that are often altered in various diseases, including many blood cancers. One such network is the nuclear factor-κB (NF-κB) pathways that universally stimulate transcription of proteins which generally promote cell survival, inhibit apoptosis, allow cellular growth, induce angiogenesis and generate many pro-inflammatory responses. The NF-κB signalling pathway is often constitutively activated in blood cell cancers including myelodysplastic syndrome (MDS), acute myeloid leukaemia (AML), acute lymphocytic leukaemia (ALL), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), lymphomas and in multiple myeloma (MM). This review focuses on the function of miRNAs that directly target NF-κB signaling cascade. Recent findings that connect this pathway through various miRNA families to human blood cancers are reviewed, and support for using miRNA-based therapy as a novel method to counteract this tumour-promoting signalling event is discussed.

18.
Oncotarget ; 2(9): 658-68, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21911919

RESUMO

Haem oxygenase-1 (HO-1) is increasingly regarded as a pro-tumoral target in the treatment of human cancers. Currently, little is known about HO-1 and its role in human acute myeloid leukaemia (AML) to regulate apoptosis in response to chemotherapy. Recently, we showed that HO-1 protects AML samples from tumour necrosis factor-α (TNF)-induced apoptosis - it being regulated by transcription factors Nrf2, NF-ĸB and AP-1. This study aims to analyse the role of HO-1 in regulating apoptosis in AML cells in response to two front-line chemotherapeutic agents used for AML, cytarabine and daunorubicin. Here we show that HO-1 expression in AML samples was increased in response to both cytarabine and daunorubicin treatment, and micro RNA (miRNA) silenced HO-1 expression in combination with either daunorubicin or cytarabine induced a greater apoptotic responses in AML cells. Moreover, we showed that both daunorubicin and cytarabine induced reactive oxygen species (ROS) accumulation to induce apoptosis in AML. However, ROS-dependent induction of HO-1 was limiting the apoptotic response that is seen in AML towards cytarabine and daunorubicin treatment. These findings suggest concurrent inhibition of HO-1 expression in conjunction with chemotherapeutic treatment would improve the number of cases who reach complete remission.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citarabina/farmacologia , Daunorrubicina/farmacologia , Heme Oxigenase-1/biossíntese , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citarabina/administração & dosagem , Daunorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Feminino , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/genética , Heme Oxigenase-1/imunologia , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/metabolismo
19.
Cell Cycle ; 10(7): 1067-72, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21403465

RESUMO

Death-receptor induced apoptosis is regulated by FLIP [FLICE (Fas-associated protein with death domain-like IL-1ß-converting enzyme)-inhibitory protein] via modification of caspase-8 activation. As an important modulator of apoptosis, the long isoform, FLIPL, regulates life and death in many various types of normal and tumor cells and tissues to render resistance to death receptor-mediated apoptosis. In addition, FLIPL has been shown to be involved in regulation of intrinsic (mitochondrial) pathways of apoptosis as well as regulating other proteins involved in cytoprotection and cell cycle progression. Therefore, understanding the role of FLIPL in complex regulatory networks of cell survival/death mechanisms is vital for future developments to control diseases such as cancer. Here, we shown that silencing FLIPL in HEK 293 cells changed the expression levels of proteins that are involved in both extrinsic and intrinsic apoptosis, as well as regulating tumor necrosis factor-α (TNF)-mediated apoptotic patterns. We also show that FLIPL-silenced cells have a lower rate of proliferation and cell cycle progression when compared to control cells. Moreover, treatment with TNF restored proliferation rates in FLIPL-silenced cells back to more normal levels when compared to control cells. These results suggest that cells have evolved complex compensatory mechanisms to overcome the absence of a key apoptotic regulatory proteins.


Assuntos
Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Regulação da Expressão Gênica/fisiologia , Western Blotting , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular , Proliferação de Células , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Marcação de Genes , Vetores Genéticos , Humanos , Lentivirus , MicroRNAs/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/metabolismo
20.
Oncotarget ; 1(5): 359-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21307400

RESUMO

Acute myeloid leukemia (AML) comprises a heterogeneous group of clonal disorders of hematopoietic progenitors. We previously showed that heme oxygenase-1 (HO-1/Hsp32) underlies resistance of AML to TNF-induced apoptosis. Here we show for the first time that the modulatory protein, FLICE-inhibitory protein (FLIP) indirectly regulates induction of HO-1 in response to TNF in human AML blasts, but not non-cancerous control cells. In AML cells, TNF-induced FLIP expression was an NF-κB-dependent event, and silencing of FLIP isoforms (FLIPL, FLIPS and FLIPR) induced pro-apoptotic responses to TNF, with FLIPL knock-down providing the greatest apoptotic switch. However, FLIPL knock-down consequently increased expression of HO-1; a response that occurred in AML (but not non-cancerous) cells to protect a proportion of them from apoptotic death. Our results show that increases in HO-1 induced an apoptotic-resistant form in AML cells in the absence of FLIPL. This is the first time that FLIPL has been shown to regulate the expression of HO-1. These data reveal unique regulatory networks in cancerous AML cells whereby FLIP regulation of HO-1 provides AML cells with secondary anti-apoptotic protection against extrinsic factors (eg TNF/chemotherapies) that try to switch on death signals in these highly death-resistant cells. Future AML therapies should target these mechanisms.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Resistencia a Medicamentos Antineoplásicos , Heme Oxigenase-1/metabolismo , Leucemia Mieloide Aguda/enzimologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Células HL-60 , Heme Oxigenase-1/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , NF-kappa B/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA