Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
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
2.
Blood ; 133(5): 446-456, 2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30401703

RESUMO

Acute myeloid leukemia (AML) is an age-related disease that is highly dependent on the bone marrow (BM) microenvironment. With increasing age, tissues accumulate senescent cells, characterized by an irreversible arrest of cell proliferation and the secretion of a set of proinflammatory cytokines, chemokines, and growth factors, collectively known as the senescence-associated secretory phenotype (SASP). Here, we report that AML blasts induce a senescent phenotype in the stromal cells within the BM microenvironment and that the BM stromal cell senescence is driven by p16INK4a expression. The p16INK4a-expressing senescent stromal cells then feed back to promote AML blast survival and proliferation via the SASP. Importantly, selective elimination of p16INK4a+ senescent BM stromal cells in vivo improved the survival of mice with leukemia. Next, we find that the leukemia-driven senescent tumor microenvironment is caused by AML-induced NOX2-derived superoxide. Finally, using the p16-3MR mouse model, we show that by targeting NOX2 we reduced BM stromal cell senescence and consequently reduced AML proliferation. Together, these data identify leukemia-generated NOX2-derived superoxide as a driver of protumoral p16INK4a-dependent senescence in BM stromal cells. Our findings reveal the importance of a senescent microenvironment for the pathophysiology of leukemia. These data now open the door to investigate drugs that specifically target the "benign" senescent cells that surround and support AML.


Assuntos
Medula Óssea/patologia , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Leucemia Mieloide Aguda/patologia , Microambiente Tumoral , Animais , Medula Óssea/metabolismo , Proliferação de Células , Técnicas de Cocultura , Feminino , Humanos , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos Endogâmicos C57BL , NADPH Oxidase 2/metabolismo , Superóxidos/metabolismo , Células Tumorais Cultivadas
4.
J Hematol Oncol ; 11(1): 66, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29769142

RESUMO

Multiple myeloma (MM) remains an incurable malignancy despite the recent advancements in its treatment. The protective effects of the niche in which it develops has been well documented; however, little has been done to investigate the MM cell's ability to 're-program' cells within its environment to benefit disease progression. Here, we show that MM-derived macrophage migratory inhibitory factor (MIF) stimulates bone marrow stromal cells to produce the disease critical cytokines IL-6 and IL-8, prior to any cell-cell contact. Furthermore, we provide evidence that this IL-6/8 production is mediated by the transcription factor cMYC. Pharmacological inhibition of cMYC in vivo using JQ1 led to significantly decreased levels of serum IL-6-a highly positive prognostic marker in MM patients. CONCLUSIONS: Our presented findings show that MM-derived MIF causes BMSC secretion of IL-6 and IL-8 via BMSC cMYC. Furthermore, we show that the cMYC inhibitor JQ1 can reduce BMSC secreted IL-6 in vivo, irrespective of tumor burden. These data provide evidence for the clinical evaluation of both MIF and cMYC inhibitors in the treatment of MM.


Assuntos
Células da Medula Óssea/patologia , Interleucina-6/metabolismo , Oxirredutases Intramoleculares/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Mieloma Múltiplo/química , Células Estromais/patologia , Humanos , Interleucina-8/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
5.
Oncogene ; 37(20): 2676-2686, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29487418

RESUMO

Approximately 80% of patients diagnosed with acute myeloid leukemia (AML) die as a consequence of failure to eradicate the tumor from the bone marrow microenvironment. We have recently shown that stroma-derived interleukin-8 (IL-8) promotes AML growth and survival in the bone marrow in response to AML-derived macrophage migration inhibitory factor (MIF). In the present study we show that high constitutive expression of MIF in AML blasts in the bone marrow is hypoxia-driven and, through knockdown of MIF, HIF1α and HIF2α, establish that hypoxia supports AML tumor proliferation through HIF1α signaling. In vivo targeting of leukemic cell HIF1α inhibits AML proliferation in the tumor microenvironment through transcriptional regulation of MIF, but inhibition of HIF2α had no measurable effect on AML blast survival. Functionally, targeted inhibition of MIF in vivo improves survival in models of AML. Here we present a mechanism linking HIF1α to a pro-tumoral chemokine factor signaling pathway and in doing so, we establish a potential strategy to target AML.


Assuntos
Medula Óssea/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Oxirredutases Intramoleculares/genética , Leucemia Mieloide Aguda/genética , Fatores Inibidores da Migração de Macrófagos/genética , Regulação para Cima , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Técnicas de Silenciamento de Genes , Humanos , Oxirredutases Intramoleculares/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos , Transplante de Neoplasias , Transdução de Sinais , Microambiente Tumoral
6.
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
7.
Cancers (Basel) ; 9(4)2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28350342

RESUMO

Despite the development of novel treatments in the past 15 years, many blood cancers still remain ultimately fatal and difficult to treat, particularly acute myeloid leukaemia (AML) and multiple myeloma (MM). While significant progress has been made characterising small-scale genetic mutations and larger-scale chromosomal translocations that contribute to the development of various blood cancers, less is understood about the complex microenvironment of the bone marrow (BM), which is known to be a key player in the pathogenesis of chronic lymphocytic leukaemia (CLL), AML and MM. This niche acts as a sanctuary for the cancerous cells, protecting them from chemotherapeutics and encouraging clonal cell survival. It does this by upregulating a plethora of signalling cascades within the malignant cell, with the phosphatidylinositol-3-kinase (PI3K) pathway taking a critical role. This review will focus on how the PI3K pathway influences disease progression and the individualised role of the PI3K subunits. We will also summarise the current clinical trials for PI3K inhibitors and how these trials impact the treatment of blood cancers.

8.
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
9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA