Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Front Immunol ; 15: 1278807, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38576613

RESUMO

Pancreatic inflammation is a risk factor for the development of pancreatic cancer. Increased presence of inflammatory macrophages can be found in response to a KRAS mutation in acinar cells or in response to experimentally-induced pancreatitis. Inflammatory macrophages induce pancreatic acinar cells to undergo dedifferentiation to a duct-like progenitor stage, a process called acinar-to-ductal metaplasia (ADM). Occurrence of ADM lesions are believed to be the initiating event in tumorigenesis. Here we will discuss how macrophage-induced oxidative stress contributes to ADM and how ADM cells shape the fibrotic stroma needed for further progression.


Assuntos
Neoplasias Pancreáticas , Pancreatite , Humanos , Espécies Reativas de Oxigênio , Transdução de Sinais/genética , Neoplasias Pancreáticas/patologia , Pancreatite/patologia , Macrófagos/patologia
2.
iScience ; 26(6): 106820, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37250781

RESUMO

The innate immune system has a key role in pancreatic cancer initiation, but the specific contribution of different macrophage populations is still ill-defined. While inflammatory (M1) macrophages have been shown to drive acinar-to-ductal metaplasia (ADM), a cancer initiating event, alternatively activated (M2) macrophages have been attributed to lesion growth and fibrosis. Here, we determined cytokines and chemokines secreted by both macrophage subtypes. Then, we analyzed their role in ADM initiation and lesion growth, finding that while M1 secrete TNF, CCL5, and IL-6 to drive ADM, M2 induce this dedifferentiation process via CCL2, but the effects are not additive. This is because CCL2 induces ADM by generating ROS and upregulating EGFR signaling, thus using the same mechanism as cytokines from inflammatory macrophages. Therefore, while effects on ADM are not additive between macrophage polarization types, both act synergistically on the growth of low-grade lesions by activating different MAPK pathways.

3.
iScience ; 25(5): 104327, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35602933

RESUMO

Desmoplasia around pancreatic lesions is a barrier for immune cells and a hallmark of developing and established pancreatic cancer. However, the contribution of the innate immune system to this process is ill-defined. Using the KC mouse model and primary cells in vitro, we show that alternatively activated macrophages (AAM) crosstalk with pancreatic lesion cells and pancreatic stellate cells (PSCs) to mediate fibrosis and progression of lesions. TGFß1 secreted by AAM not only drives activation of quiescent PSCs but also in activated PSCs upregulates expression of TIMP1, a factor previously shown as crucial in fibrosis. Once activated, PSCs auto-stimulate proliferation via CXCL12. Furthermore, we found that TIMP1/CD63 signaling mediates PanIN lesion growth and TGFß1 contributes to a cadherin switch and drives structural collapse of lesions, indicating a potential progression step. Taken together, our data indicate TGFß1 produced by Ym1+ AAM as a major driver of processes that initiate the development of pancreatic cancer.

4.
Antioxidants (Basel) ; 11(1)2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-35052641

RESUMO

Pancreatic acinar-to-ductal metaplasia (ADM) is a reversible process that occurs after pancreatic injury, but becomes permanent and leads to pancreatic lesions in the presence of an oncogenic mutation in KRAS,. While inflammatory macrophage-secreted chemokines, growth factors that activate epidermal growth factor receptor (EGFR) and oncogenic KRAS have been implicated in the induction of ADM, it is currently unclear whether a common underlying signaling mechanism exists that drives this process. In this study, we show that different inducers of ADM increase levels of hydrogen peroxide, most likely generated at the mitochondria, and upregulate the expression of Protein Kinase D1 (PKD1), a kinase that can be activated by hydrogen peroxide. PKD1 expression in acinar cells affects their survival and mediates ADM, which is in part due to the PKD1 target NF-κB. Overall, our data implicate ROS-PKD1 signaling as a common feature of different inducers of pancreatic ADM.

5.
iScience ; 24(1): 102019, 2021 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-33521594

RESUMO

Doublecortin-like kinase 1 (DCLK1)-positive pancreatic cancer stem cells develop at a precancerous stage and may contribute to the lack of efficacy of pancreatic cancer therapy. Although PanIN cells express oncogenic KRas and have an increased activity of epidermal growth factor receptor (EGFR), we demonstrate that, in DCLK1+ PanIN cells, EGFR signaling is not propagated to the nucleus. Mimicking blockage of EGFR with erlotinib in PanIN organoid culture or in p48cre;KrasG12D mice led to a significant increase in DCLK1+ PanIN cells. As a mechanism of how EGFR inhibition leads to formation of DCLK1+ cells, we identify an increase in hydrogen peroxide contributing to activation of Protein Kinase D1 (PKD1). Active PKD1 then drives stemness and abundance of DCLK1+ cells in lesions. Our data suggest a signaling mechanism that leads to the development of DCLK1+ pancreatic cancer stem cells, which can be exploited to target this population in potential therapeutic approaches.

6.
Sci Rep ; 9(1): 16588, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31719634

RESUMO

Current treatment options for patients with pancreatic cancer are suboptimal, resulting in a five year survival rate of about 9%. Difficulties with treatment are due to an immunosuppressive, fibrotic tumor microenvironment that prevents drugs from reaching tumor cells, but also to the limited efficacy of existing FDA-approved chemotherapeutic compounds. We here show that the nucleoside analog Sangivamycin and its closely-related compound Toyocamycin target PDA cell lines, and are significantly more efficient than Gemcitabine. Using KINOMEscan screening, we identified the kinase Haspin, which is overexpressed in PDA cell lines and human PDA samples, as a main target for both compounds. Inhibition of Haspin leads to a decrease in Histone H3 phosphorylation and prevents Histone H3 binding to survivin, thus providing mechanistic insight of how Sangivamycin targets cell proliferation, mitosis and induces apoptotic cell death. In orthotopically implanted tumors in mice, Sangivamycin was efficient in decreasing the growth of established tumors. In summary, we show that Sangivamycin and derivatives can be an efficient new option for treatment of PDA.


Assuntos
Apoptose , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Nucleosídeos de Pirimidina/farmacologia , Survivina/antagonistas & inibidores , Animais , Antibióticos Antineoplásicos/farmacologia , Biomarcadores Tumorais , Proliferação de Células , Histonas/genética , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação , Prognóstico , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Survivina/genética , Survivina/metabolismo , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 9(91): 36358-36370, 2018 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-30555634

RESUMO

Phosphatidylinositol-4-phosphate 5-kinase type-1C (PIP5K1C) is a lipid kinase that regulates focal adhesion dynamics and cell attachment through site-specific formation of phosphatidylinositol-4,5-bisphosphate (PI4,5P2). By comparing normal breast tissue to carcinoma in situ and invasive ductal carcinoma subtypes, we here show that the phosphorylation status of PIP5K1C at serine residue 448 (S448) can be predictive for breast cancer progression to an aggressive phenotype, while PIP5K1C expression levels are not indicative for this event. PIP5K1C phosphorylation at S448 is downregulated in invasive ductal carcinoma, and similarly, the expression levels of PKD1, the kinase that phosphorylates PIP5K1C at this site, are decreased. Overall, since PKD1 is a negative regulator of cell migration and invasion in breast cancer, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.

8.
Sci Rep ; 7(1): 9524, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28842658

RESUMO

Dependent on their cellular localization, Protein Kinase D (PKD) enzymes regulate different processes including Golgi transport, cell signaling and response to oxidative stress. The localization of PKD within cells is mediated by interaction with different lipid or protein binding partners. With the example of PKD2, we here show that phosphorylation events can also contribute to localization of subcellular pools of this kinase. Specifically, in the present study, we show that tyrosine phosphorylation of PKD2 at residue Y87 defines its localization to the focal adhesions and leads to activation. This phosphorylation occurs downstream of RhoA signaling and is mediated via Src. Moreover, mutation of this residue blocks PKD2's interaction with Focal Adhesion Kinase (FAK). The presence and regulation of PKD2 at focal adhesions identifies a novel function for this kinase as a modulator of cell adhesion and migration.


Assuntos
Adesão Celular , Adesões Focais , Proteínas Quinases/metabolismo , Quinases da Família src/metabolismo , Movimento Celular , Imunofluorescência , Imuno-Histoquímica , Fosforilação , Proteína Quinase D2 , Proteína rhoA de Ligação ao GTP/metabolismo
9.
Cell Rep ; 19(7): 1322-1333, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28514653

RESUMO

The contributions of the innate immune system to the development of pancreatic cancer are still ill defined. Inflammatory macrophages can initiate metaplasia of pancreatic acinar cells to a duct-like phenotype (acinar-to-ductal metaplasia [ADM]), which then gives rise to pancreatic intraepithelial neoplasia (PanIN) when oncogenic KRas is present. However, it remains unclear when and how this inflammatory macrophage population is replaced by tumor-promoting macrophages. Here, we demonstrate the presence of interleukin-13 (IL-13), which can convert inflammatory into Ym1+ alternatively activated macrophages, at ADM/PanIN lesions. We further show that Ym1+ macrophages release factors, such as IL-1ra and CCL2, to drive pancreatic fibrogenesis and tumorigenesis. Treatment of mice expressing oncogenic KRas under an acinar cell-specific promoter with a neutralizing antibody for IL-13 significantly decreased the accumulation of alternatively activated macrophages at these lesions, resulting in decreased fibrosis and lesion growth.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Interleucina-13/metabolismo , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Polaridade Celular , Proliferação de Células , Fibrose , Inflamação/patologia , Macrófagos/patologia , Metaplasia , Camundongos , Testes de Neutralização , Ductos Pancreáticos/metabolismo
10.
Front Oncol ; 7: 41, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28361035

RESUMO

Due to alterations in their metabolic activity and decreased mitochondrial efficiency, cancer cells often show increased generation of reactive oxygen species (ROS), but at the same time, to avoid cytotoxic signaling and to facilitate tumorigenic signaling, have mechanism in place that keep ROS in check. This requires signaling molecules that convey increases in oxidative stress to signal to the nucleus to upregulate antioxidant genes. Protein kinase D1 (PKD1), the serine/threonine kinase, is one of these ROS sensors. In this mini-review, we highlight the mechanisms of how PKD1 is activated in response to oxidative stress, so far known downstream effectors, as well as the importance of PKD1-initiated signaling for development and progression of pancreatic cancer.

11.
Sci Rep ; 7(1): 887, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28428613

RESUMO

Protein kinases are essential molecules in life and their crucial function requires tight regulation. Many kinases are regulated via phosphorylation within their activation loop. This loop is embedded in the activation segment, which additionally contains the Mg2+ binding loop and a P + 1 loop that is important in substrate binding. In this report, we identify Abl-mediated phosphorylation of a highly conserved Tyr residue in the P + 1 loop of protein kinase D2 (PKD2) during oxidative stress. Remarkably, we observed that the three human PKD isoforms display very different degrees of P + 1 loop Tyr phosphorylation and we identify one of the molecular determinants for this divergence. This is paralleled by a different activation mechanism of PKD1 and PKD2 during oxidative stress. Tyr phosphorylation in the P + 1 loop of PKD2 increases turnover for Syntide-2, while substrate specificity and the role of PKD2 in NF-κB signaling remain unaffected. Importantly, Tyr to Phe substitution renders the kinase inactive, jeopardizing its use as a non-phosphorylatable mutant. Since large-scale proteomics studies identified P + 1 loop Tyr phosphorylation in more than 70 Ser/Thr kinases in multiple conditions, our results do not only demonstrate differential regulation/function of PKD isoforms under oxidative stress, but also have implications for kinase regulation in general.


Assuntos
Estresse Oxidativo , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Sequência Conservada , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , NF-kappa B/metabolismo , Peptídeos/metabolismo , Fosforilação , Domínios Proteicos , Proteína Quinase C/química , Proteína Quinase C/genética , Tirosina/genética , Tirosina/metabolismo
12.
Sci Rep ; 6: 35963, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27775029

RESUMO

Focal adhesions (FAs) are highly dynamic structures that are assembled and disassembled on a continuous basis. The balance between the two processes mediates various aspects of cell behavior, ranging from cell adhesion and spreading to directed cell migration. The turnover of FAs is regulated at multiple levels and involves a variety of signaling molecules and adaptor proteins. In the present study, we show that in response to integrin engagement, a subcellular pool of Protein Kinase D1 (PKD1) localizes to the FAs. PKD1 affects FAs by decreasing turnover and promoting maturation, resulting in enhanced cell adhesion. The effects of PKD1 are mediated through direct phosphorylation of FA-localized phosphatidylinositol-4-phosphate 5-kinase type-l γ (PIP5Klγ) at serine residue 448. This phosphorylation occurs in response to Fibronectin-RhoA signaling and leads to a decrease in PIP5Klγs' lipid kinase activity and binding affinity for Talin. Our data reveal a novel function for PKD1 as a regulator of FA dynamics and by identifying PIP5Klγ as a novel PKD1 substrate provide mechanistic insight into this process.


Assuntos
Adesão Celular , Adesões Focais/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Linhagem Celular , Humanos , Integrinas/metabolismo , Camundongos , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais
13.
Sci Rep ; 6: 33758, 2016 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-27649783

RESUMO

Increased expression of PRKD1 and its gene product protein kinase D1 (PKD1) are linked to oncogenic signaling in pancreatic ductal adenocarcinoma, but a direct functional relationship to oncogenic KRas has not been established so far. We here describe the PRKD1 gene promoter as a target for oncogenic KRas signaling. We demonstrate that KRas-induced activation of the canonical NF-κB pathway is one mechanism of how PRKD1 expression is increased and identify the binding sites for NF-κB in the PRKD1 promoter. Altogether, these results describe a novel mechanism governing PRKD1 gene expression in PDA and provide a functional link between oncogenic KRas, NF-κB and expression of PRKD1.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , NF-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas , Proteína Quinase C/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Humanos , NF-kappa B/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteína Quinase C/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
14.
Cell Rep ; 14(10): 2325-36, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947075

RESUMO

The development of pancreatic cancer requires the acquisition of oncogenic KRas mutations and upregulation of growth factor signaling, but the relationship between these is not well established. Here, we show that mutant KRas alters mitochondrial metabolism in pancreatic acinar cells, resulting in increased generation of mitochondrial reactive oxygen species (mROS). Mitochondrial ROS then drives the dedifferentiation of acinar cells to a duct-like progenitor phenotype and progression to PanIN. This is mediated via the ROS-receptive kinase protein kinase D1 and the transcription factors NF-κB1 and NF-κB2, which upregulate expression of the epidermal growth factor, its ligands, and their sheddase ADAM17. In vivo, interception of KRas-mediated generation of mROS reduced the formation of pre-neoplastic lesions. Hence, our data provide insight into how oncogenic KRas interacts with growth factor signaling to induce the formation of pancreatic cancer.


Assuntos
Células Acinares/metabolismo , Receptores ErbB/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína ADAM17/metabolismo , Células Acinares/citologia , Animais , Células Cultivadas , Fator de Crescimento Epidérmico/metabolismo , Humanos , Ligantes , Camundongos , Mutagênese Sítio-Dirigida , Subunidade p52 de NF-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Regulação para Cima
15.
Cell Metab ; 22(4): 536-7, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26445507

RESUMO

Although metabolic reprogramming is a hallmark of oncogenic transformation and growth of primary tumor cells, little is known about how metabolic alterations impact metastasis. In this issue of Cell Metabolism, Dupuy et al. (2015) show that primary breast cancer cells display metabolic heterogeneity, and that their distinct metabolic programs define their sites of metastasis.


Assuntos
Neoplasias da Mama/patologia , Engenharia Metabólica , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Feminino , Humanos
16.
Oncotarget ; 6(30): 29740-52, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26336132

RESUMO

Vasodilator-stimulated phosphoprotein (VASP) signaling is critical for dynamic actin reorganization processes that define the motile phenotype of cells. Here we show that VASP is generally highly expressed in normal breast tissue and breast cancer. We also show that the phosphorylation status of VASP at S322 can be predictive for breast cancer progression to an aggressive phenotype. Our data indicate that phosphorylation at S322 is gradually decreased from normal breast to DCIS, luminal/ER+, HER2+ and basal-like/TN phenotypes. Similarly, the expression levels of PKD2, the kinase that phosphorylates VASP at this site, are decreased in invasive ductal carcinoma samples of all three groups. Overall, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Ductal/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Serina/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal/genética , Carcinoma Ductal/patologia , Moléculas de Adesão Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Progressão da Doença , Células HeLa , Humanos , Immunoblotting , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Proteínas dos Microfilamentos/genética , Microscopia Confocal , Mutação , Invasividade Neoplásica , Fosfoproteínas/genética , Fosforilação , Prognóstico , Proteína Quinase D2 , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Serina/genética , Análise Serial de Tecidos
17.
Nat Commun ; 6: 6200, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25698580

RESUMO

The transdifferentiation of pancreatic acinar cells to a ductal phenotype (acinar-to-ductal metaplasia, ADM) occurs after injury or inflammation of the pancreas and is a reversible process. However, in the presence of activating Kras mutations or persistent epidermal growth factor receptor (EGF-R) signalling, cells that underwent ADM can progress to pancreatic intraepithelial neoplasia (PanIN) and eventually pancreatic cancer. In transgenic animal models, ADM and PanINs are initiated by high-affinity ligands for EGF-R or activating Kras mutations, but the underlying signalling mechanisms are not well understood. Here, using a conditional knockout approach, we show that protein kinase D1 (PKD1) is sufficient to drive the reprogramming process to a ductal phenotype and progression to PanINs. Moreover, using 3D explant culture of primary pancreatic acinar cells, we show that PKD1 acts downstream of TGFα and Kras, to mediate formation of ductal structures through activation of the Notch pathway.


Assuntos
Células Acinares/enzimologia , Células Acinares/patologia , Carcinoma in Situ/enzimologia , Reprogramação Celular , Progressão da Doença , Neoplasias Pancreáticas/enzimologia , Proteína Quinase C/metabolismo , Células Acinares/efeitos dos fármacos , Animais , Carcinoma in Situ/patologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Reprogramação Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Ductos Pancreáticos/efeitos dos fármacos , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/patologia , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores Notch/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Regulação para Cima/efeitos dos fármacos
18.
Cancer Discov ; 5(1): 52-63, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25361845

RESUMO

UNLABELLED: Desmoplasia and an inflammatory environment are defining features of pancreatic cancer. Unclear is how pancreatic cells that undergo oncogenic transformation can cross-talk with immune cells and how this contributes to the development of pancreatic lesions. Here, we demonstrate that pancreatic acinar cells expressing mutant KRAS can expedite their transformation to a duct-like phenotype by inducing local inflammation. Specifically, we show that KRAS(G12D) induces the expression of intercellular adhesion molecule-1 (ICAM-1), which serves as chemoattractant for macrophages. Infiltrating macrophages amplify the formation of KRAS(G12D)-caused abnormal pancreatic structures by remodeling the extracellular matrix and providing cytokines such as TNF. Depletion of macrophages or treatment with a neutralizing antibody for ICAM-1 in mice expressing oncogenic Kras under an acinar cell-specific promoter resulted in both a decreased formation of abnormal structures and decreased progression of acinar-to-ductal metaplasia to pancreatic intraepithelial neoplastic lesions. SIGNIFICANCE: We here show that oncogenic KRAS in pancreatic acinar cells upregulates the expression of ICAM-1 to attract macrophages. Hence, our results reveal a direct cooperative mechanism between oncogenic Kras mutations and the inflammatory environment to drive the initiation of pancreatic cancer.


Assuntos
Células Acinares/metabolismo , Genes ras , Molécula 1 de Adesão Intercelular/genética , Macrófagos/imunologia , Mutação , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Lesões Pré-Cancerosas , Células Acinares/patologia , Animais , Linhagem Celular Tumoral , Quimiotaxia de Leucócito , Citocinas/biossíntese , Modelos Animais de Doenças , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos/metabolismo , Camundongos , Neoplasias Pancreáticas/patologia , Peptídeo Hidrolases/biossíntese
19.
PLoS One ; 9(5): e98090, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24840177

RESUMO

BACKGROUND: Protein kinase D (PKD) enzymes regulate cofilin-driven actin reorganization and directed cell migration through both p21-activated kinase 4 (PAK4) and the phosphatase slingshot 1L (SSH1L). The relative contributions of different endogenous PKD isoforms to both signaling pathways have not been elucidated, sufficiently. METHODOLOGY/PRINCIPAL FINDINGS: We here analyzed two cell lines (HeLa and MDA-MB-468) that express the subtypes protein kinase D2 (PKD2) and protein kinase D3 (PKD3). We show that under normal growth conditions both isoforms can form a complex, in which PKD3 is basally-active and PKD2 is inactive. Basal activity of PKD3 mediates PAK4 activity and downstream signaling, but does not significantly inhibit SSH1L. This signaling constellation was required for facilitating directed cell migration. Activation of PKD2 and further increase of PKD3 activity leads to additional phosphorylation and inhibition of endogenous SSH1L. Net effect is a dramatic increase in phospho-cofilin and a decrease in cell migration, since now both PAK4 and SSH1L are regulated by the active PKD2/PKD3 complex. CONCLUSIONS/SIGNIFICANCE: Our data suggest that PKD complexes provide an interface for both cofilin regulatory pathways. Dependent on the activity of involved PKD enzymes signaling can be balanced to guarantee a functional cofilin activity cycle and increase cell migration, or imbalanced to decrease cell migration. Our data also provide an explanation of how PKD isoforms mediate different effects on directed cell migration.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Movimento Celular/fisiologia , Complexos Multiproteicos/metabolismo , Isoformas de Proteínas/metabolismo , Proteína Quinase C/genética , Transdução de Sinais/fisiologia , Movimento Celular/genética , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Complexos Multiproteicos/genética , Oligonucleotídeos/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Quinases Ativadas por p21/metabolismo
20.
Breast Cancer Res Treat ; 144(1): 79-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24510012

RESUMO

The treatment of patients with invasive breast cancer remains a major issue because of the acquisition of drug resistance to conventional chemotherapy. Here we propose a new therapeutic strategy by combining DNA methyltransferase inhibitors (DMTIs) with suramin. Cytotoxic effects of suramin or combination treatment with DMTIs were determined in highly invasive breast cancer cell lines MDA-MB-231, BT-20 and HCC1954, or control cells. In addition, effects on cell invasion were determined in 3-dimensional cell culture assays. DMTI-mediated upregulation of Protein Kinase D1 (PKD1) expression was shown by Western blotting. Effects of suramin on PKD1 activity was determined in vitro and in cells. The importance of PKD1 in mediating the effects of such combination treatment in cell invasion was demonstrated using 3D cell culture assays. A proof of principal animal experiment was performed showing that PKD1 is critical for breast cancer growth. We show that when used in combination, suramin and DMTIs impair the invasive phenotype of breast cancer cells. We show that PKD1, a kinase that previously has been described as a suppressor of tumor cell invasion, is an interface for both FDA-approved drugs, since the additive effects observed are due to DMTI-mediated re-expression and suramin-induced activation of PKD1. Our data reveal a mechanism of how a combination treatment with non-toxic doses of suramin and DMTIs may be of therapeutic benefit for patients with aggressive, multi-drug resistant breast cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Animais , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Decitabina , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Ftalimidas/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suramina/administração & dosagem , Triptofano/administração & dosagem , Triptofano/análogos & derivados , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA