345 results on '"Peter Storz"'
Search Results
2. Cytokine CCL9 Mediates Oncogenic KRAS-Induced Pancreatic Acinar-to-Ductal Metaplasia by Promoting Reactive Oxygen Species and Metalloproteinases
- Author
-
Geou-Yarh Liou, Crystal J. Byrd, Peter Storz, and Justin K. Messex
- Subjects
KRASG12D ,CCL9 ,PDAC initiation ,ROS ,MMP14 ,MMP3 ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC.
- Published
- 2024
- Full Text
- View/download PDF
3. Roles of differently polarized macrophages in the initiation and progressionof pancreatic cancer
- Author
-
Peter Storz
- Subjects
pancreatic cancer ,PDA ,macrophages ,inflammatory ,M1 ,M2 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
During development of pancreatic cancer macrophage-mediated inflammatory processes and the formation of cancerous lesions are tightly connected. Based on insight from mouse models we provide an overview on the functions of classically-activated pro-inflammatory and alternatively-activated anti-inflammatory macrophages in the initiation and progression of pancreatic cancer. We highlight their roles in earliest events of tumor initiation such as acinar-to-ductal metaplasia (ADM), organization of the fibrotic lesion microenvironment, and growth of low-grade (LG) lesions. We then discuss their roles as tumor-associated macrophages (TAM) in progression to high-grade (HG) lesions with a cancerous invasive phenotype and an immunosuppressive microenvironment. Another focus is on how targeting these macrophage populations can affect immunosuppression, fibrosis and responses to chemotherapy, and eventually how this knowledge could be used for novel therapy approaches for patients with pancreatic ductal adenocarcinoma (PDA).
- Published
- 2023
- Full Text
- View/download PDF
4. Inflammatory and alternatively activated macrophages independently induce metaplasia but cooperatively drive pancreatic precancerous lesion growth
- Author
-
Geou-Yarh Liou, Alicia K. Fleming Martinez, Heike R. Döppler, Ligia I. Bastea, and Peter Storz
- Subjects
Cancer ,Cell biology ,Immunology ,Molecular biology ,Science - Abstract
Summary: 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.
- Published
- 2023
- Full Text
- View/download PDF
5. Ym1+ macrophages orchestrate fibrosis, lesion growth, and progression during development of murine pancreatic cancer
- Author
-
Alicia K. Fleming Martinez, Heike R. Döppler, Ligia I. Bastea, Brandy H. Edenfield, Geou-Yarh Liou, and Peter Storz
- Subjects
Microenvironment ,Immunology ,Cancer ,Science - Abstract
Summary: 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. TGFβ1 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 TGFβ1 contributes to a cadherin switch and drives structural collapse of lesions, indicating a potential progression step. Taken together, our data indicate TGFβ1 produced by Ym1+ AAM as a major driver of processes that initiate the development of pancreatic cancer.
- Published
- 2022
- Full Text
- View/download PDF
6. CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions
- Author
-
Veethika Pandey, Alicia Fleming-Martinez, Ligia Bastea, Heike R Doeppler, Jillian Eisenhauer, Tam Le, Brandy Edenfield, and Peter Storz
- Subjects
pancreatic cancer ,macrophages ,CXCL10 ,CXCR3 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The development of pancreatic cancer requires recruitment and activation of different macrophage populations. However, little is known about how macrophages are attracted to the pancreas after injury or an oncogenic event, and how they crosstalk with lesion cells or other cells of the lesion microenvironment. Here, we delineate the importance of CXCL10/CXCR3 signaling during the early phase of murine pancreatic cancer. We show that CXCL10 is produced by pancreatic precancerous lesion cells in response to IFNγ signaling and that inflammatory macrophages are recipients for this chemokine. CXCL10/CXCR3 signaling in macrophages mediates their chemoattraction to the pancreas, enhances their proliferation, and maintains their inflammatory identity. Blocking of CXCL10/CXCR3 signaling in vivo shifts macrophage populations to a tumor-promoting (Ym1+, Fizz+, Arg1+) phenotype, increases fibrosis, and mediates progression of lesions, highlighting the importance of this pathway in PDA development. This is reversed when CXCL10 is overexpressed in PanIN cells.
- Published
- 2021
- Full Text
- View/download PDF
7. Dysfunctional EGFR and oxidative stress-induced PKD1 signaling drive formation of DCLK1+ pancreatic stem cells
- Author
-
Alicia K. Fleming Martinez, Heike R. Döppler, Ligia I. Bastea, Brandy Edenfield, Tushar Patel, Michael Leitges, Geou-Yarh Liou, and Peter Storz
- Subjects
Cell Biology ,Stem Cell Research ,Cancer ,Science - Abstract
Summary: 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.
- Published
- 2021
- Full Text
- View/download PDF
8. Generation of Hydrogen Peroxide and Downstream Protein Kinase D1 Signaling Is a Common Feature of Inducers of Pancreatic Acinar-to-Ductal Metaplasia
- Author
-
Heike R. Döppler, Geou-Yarh Liou, and Peter Storz
- Subjects
hydrogen peroxide ,oxidative stress ,ADM ,acinar-to-ductal metaplasia ,pancreas ,PKD ,Therapeutics. Pharmacology ,RM1-950 - Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
9. The Presence of Interleukin-13 at Pancreatic ADM/PanIN Lesions Alters Macrophage Populations and Mediates Pancreatic Tumorigenesis
- Author
-
Geou-Yarh Liou, Ligia Bastea, Alicia Fleming, Heike Döppler, Brandy H. Edenfield, David W. Dawson, Lizhi Zhang, Nabeel Bardeesy, and Peter Storz
- Subjects
pancreatic cancer ,PanIN ,metaplasia ,Tuft cells ,macrophages ,polarization ,IL-13 ,interleukin-13 ,CCL-2 ,IL-1ra ,Biology (General) ,QH301-705.5 - Abstract
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.
- Published
- 2017
- Full Text
- View/download PDF
10. Differential regulation of PKD isoforms in oxidative stress conditions through phosphorylation of a conserved Tyr in the P+1 loop
- Author
-
Mathias Cobbaut, Rita Derua, Heike Döppler, Hua Jane Lou, Sandy Vandoninck, Peter Storz, Benjamin E. Turk, Thomas Seufferlein, Etienne Waelkens, Veerle Janssens, and Johan Van Lint
- Subjects
Medicine ,Science - Abstract
Abstract 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.
- Published
- 2017
- Full Text
- View/download PDF
11. Mutant KRas-Induced Mitochondrial Oxidative Stress in Acinar Cells Upregulates EGFR Signaling to Drive Formation of Pancreatic Precancerous Lesions
- Author
-
Geou-Yarh Liou, Heike Döppler, Kathleen E. DelGiorno, Lizhi Zhang, Michael Leitges, Howard C. Crawford, Michael P. Murphy, and Peter Storz
- Subjects
pancreatic cancer ,PanIN ,Kras ,oxidative stress ,mitochondria ,growth factor signaling ,Biology (General) ,QH301-705.5 - Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
12. Vasodilator-Stimulated Phosphoprotein Activity Is Required for Coxiella burnetii Growth in Human Macrophages.
- Author
-
Punsiri M Colonne, Caylin G Winchell, Joseph G Graham, Frances I Onyilagha, Laura J MacDonald, Heike R Doeppler, Peter Storz, Richard C Kurten, Paul A Beare, Robert A Heinzen, and Daniel E Voth
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Coxiella burnetii is an intracellular bacterial pathogen that causes human Q fever, an acute flu-like illness that can progress to chronic endocarditis and liver and bone infections. Humans are typically infected by aerosol-mediated transmission, and C. burnetii initially targets alveolar macrophages wherein the pathogen replicates in a phagolysosome-like niche known as the parasitophorous vacuole (PV). C. burnetii manipulates host cAMP-dependent protein kinase (PKA) signaling to promote PV formation, cell survival, and bacterial replication. In this study, we identified the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) as a PKA substrate that is increasingly phosphorylated at S157 and S239 during C. burnetii infection. Avirulent and virulent C. burnetii triggered increased levels of phosphorylated VASP in macrophage-like THP-1 cells and primary human alveolar macrophages, and this event required the Cα subunit of PKA. VASP phosphorylation also required bacterial protein synthesis and secretion of effector proteins via a type IV secretion system, indicating the pathogen actively triggers prolonged VASP phosphorylation. Optimal PV formation and intracellular bacterial replication required VASP activity, as siRNA-mediated depletion of VASP reduced PV size and bacterial growth. Interestingly, ectopic expression of a phospho-mimetic VASP (S239E) mutant protein prevented optimal PV formation, whereas VASP (S157E) mutant expression had no effect. VASP (S239E) expression also prevented trafficking of bead-containing phagosomes to the PV, indicating proper VASP activity is critical for heterotypic fusion events that control PV expansion in macrophages. Finally, expression of dominant negative VASP (S157A) in C. burnetii-infected cells impaired PV formation, confirming importance of the protein for proper infection. This study provides the first evidence of VASP manipulation by an intravacuolar bacterial pathogen via activation of PKA in human macrophages.
- Published
- 2016
- Full Text
- View/download PDF
13. NFATc4 Regulates Sox9 Gene Expression in Acinar Cell Plasticity and Pancreatic Cancer Initiation
- Author
-
Elisabeth Hessmann, Jin-San Zhang, Nai-Ming Chen, Marie Hasselluhn, Geou-Yarh Liou, Peter Storz, Volker Ellenrieder, Daniel D. Billadeau, and Alexander Koenig
- Subjects
Internal medicine ,RC31-1245 - Abstract
Acinar transdifferentiation toward a duct-like phenotype constitutes the defining response of acinar cells to external stress signals and is considered to be the initial step in pancreatic carcinogenesis. Despite the requirement for oncogenic Kras in pancreatic cancer (PDAC) development, oncogenic Kras is not sufficient to drive pancreatic carcinogenesis beyond the level of premalignancy. Instead, secondary events, such as inflammation-induced signaling activation of the epidermal growth factor (EGFR) or induction of Sox9 expression, are required for tumor formation. Herein, we aimed to dissect the mechanism that links EGFR signaling to Sox9 gene expression during acinar-to-ductal metaplasia in pancreatic tissue adaptation and PDAC initiation. We show that the inflammatory transcription factor NFATc4 is highly induced and localizes in the nucleus in response to inflammation-induced EGFR signaling. Moreover, we demonstrate that NFATc4 drives acinar-to-ductal conversion and PDAC initiation through direct transcriptional induction of Sox9. Therefore, strategies designed to disrupt NFATc4 induction might be beneficial in the prevention or therapy of PDAC.
- Published
- 2016
- Full Text
- View/download PDF
14. Protein kinase d isoforms differentially modulate cofilin-driven directed cell migration.
- Author
-
Heike Döppler, Ligia I Bastea, Sahra Borges, Samantha J Spratley, Sarah E Pearce, and Peter Storz
- Subjects
Medicine ,Science - Abstract
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.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.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.
- Published
- 2014
- Full Text
- View/download PDF
15. Downregulation of TRAF2 mediates NIK-induced pancreatic cancer cell proliferation and tumorigenicity.
- Author
-
Heike Döppler, Geou-Yarh Liou, and Peter Storz
- Subjects
Medicine ,Science - Abstract
Increased levels of NF-κB are hallmarks of pancreatic ductal adenocarcinoma (PDAC) and both classical and alternative NF-κB activation pathways have been implicated.Here we show that activation of the alternative pathway is a source for the high basal NF-κB activity in PDAC cell lines. Increased activity of the p52/RelB NF-κB complex is mediated through stabilization and activation of NF-κB-inducing kinase (NIK). We identify proteasomal downregulation of TNF receptor-associated factor 2 (TRAF2) as a mechanism by which levels of active NIK are increased in PDAC cell lines. Such upregulation of NIK expression and activity levels relays to increased proliferation and anchorage-independent growth, but not migration or survival of PDAC cells.Rapid growth is one characteristic of pancreatic cancer. Our data indicates that the TRAF2/NIK/NF-κB2 pathway regulates PDAC cell tumorigenicity and could be a valuable target for therapy of this cancer.
- Published
- 2013
- Full Text
- View/download PDF
16. Protein kinase D1 maintains the epithelial phenotype by inducing a DNA-bound, inactive SNAI1 transcriptional repressor complex.
- Author
-
Ligia I Bastea, Heike Döppler, Bolanle Balogun, and Peter Storz
- Subjects
Medicine ,Science - Abstract
Protein kinase D1 is downregulated in its expression in invasive ductal carcinoma of the breast and in invasive breast cancer cells, but its functions in normal breast epithelial cells is largely unknown. The epithelial phenotype is maintained by cell-cell junctions formed by E-cadherin. In cancer cells loss of E-cadherin expression contributes to an invasive phenotype. This can be mediated by SNAI1, a transcriptional repressor for E-cadherin that contributes to epithelial-to-mesenchymal transition (EMT).Here we show that PKD1 in normal murine mammary gland (NMuMG) epithelial cells is constitutively-active in its basal state and prevents a transition to a mesenchymal phenotype. Investigation of the involved mechanism suggested that PKD1 regulates the expression of E-cadherin at the promoter level through direct phosphorylation of the transcriptional repressor SNAI1. PKD1-mediated phosphorylation of SNAI1 occurs in the nucleus and generates a nuclear, inactive DNA/SNAI1 complex that shows decreased interaction with its co-repressor Ajuba. Analysis of human tissue samples with a newly-generated phosphospecific antibody for PKD1-phosphorylated SNAI1 showed that regulation of SNAI1 through PKD1 occurs in vivo in normal breast ductal tissue and is decreased or lost in invasive ductal carcinoma.Our data describe a mechanism of how PKD1 maintains the breast epithelial phenotype. Moreover, they suggest, that the analysis of breast tissue for PKD-mediated phosphorylation of SNAI1 using our novel phosphoS11-SNAI1-specific antibody may allow predicting the invasive potential of breast cancer cells.
- Published
- 2012
- Full Text
- View/download PDF
17. Supplementary Figure S3 from Mutant KRAS–Induced Expression of ICAM-1 in Pancreatic Acinar Cells Causes Attraction of Macrophages to Expedite the Formation of Precancerous Lesions
- Author
-
Peter Storz, David W. Dawson, Lizhi Zhang, Brandy Edenfield, Brian Necela, Heike Döppler, and Geou-Yarh Liou
- Abstract
Supplementary Figure S3 shows analysis for microvascularization (A), alcian blue staining of tissue sections (B), and ICAM-1 expression in human samples (C).
- Published
- 2023
18. Data from Effective Targeting of Estrogen Receptor–Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101
- Author
-
Peter Storz, Xochiquetzal J. Geiger, Derek C. Radisky, E. Aubrey Thompson, Edith A. Perez, and Sahra Borges
- Abstract
Invasive ductal carcinomas (IDC) of the breast are associated with altered expression of hormone receptors (HR), amplification or overexpression of HER2, or a triple-negative phenotype. The most aggressive cases of IDC are characterized by a high proliferation rate, a great propensity to metastasize, and their ability to resist to standard chemotherapy, hormone therapy, or HER2-targeted therapy. Using progression tissue microarrays, we here demonstrate that the serine/threonine kinase protein kinase D3 (PKD3) is highly upregulated in estrogen receptor (ER)–negative (ER−) tumors. We identify direct binding of the ER to the PRKD3 gene promoter as a mechanism of inhibition of PKD3 expression. Loss of ER results in upregulation of PKD3, leading to all hallmarks of aggressive IDC, including increased cell proliferation, migration, and invasion. This identifies ER− breast cancers as ideal for treatment with the PKD inhibitor CRT0066101. We show that similar to a knockdown of PKD3, treatment with this inhibitor targets all tumorigenic processes in vitro and decreases growth of primary tumors and metastasis in vivo. Our data strongly support the development of PKD inhibitors for clinical use for ER− breast cancers, including the triple-negative phenotype. Mol Cancer Ther; 14(6); 1306–16. ©2015 AACR.
- Published
- 2023
19. Supplementary Methods from Mutant KRAS–Induced Expression of ICAM-1 in Pancreatic Acinar Cells Causes Attraction of Macrophages to Expedite the Formation of Precancerous Lesions
- Author
-
Peter Storz, David W. Dawson, Lizhi Zhang, Brandy Edenfield, Brian Necela, Heike Döppler, and Geou-Yarh Liou
- Abstract
Supplementary Methods
- Published
- 2023
20. Supplementary Tables 1 through 3 from Effective Targeting of Estrogen Receptor–Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101
- Author
-
Peter Storz, Xochiquetzal J. Geiger, Derek C. Radisky, E. Aubrey Thompson, Edith A. Perez, and Sahra Borges
- Abstract
Supplemental Table S1 shows differential expression of PRKD3 in breast cancer (triple negative vs triple positive); Supplemental Table S2 shows the differential expression of PRKD3 in breast cancer by ER status; and Supplemental Table S3 shows the level of PRKD1, PRKD2 and PRKD3 expression in invasive breast cancer cell lines according to their hormone receptor status. The file also contains the references for Tables S1 and S2.
- Published
- 2023
21. Supplementary Figures 1 through 9 from Effective Targeting of Estrogen Receptor–Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101
- Author
-
Peter Storz, Xochiquetzal J. Geiger, Derek C. Radisky, E. Aubrey Thompson, Edith A. Perez, and Sahra Borges
- Abstract
Supplemental Figure S1 shows the differential expression of PKD3 in invasive ductal carcinoma (IDC) by HER2 status; Supplemental Figure S2 shows the status of PKD1, PKD2 and PKD3 expression in invasive breast cancer cell lines; Supplemental Figure S3 depicts the effect of PKD3 knockdown on MDA-MB-231 cell proliferation; Supplemental Figure S4 depicts the effect of PKD3 knockdown on MDA-MB-231 cell migration; Supplemental Figure S5 shows the effect of PKD3 knockdown on tumor growth (A-C)and local invasion (D); Supplemental Figure S6 depicts the effect of CRT0066101 on MDA-MB-231 cell proliferation; Supplemental Figure S7 depicts the effect of CRT0066101 on MDA-MB-231 cell migration; Supplemental Figure S8 shows the impact of PKD2 and PKD3 knockdown alone or in combination on MDA-MB-231 cell migration; and Supplemental Figure S9 depicts the effect of CRT0066101 on tumor growth over time in orthotopic xenografts.
- Published
- 2023
22. Data from Mutant KRAS–Induced Expression of ICAM-1 in Pancreatic Acinar Cells Causes Attraction of Macrophages to Expedite the Formation of Precancerous Lesions
- Author
-
Peter Storz, David W. Dawson, Lizhi Zhang, Brandy Edenfield, Brian Necela, Heike Döppler, and Geou-Yarh Liou
- Abstract
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 KRASG12D induces the expression of intercellular adhesion molecule-1 (ICAM-1), which serves as chemoattractant for macrophages. Infiltrating macrophages amplify the formation of KRASG12D-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. Cancer Discov; 5(1); 52–63. ©2014 AACR.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2023
23. Data from Pomalidomide Alters Pancreatic Macrophage Populations to Generate an Immune-Responsive Environment at Precancerous and Cancerous Lesions
- Author
-
Peter Storz, Han W. Tun, John A. Copland, Brandy Edenfield, Yushi Qiu, Zhimin Li, Heike Doeppler, Christina A. von Roemeling, Alicia K. Fleming, Veethika Pandey, Geou-Yarh Liou, and Ligia I. Bastea
- Abstract
During development of pancreatic cancer, alternatively activated macrophages contribute to fibrogenesis, pancreatic intraepithelial neoplasia (PanIN) lesion growth, and generation of an immunosuppressive environment. Here, we show that the immunomodulatory agent pomalidomide depletes pancreatic lesion areas of alternatively activated macrophage populations. Pomalidomide treatment resulted in downregulation of interferon regulatory factor 4, a transcription factor for M2 macrophage polarization. Pomalidomide-induced absence of alternatively activated macrophages led to a decrease in fibrosis at PanIN lesions and in syngeneic tumors; this was due to generation of an inflammatory, immune-responsive environment with increased expression of IL1α and presence of activated (IFNγ-positive) CD4+ and CD8+ T-cell populations. Our results indicate that pomalidomide could be used to decrease fibrogenesis in pancreatic cancer and may be ideal as a combination treatment with chemotherapeutic drugs or other immunotherapies.Significance:These findings reveal new insights into how macrophage populations within the pancreatic cancer microenvironment can be modulated, providing the means to turn the microenvironment from immunosuppressive to immune-responsive.
- Published
- 2023
24. Supplementary Table and Supplementary Figures S1-S7 from Pomalidomide Alters Pancreatic Macrophage Populations to Generate an Immune-Responsive Environment at Precancerous and Cancerous Lesions
- Author
-
Peter Storz, Han W. Tun, John A. Copland, Brandy Edenfield, Yushi Qiu, Zhimin Li, Heike Doeppler, Christina A. von Roemeling, Alicia K. Fleming, Veethika Pandey, Geou-Yarh Liou, and Ligia I. Bastea
- Abstract
Supplementary Table 1: Table with antibodies and dilutions used Supplementary Figure S1: Treatment scheme and analyses of stroma Supplementary Figure S2: Additional analyses of cell death and cell proliferation Supplementary Figure S3: Presence of Ym1+ macrophage and IL-13 in PanIN areas Supplementary Figure S4: Additional data on the role of IRF4 in macrophage polarization Supplementary Figure S5: Presence of macrophage-secreted factors and T cells Supplementary Figure S6: Treatment scheme and analyses of presence of immune cells Supplementary Figure S7: Blood counts and coagulation profile
- Published
- 2023
25. Elevating microglia TREM2 reduces amyloid seeding and suppresses disease-associated microglia
- Author
-
Na Zhao, Wenhui Qiao, Fuyao Li, Yingxue Ren, Jiaying Zheng, Yuka A. Martens, Xusheng Wang, Ling Li, Chia-Chen Liu, Kai Chen, Yiyang Zhu, Tadafumi C. Ikezu, Zonghua Li, Axel D. Meneses, Yunjung Jin, Joshua A. Knight, Yixing Chen, Ligia Bastea, Cynthia Linares, Berkiye Sonustun, Lucy Job, Madeleine L. Smith, Manling Xie, Yong U. Liu, Anthony D. Umpierre, Koichiro Haruwaka, Zachary S. Quicksall, Peter Storz, Yan W. Asmann, Long-Jun Wu, and Guojun Bu
- Subjects
Amyloid ,Mice ,Membrane Glycoproteins ,Alzheimer Disease ,Immunology ,Animals ,Brain ,Humans ,Immunology and Allergy ,Amyloidosis ,Microglia ,Receptors, Immunologic - Abstract
TREM2 is exclusively expressed by microglia in the brain and is strongly linked to the risk for Alzheimer’s disease (AD). As microglial responses modulated by TREM2 are central to AD pathogenesis, enhancing TREM2 signaling has been explored as an AD therapeutic strategy. However, the effective therapeutic window targeting TREM2 is unclear. Here, by using microglia-specific inducible mouse models overexpressing human wild-type TREM2 (TREM2-WT) or R47H risk variant (TREM2-R47H), we show that TREM2-WT expression reduces amyloid deposition and neuritic dystrophy only during the early amyloid seeding stage, whereas TREM2-R47H exacerbates amyloid burden during the middle amyloid rapid growth stage. Single-cell RNA sequencing reveals suppressed disease-associated microglia (DAM) signature and reduced DAM population upon TREM2-WT expression in the early stage, whereas upregulated antigen presentation pathway is detected with TREM2-R47H expression in the middle stage. Together, our findings highlight the dynamic effects of TREM2 in modulating AD pathogenesis and emphasize the beneficial effect of enhancing TREM2 function in the early stage of AD development.
- Published
- 2022
26. Peripheral apoE4 enhances Alzheimer’s pathology and impairs cognition by compromising cerebrovascular function
- Author
-
Chia-Chen Liu, Jing Zhao, Yuan Fu, Yasuteru Inoue, Yingxue Ren, Yuanxin Chen, Sydney V. Doss, Francis Shue, Suren Jeevaratnam, Ligia Bastea, Na Wang, Yuka A. Martens, Wenhui Qiao, Minghui Wang, Na Zhao, Lin Jia, Yu Yamazaki, Akari Yamazaki, Cassandra L. Rosenberg, Zhen Wang, Dehui Kong, Zonghua Li, Lindsey A. Kuchenbecker, Zachary A. Trottier, Lindsey Felton, Justin Rogers, Zachary S. Quicksall, Cynthia Linares, Joshua Knight, Yixing Chen, Aishe Kurti, Takahisa Kanekiyo, John D. Fryer, Yan W. Asmann, Peter Storz, Xusheng Wang, Junmin Peng, Bin Zhang, Betty Y. S. Kim, and Guojun Bu
- Subjects
General Neuroscience ,Apolipoprotein E4 ,Induced Pluripotent Stem Cells ,Apolipoprotein E3 ,Brain ,Mice, Transgenic ,Article ,Mice ,Apolipoproteins E ,Cognition ,Alzheimer Disease ,Animals ,Humans ,Protein Isoforms - Abstract
The ε4 allele of the apolipoprotein E (APOE) gene, a genetic risk factor for Alzheimer's disease, is abundantly expressed in both the brain and periphery. Here, we present evidence that peripheral apoE isoforms, separated from those in the brain by the blood-brain barrier, differentially impact Alzheimer's disease pathogenesis and cognition. To evaluate the function of peripheral apoE, we developed conditional mouse models expressing human APOE3 or APOE4 in the liver with no detectable apoE in the brain. Liver-expressed apoE4 compromised synaptic plasticity and cognition by impairing cerebrovascular functions. Plasma proteome profiling revealed apoE isoform-dependent functional pathways highlighting cell adhesion, lipoprotein metabolism and complement activation. ApoE3 plasma from young mice improved cognition and reduced vessel-associated gliosis when transfused into aged mice, whereas apoE4 compromised the beneficial effects of young plasma. A human induced pluripotent stem cell-derived endothelial cell model recapitulated the plasma apoE isoform-specific effect on endothelial integrity, further supporting a vascular-related mechanism. Upon breeding with amyloid model mice, liver-expressed apoE4 exacerbated brain amyloid pathology, whereas apoE3 reduced it. Our findings demonstrate pathogenic effects of peripheral apoE4, providing a strong rationale for targeting peripheral apoE to treat Alzheimer's disease.
- Published
- 2022
27. Abstract 4779: MnSOD mimetic mitoquinone mesylate (MitoQ) increases DCLK1+pancreatic cancer stem cells
- Author
-
Jeffrey Mario Perera, Alicia K. Fleming Martinez, and Peter Storz
- Subjects
Cancer Research ,Oncology - Abstract
Mitochondrially-generated reactive oxygen species (mROS) have been shown to induce acinar-to-ductal metaplasia (ADM). ADM, in presence of an oncogenic KRAS mutation, is an initiating step for progression to pancreatic intraepithelial neoplasia (PanIN) and eventually pancreatic ductal adenocarcinoma (PDA). Here, we utilize primary murine cells as well as the p48Cre;LSL-KrasG12D (KC) mouse model to explore the use of the mitochondria-targeted mimetic of manganese superoxide dismutase MitoQ for targeting PDA initiation and progression. Previously published data by our laboratory has shown that treatment with MitoQ decreases the abundance of pancreatic low-grade lesions in KC mice. However, we here demonstrate that superoxide depletion by MitoQ at the same time increases formation of DCLK1+ cancer stem cells in such lesions. We here evaluate if the detoxification of mitochondrial superoxide results in a build-up of hydrogen peroxide, which is a major driver of the DCLK1+ cell type. We determine how altered expression of ROS detoxifying enzymes in different cell types contribute to PDA stem cell formation and PDA development. Eventually we test if the increase in DCLK1+ cancer stem cells can contribute to changes in the microenvironment and accelerated progression of abnormal tissue. Citation Format: Jeffrey Mario Perera, Alicia K. Fleming Martinez, Peter Storz. MnSOD mimetic mitoquinone mesylate (MitoQ) increases DCLK1+pancreatic cancer stem cells. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4779.
- Published
- 2023
28. Abstract 4775: Knockout of Sod2 accelerates KrasG12D-driven formation of pancreatic cancerous lesions
- Author
-
Alicia K. Fleming Martinez, Brandy H. Edenfield, Irene Esposito, and Peter Storz
- Subjects
Cancer Research ,Oncology - Abstract
Recent studies demonstrate that mitochondrially-generated reactive oxygen species (ROS) downstream of oncogenic KRAS drive acinar-to-ductal metaplasia (ADM), a key first step in the pancreatic ductal adenocarcinoma (PDA) progression model. MnSOD/SOD2 is a mitochondrial enzyme that converts superoxide into hydrogen peroxide. Previous studies have shown that SOD2 is biologically relevant to the study of pancreatic cancer. Some patients with a SOD2 polymorphism resulting in less active MnSOD at the mitochondrial matrix are at increased risk of developing pancreatic cancer. Additionally, MnSOD is lost in high-grade human PanIN, further suggesting a role in tumor development. To elucidate the role of MnSOD in pancreatic cancer initiation and progression, we crossed Sod2lox/lox mice into the p48Cre;LSL-KrasG12D (KC) mouse model. We found increased ROS, abundantly more abnormal tissue development and presence of flat lesions indicating accelerated early progression. However, Kaplan Meyer survival analysis of KC and KC;Sod2−/− mice did not indicate faster progression to cancer, suggesting that for later steps additional signaling is needed. In summary, we identify MnSOD as a key antioxidant molecule preventing initiation of PDA. Citation Format: Alicia K. Fleming Martinez, Brandy H. Edenfield, Irene Esposito, Peter Storz. Knockout of Sod2 accelerates KrasG12D-driven formation of pancreatic cancerous lesions. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4775.
- Published
- 2023
29. Carcinogenesis of Pancreatic Ductal Adenocarcinoma
- Author
-
Peter Storz and Howard C. Crawford
- Subjects
0301 basic medicine ,Time Factors ,Regulatory T cell ,education ,Pancreatic stellate cell ,Pancreatic Intraepithelial Neoplasia ,Tumor-associated macrophage ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Cell Lineage ,Genetic Predisposition to Disease ,Tumor microenvironment ,Hepatology ,Intraductal papillary mucinous neoplasm ,business.industry ,Carcinoma in situ ,Gastroenterology ,medicine.disease ,Pancreatic Neoplasms ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Mutation ,Disease Progression ,Cancer research ,030211 gastroenterology & hepatology ,Carcinogenesis ,business ,Carcinoma, Pancreatic Ductal - Abstract
Although the estimated time for development of pancreatic ductal adenocarcinoma (PDA) is more than 20 years, PDAs are usually detected at late, metastatic stages. PDAs develop from duct-like cells through a multistep carcinogenesis process, from low-grade dysplastic lesions to carcinoma in situ and eventually to metastatic disease. This process involves gradual acquisition of mutations in oncogenes and tumor suppressor genes, as well as changes in the pancreatic environment from a pro-inflammatory microenvironment that favors the development of early lesions, to a desmoplastic tumor microenvironment that is highly fibrotic and immune suppressive. This review discusses our current understanding of how PDA originates.
- Published
- 2020
30. AT-101 Enhances the Antitumor Activity of Lenalidomide in Patients with Multiple Myeloma
- Author
-
Sikander Ailawadhi, Ricardo D. Parrondo, Navnita Dutta, Bing Han, Gina Ciccio, Yesesri Cherukuri, Victoria R. Alegria, Betsy R. LaPlant, Vivek Roy, Taimur Sher, Brett Edwards, Stephanie Lanier, Alak Manna, Keisha Heslop, Thomas Caulfield, Emir Maldosevic, Peter Storz, Rami Manochakian, Yan Asmann, Asher A. Chanan-Khan, and Aneel Paulus
- Subjects
Cancer Research ,Oncology ,multiple myeloma ,Bcl-2 inhibition ,apoptosis - Abstract
Bcl-2 and Mcl-1 proteins play a role in multiple myeloma (MM) cell survival, for which targeted inhibitors are being developed. AT-101 is an oral drug, which disrupts Bcl-2 and Mcl-1 function, impedes mitochondrial bioenergetic processes and induces apoptosis in MM cells. When combined with lenalidomide and dexamethasone (Rd), AT-101 significantly reduced tumor burden in an in vivo xenograft model of MM. These data provided rationale for a phase I/II study to establish the effective dose of AT-101 in combination with Rd (ARd regimen) in relapsed/refractory MM. A total of 10 patients were enrolled, most with high-risk cytogenetics (80%) and prior stem cell transplant (70%). Three patients were lenalidomide-refractory, 2 were bortezomib-refractory and 3 were daratumumab-refractory. The ARd combination was well tolerated with most common grade 3/4 adverse events being cytopenia’s. The overall response rate was 40% and clinical benefit rate was 90%. The median progression free survival was 14.9 months (95% CI 7.1-NE). Patients responsive to ARd showed a decrease in Bcl-2:Bim or Mcl-1:Noxa protein complexes, increased CD8+ T and NK cells and depletion of T and B-regulatory cells. The ARd regimen demonstrated an acceptable safety profile and promising efficacy in patients with relapsed/refractory MM prompting further investigation in additional patients.
- Published
- 2023
31. Ym1
- Author
-
Alicia K, Fleming Martinez, Heike R, Döppler, Ligia I, Bastea, Brandy H, Edenfield, Geou-Yarh, Liou, and Peter, Storz
- Abstract
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
- Published
- 2021
32. CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions
- Author
-
Alicia Fleming-Martinez, Heike Doeppler, Veethika Pandey, Ligia I. Bastea, Brandy Edenfield, Tam Le, Peter Storz, and Jillian Eisenhauer
- Subjects
Male ,Chemokine ,Mouse ,pancreatic cancer ,CXCR3 ,Mice ,Fibrosis ,immune system diseases ,Tumor Microenvironment ,Macrophage ,Biology (General) ,Cells, Cultured ,Cancer Biology ,biology ,General Neuroscience ,CXCL10 ,hemic and immune systems ,General Medicine ,macrophages ,medicine.anatomical_structure ,Disease Progression ,Medicine ,Female ,medicine.symptom ,Pancreas ,Signal Transduction ,Research Article ,Human ,Receptors, CXCR3 ,QH301-705.5 ,Science ,General Biochemistry, Genetics and Molecular Biology ,Lesion ,Pancreatic cancer ,medicine ,Animals ,Inflammation ,General Immunology and Microbiology ,Cell Biology ,medicine.disease ,Chemokine CXCL10 ,Pancreatic Neoplasms ,Disease Models, Animal ,biology.protein ,Cancer research - Abstract
The development of pancreatic cancer requires recruitment and activation of different macrophage populations. However, little is known about how macrophages are attracted to the pancreas after injury or an oncogenic event, and how they crosstalk with lesion cells or other cells of the lesion microenvironment. Here, we delineate the importance of CXCL10/CXCR3 signaling during the early phase of murine pancreatic cancer. We show that CXCL10 is produced by pancreatic precancerous lesion cells in response to IFNγ signaling and that inflammatory macrophages are recipients for this chemokine. CXCL10/CXCR3 signaling in macrophages mediates their chemoattraction to the pancreas, enhances their proliferation, and maintains their inflammatory identity. Blocking of CXCL10/CXCR3 signaling in vivo shifts macrophage populations to a tumor-promoting (Ym1+, Fizz+, Arg1+) phenotype, increases fibrosis, and mediates progression of lesions, highlighting the importance of this pathway in PDA development. This is reversed when CXCL10 is overexpressed in PanIN cells.
- Published
- 2021
33. Author response: CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions
- Author
-
Veethika Pandey, Alicia Fleming-Martinez, Ligia Bastea, Heike R Doeppler, Jillian Eisenhauer, Tam Le, Brandy Edenfield, and Peter Storz
- Published
- 2021
34. Protein kinase D up-regulates transcription of VEGF receptor-2 in endothelial cells by suppressing nuclear localization of the transcription factor AP2β
- Author
-
Tao Shen, Ramcharan Singh Angom, Resham Bhattacharya, Fei Wang, Luke H. Hoeppner, Ying Wang, Shamit K. Dutta, Sushovan Guha, Enfeng Wang, Isobel A. Scarisbrick, Debabrata Mukhopadhyay, Peter Storz, and Heike Doeppler
- Subjects
0301 basic medicine ,Gene knockdown ,030102 biochemistry & molecular biology ,Chemistry ,Angiogenesis ,Kinase insert domain receptor ,Cell Biology ,Biochemistry ,Cell biology ,03 medical and health sciences ,Vascular endothelial growth factor A ,030104 developmental biology ,Vasculogenesis ,embryonic structures ,cardiovascular system ,Signal transduction ,Protein kinase A ,Molecular Biology ,Transcription factor - Abstract
Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer. In human umbilical vein endothelial cells (HUVECs), knockdown of protein kinase D-1 (PKD1) or PKD2 down-regulates VEGFR-2 expression and inhibits VEGF-induced cell proliferation and migration. However, how PKD regulates VEGF signaling is unclear. Previous bioinformatics analyses have identified binding sites for the transcription factor activating enhancer-binding protein 2 (AP2) in the VEGFR-2 promoter. Using ChIP analyses, here we found that PKD knockdown in HUVECs increases binding of AP2β to the VEGFR-2 promoter. Luciferase reporter assays with serial deletions of AP2-binding sites within the VEGFR-2 promoter revealed that its transcriptional activity negatively correlates with the number of these sites. Next we demonstrated that AP2β up-regulation decreases VEGFR-2 expression and that loss of AP2β enhances VEGFR-2 expression in HUVECs. In vivo experiments confirmed increased VEGFR-2 immunostaining in the spinal cord of AP2β knockout mouse embryos. Mechanistically, we observed that PKD phosphorylates AP2β at Ser258 and Ser277 and suppresses its nuclear accumulation. Inhibition of PKD activity with a pan-PKD inhibitor increased AP2β nuclear localization, and overexpression of both WT and constitutively active PKD1 or PKD2 reduced AP2β nuclear localization through a Ser258- and Ser277-dependent mechanism. Furthermore, substitution of Ser277 in AP2β increased its binding to the VEGFR-2 promoter. Our findings uncover evidence of a molecular pathway that regulates VEGFR-2 expression, insights that may shed light on the etiology of diseases associated with aberrant VEGF/VEGFR signaling.
- Published
- 2019
35. Dysfunctional EGFR and oxidative stress-induced PKD1 signaling drive formation of DCLK1+ pancreatic stem cells
- Author
-
Brandy Edenfield, Michael Leitges, Peter Storz, Alicia K. Fleming Martinez, Geou Yarh Liou, Heike Döppler, Ligia I. Bastea, and Tushar Patel
- Subjects
0301 basic medicine ,endocrine system diseases ,Population ,02 engineering and technology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Pancreatic cancer ,medicine ,Epidermal growth factor receptor ,education ,lcsh:Science ,Cancer ,education.field_of_study ,Multidisciplinary ,biology ,Chemistry ,Kinase ,Cell Biology ,021001 nanoscience & nanotechnology ,medicine.disease ,Stem Cell Research ,030104 developmental biology ,Cancer research ,biology.protein ,lcsh:Q ,Erlotinib ,Protein kinase D1 ,KRAS ,Stem cell ,0210 nano-technology ,medicine.drug - Abstract
Summary 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., Graphical Abstract For a Figure360 author presentation of this figure, see https://doi.org/10.1016/j.isci.2020.102019., Highlights • DCLK1+ PanIN cells have abrogated EGFR signaling and increased oxidative stress • Inhibition of EGFR signaling increases DCLK1+ cells, oxidative stress, and PKD1 • PKD1 contributes to the abundance of DCLK1+ stem cells in PanIN lesions • Stem cell markers, including OCT4 and CD133, are increased by PKD1, Cell Biology; Stem Cell Research; Cancer
- Published
- 2021
36. Early detection and imaging strategies to reveal and target developing pancreatic cancer
- Author
-
Peter Storz, Howard C. Crawford, and Michael B. Wallace
- Subjects
0301 basic medicine ,Diagnostic Imaging ,congenital, hereditary, and neonatal diseases and abnormalities ,Pancreatic ductal adenocarcinoma ,health care facilities, manpower, and services ,education ,Pancreatic Intraepithelial Neoplasia ,Early detection ,Adenocarcinoma ,Article ,03 medical and health sciences ,0302 clinical medicine ,health services administration ,Pancreatic cancer ,Medicine ,Humans ,Pharmacology (medical) ,Early Detection of Cancer ,business.industry ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,business ,Carcinoma, Pancreatic Ductal - Abstract
Here, we summarize the current strategies for early detection of pancreatic ductal adenocarcinoma (PDA) including traditional and novel detection methods. PDA remains the deadliest of the common ca...
- Published
- 2020
37. Pomalidomide-induced changes in the pancreatic tumor microenvironment and potential for therapy
- Author
-
Peter Storz
- Subjects
0303 health sciences ,Cancer Research ,business.industry ,Macrophages ,Pomalidomide ,medicine.disease ,Pancreatic Cancer ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Pancreatic tumor ,030220 oncology & carcinogenesis ,Pancreatic cancer ,Research Perspective ,medicine ,Cancer research ,business ,030304 developmental biology ,medicine.drug - Published
- 2019
38. Protein kinase C isoforms in the normal pancreas and in pancreatic disease
- Author
-
Peter Storz and Alicia K. Fleming
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Pancreatic disease ,Biology ,Article ,03 medical and health sciences ,Insulin resistance ,Pancreatic cancer ,Internal medicine ,Diabetes Mellitus ,medicine ,SLC26A6 ,Humans ,Protein Isoforms ,Pancreas ,Protein Kinase C ,Protein kinase C ,Cell Biology ,medicine.disease ,KEAP1 ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Pancreatitis ,biology.protein ,Insulin Resistance - Abstract
Protein Kinase C isoforms have been implicated in regulating multiple processes within the healthy pancreas. Moreover, their dysregulation contributes to all aspects of pancreatic disease. In this review, with a focus on acinar, ductal, and islet cells, we highlight the roles and contributions of the different PKC isoforms to normal pancreas function. We also discuss the contribution of PKC enzymes to pancreatic diseases, including insulin resistance and diabetes mellitus, as well as pancreatitis and the development and progression of pancreatic cancer.
- Published
- 2017
39. Glycogen synthase kinase-3β ablation limits pancreatitis-induced acinar-to-ductal metaplasia
- Author
-
Daniel Schmitt, Geou Yarh Liou, Daniel D. Billadeau, Li Ding, Jin San Zhang, and Peter Storz
- Subjects
0301 basic medicine ,endocrine system diseases ,Transdifferentiation ,Pancreatic Intraepithelial Neoplasia ,P70-S6 Kinase 1 ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,GSK-3 ,Metaplasia ,Pancreatic cancer ,Cancer research ,medicine ,biology.protein ,medicine.symptom ,Pancreas ,Glycogen synthase - Abstract
Acinar-to-ductal metaplasia (ADM) is a reversible epithelial transdifferentiation process that occurs in the pancreas in response to acute inflammation. ADM can rapidly progress towards pre-malignant pancreatic intraepithelial neoplasia (PanIN) lesions in the presence of mutant KRas and ultimately pancreatic adenocarcinoma (PDAC). In the present work, we elucidate the role and related mechanism of glycogen synthase kinase-3beta (GSK-3β) in ADM development using in vitro 3D cultures and genetically engineered mouse models. We show that GSK-3β promotes TGF-α-induced ADM in 3D cultured primary acinar cells, whereas deletion of GSK-3β attenuates caerulein-induced ADM formation and PanIN progression in KrasG12D transgenic mice. Furthermore, we demonstrate that GSK-3β ablation influences ADM formation and PanIN progression by suppressing oncogenic KRas-driven cell proliferation. Mechanistically, we show that GSK-3β regulates proliferation by increasing the activation of S6 kinase. Taken together, these results indicate that GSK-3β participates in early pancreatitis-induced ADM and thus could be a target for the treatment of chronic pancreatitis and the prevention of PDAC progression. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2017
40. A Summary of the 2016 James W. Freston Conference of the American Gastroenterological Association: Intestinal Metaplasia in the Esophagus and Stomach
- Author
-
David H. Wang, Anil K. Rustgi, Richard M. Peek, James G. Fox, Peter Storz, Jason C. Mills, Nicholas A. Wright, Juanita L. Merchant, Pauline K. Lund, Yoku Hayakawa, Parakrama Chandrasoma, Stuart J. Spechler, Thai H. Pham, Ramesh A. Shivdasani, Robert D. Odze, Ernst J. Kuipers, Nicholas J. Shaheen, James R. Goldenring, Robert M. Genta, Andrea Todisco, Frank McKeon, Rhonda F. Souza, Jianwen Que, Timothy C. Wang, and Gastroenterology & Hepatology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Hepatology ,Acinar to ductal metaplasia ,Extramural ,Stomach ,Gastroenterology ,Intestinal metaplasia ,Cell lineage ,Biology ,medicine.disease ,Article ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Internal medicine ,Metaplasia ,medicine ,GERD ,medicine.symptom ,Esophagus - Published
- 2017
41. The Presence of Interleukin-13 at Pancreatic ADM/PanIN Lesions Alters Macrophage Populations and Mediates Pancreatic Tumorigenesis
- Author
-
Ligia I. Bastea, David W. Dawson, Lizhi Zhang, Heike Döppler, Nabeel Bardeesy, Geou Yarh Liou, Peter Storz, Brandy Edenfield, and Alicia K. Fleming
- Subjects
0301 basic medicine ,Carcinogenesis ,pancreatic cancer ,Pancreatic Intraepithelial Neoplasia ,Tuft cells ,interleukin-13 ,CCL2 ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,IL-1ra ,0302 clinical medicine ,Neutralization Tests ,Fibrosis ,Cell Line, Tumor ,CCL-2 ,Metaplasia ,Pancreatic cancer ,metaplasia ,medicine ,Animals ,lcsh:QH301-705.5 ,Cell Proliferation ,Inflammation ,polarization ,Innate immune system ,Pancreatic Ducts ,Cell Polarity ,medicine.disease ,macrophages ,3. Good health ,Pancreatic Neoplasms ,030104 developmental biology ,lcsh:Biology (General) ,IL-13 ,030220 oncology & carcinogenesis ,Interleukin 13 ,Immunology ,Cancer research ,PanIN ,KRAS ,medicine.symptom ,Carcinoma in Situ - Abstract
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.
- Published
- 2017
42. Protein kinase D1: gatekeeper of the epithelial phenotype and key regulator of cancer metastasis?
- Author
-
Peter Storz
- Subjects
0301 basic medicine ,Cancer Research ,Regulator ,Cancer metastasis ,Biology ,Phenotype ,3. Good health ,Epithelial phenotype ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Protein kinase D1 - Abstract
Protein kinase D1: gatekeeper of the epithelial phenotype and key regulator of cancer metastasis?
- Published
- 2018
43. Sangivamycin and its derivatives inhibit Haspin-Histone H3-survivin signaling and induce pancreatic cancer cell death
- Author
-
Elizabeth M. Reid, Peter Storz, Ligia I. Bastea, Heike Döppler, and Laeticia Hollant
- Subjects
0301 basic medicine ,Cell biology ,Survivin ,lcsh:Medicine ,Apoptosis ,Protein Serine-Threonine Kinases ,Biochemistry ,Article ,Histones ,03 medical and health sciences ,Histone H3 ,Mice ,0302 clinical medicine ,Pancreatic cancer ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Tumor Microenvironment ,Animals ,Humans ,Phosphorylation ,lcsh:Science ,Mitosis ,Cell Proliferation ,Cancer ,Tumor microenvironment ,Multidisciplinary ,Antibiotics, Antineoplastic ,Kinase ,Chemistry ,Cell growth ,lcsh:R ,Intracellular Signaling Peptides and Proteins ,medicine.disease ,Prognosis ,Pyrimidine Nucleosides ,Xenograft Model Antitumor Assays ,3. Good health ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:Q ,Protein Processing, Post-Translational ,Signal Transduction - Abstract
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.
- Published
- 2019
44. Alzheimer's Risk Factors Age, APOE Genotype, and Sex Drive Distinct Molecular Pathways
- Author
-
Yingxue Ren, Zonghua Li, Alexandra Kueider-Paisley, Yu Yamazaki, Olivia N. Attrebi, Rima Kaddurah-Daouk, Fuyao Li, Yuka A. Martens, Takahisa Kanekiyo, Akari Yamazaki, Lucy Job, Nilufer Ertekin-Taner, Matthias Arnold, Ligia I. Bastea, Tomonori Aikawa, Siamak MahmoudianDehkordi, Kai Chen, Lisa St. John-Williams, Na Zhao, Francis Shue, Berkiye Sonoustoun, Axel D. Meneses, Guojun Bu, Wenhui Qiao, J. Will Thompson, Peter Storz, Lindsey M. Felton, Jiaying Zheng, Hiroshi Oue, Masaya Tachibana, Chia Chen Liu, and Yan W. Asmann
- Subjects
0301 basic medicine ,Apolipoprotein E ,Male ,Aging ,Genotype ,Apolipoprotein E2 ,Apolipoprotein E4 ,Apolipoprotein E3 ,Gene Expression ,Mice, Transgenic ,Disease ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Apolipoproteins E ,Sex Factors ,Downregulation and upregulation ,Alzheimer Disease ,Risk Factors ,Metabolome ,Animals ,Humans ,Gene Regulatory Networks ,Receptors, Immunologic ,Gene ,Serpins ,Adaptor Proteins, Signal Transducing ,Genetics ,Membrane Glycoproteins ,TREM2 ,General Neuroscience ,Gene Expression Profiling ,Age Factors ,Brain ,Membrane Proteins ,Protective Factors ,030104 developmental biology ,Unfolded Protein Response ,Female ,030217 neurology & neurosurgery - Abstract
Evidence suggests interplay among the three major risk factors for Alzheimer's disease (AD): age, APOE genotype, and sex. Here, we present comprehensive datasets and analyses of brain transcriptomes and blood metabolomes from human apoE2-, apoE3-, and apoE4-targeted replacement mice across young, middle, and old ages with both sexes. We found that age had the greatest impact on brain transcriptomes highlighted by an immune module led by Trem2 and Tyrobp, whereas APOE4 was associated with upregulation of multiple Serpina3 genes. Importantly, these networks and gene expression changes were mostly conserved in human brains. Finally, we observed a significant interaction between age, APOE genotype, and sex on unfolded protein response pathway. In the periphery, APOE2 drove distinct blood metabolome profile highlighted by the upregulation of lipid metabolites. Our work identifies unique and interactive molecular pathways underlying AD risk factors providing valuable resources for discovery and validation research in model systems and humans.
- Published
- 2019
45. Protein kinase D up-regulates transcription of
- Author
-
Ying, Wang, Luke H, Hoeppner, Ramcharan Singh, Angom, Enfeng, Wang, Shamit, Dutta, Heike R, Doeppler, Fei, Wang, Tao, Shen, Isobel A, Scarisbrick, Sushovan, Guha, Peter, Storz, Resham, Bhattacharya, and Debabrata, Mukhopadhyay
- Subjects
Cell Nucleus ,Transcription, Genetic ,Neovascularization, Physiologic ,Cell Biology ,Vascular Endothelial Growth Factor Receptor-2 ,Up-Regulation ,HEK293 Cells ,Transcription Factor AP-2 ,Cell Movement ,Gene Knockdown Techniques ,embryonic structures ,cardiovascular system ,Human Umbilical Vein Endothelial Cells ,Serine ,Humans ,Promoter Regions, Genetic ,Protein Kinase C ,Cell Proliferation ,Protein Binding - Abstract
Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer. In human umbilical vein endothelial cells (HUVECs), knockdown of protein kinase D-1 (PKD1) or PKD2 down-regulates VEGFR-2 expression and inhibits VEGF-induced cell proliferation and migration. However, how PKD regulates VEGF signaling is unclear. Previous bioinformatics analyses have identified binding sites for the transcription factor activating enhancer-binding protein 2 (AP2) in the VEGFR-2 promoter. Using ChIP analyses, here we found that PKD knockdown in HUVECs increases binding of AP2β to the VEGFR-2 promoter. Luciferase reporter assays with serial deletions of AP2-binding sites within the VEGFR-2 promoter revealed that its transcriptional activity negatively correlates with the number of these sites. Next we demonstrated that AP2β up-regulation decreases VEGFR-2 expression and that loss of AP2β enhances VEGFR-2 expression in HUVECs. In vivo experiments confirmed increased VEGFR-2 immunostaining in the spinal cord of AP2β knockout mouse embryos. Mechanistically, we observed that PKD phosphorylates AP2β at Ser(258) and Ser(277) and suppresses its nuclear accumulation. Inhibition of PKD activity with a pan-PKD inhibitor increased AP2β nuclear localization, and overexpression of both WT and constitutively active PKD1 or PKD2 reduced AP2β nuclear localization through a Ser(258)- and Ser(277)-dependent mechanism. Furthermore, substitution of Ser(277) in AP2β increased its binding to the VEGFR-2 promoter. Our findings uncover evidence of a molecular pathway that regulates VEGFR-2 expression, insights that may shed light on the etiology of diseases associated with aberrant VEGF/VEGFR signaling.
- Published
- 2019
46. The GEF-H1/PKD3 signaling pathway promotes the maintenance of triple-negative breast cancer stem cells
- Author
-
Cristiana Lungu, Angelika Hausser, Peter Storz, Raluca Tamas, Monilola A. Olayioye, Philipp Rathert, Hannah Berreth, and Wolfgang S. Lieb
- Subjects
Cancer Research ,Paclitaxel ,Cell Survival ,Apoptosis ,Triple Negative Breast Neoplasms ,Tumor initiation ,Biology ,Article ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Cancer stem cell ,Cell Movement ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Triple-negative breast cancer ,Protein Kinase C ,Cell Proliferation ,Cell growth ,Drug Synergism ,medicine.disease ,Xenograft Model Antitumor Assays ,Pyrimidines ,Oncology ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Cancer research ,Neoplastic Stem Cells ,Female ,Stem cell ,Signal transduction ,Rho Guanine Nucleotide Exchange Factors ,Signal Transduction - Abstract
Protein kinase D3 (PKD3) is upregulated in triple-negative breast cancer (TNBC) and associated with cell proliferation and metastasis development but its precise pro-oncogenic function is unknown. Here we show that PKD3 is required for the maintenance of the TNBC stem cell population. The depletion of PKD3 in MDA-MB-231 cells reduced the cancer stem cell frequency in vitro and tumor initiation potential in vivo. We further provide evidence that the RhoGEF GEF-H1 is upstream of PKD3 activation in TNBC stem cells. Most importantly, pharmacological PKD inhibition in combination with paclitaxel synergistically decreased oncosphere and colony formation efficiency in vitro and tumor recurrence in vivo. Based on our results we propose that targeting the GEF-H1/PKD3 signaling pathway in combination with chemotherapy might provide an effective therapeutic option for TNBC.
- Published
- 2019
47. Targeting the tumor microenvironment in pancreatic ductal adenocarcinoma
- Author
-
Veethika Pandey and Peter Storz
- Subjects
0301 basic medicine ,Antineoplastic Agents ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Stroma ,Pancreatic tumor ,Pancreatic cancer ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Tumor microenvironment ,business.industry ,Cancer ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Myeloid-derived Suppressor Cell ,Cancer research ,Immunotherapy ,business ,Carcinoma, Pancreatic Ductal - Abstract
Introduction: The dismally slow improvement in patient survival over the years for pancreatic cancer patients is mainly due to two factors: the late diagnosis, at which point the disease is spread to distant organs; and the fact that tumor cells are surrounded by a dense, highly immunosuppressive microenvironment. The tumor microenvironment not only shields pancreatic cancer cells from chemotherapy but also leaves it unsusceptible to various immunotherapeutic strategies that have been proven successful in other types of cancer. Areas covered: This review highlights the main components of the pancreatic tumor microenvironment, how they cross-talk with each other to generate stroma and promote tumor growth. Additionally, we discuss the most promising treatment targets in the microenvironment whose modulation can be robustly tested in combination with standard of care chemotherapy. Currently, active clinical trials for pancreatic cancer involving components of the microenvironment are also listed. Expert opinion: Although immunotherapeutic approaches involving checkpoint inhibition are being pursued enthusiastically, there is still more work to be done with several other emerging immune targets that could provide therapeutic benefit.
- Published
- 2019
48. Mucin-1 is required for Coxsackie Virus B3-induced inflammation in pancreatitis
- Author
-
Jesse L. Cox, Nora M. Chapman, Jeffrey D. Price, Haitao Wen, Kelly A. O'Connell, Peter Storz, Whitney L. Reid, Michael A. Hollingsworth, James A. Grunkemeyer, Dahn L. Clemens, Xiang Liu, and Sarah P. Thayer
- Subjects
0301 basic medicine ,CCR2 ,Receptors, CCR2 ,lcsh:Medicine ,Coxsackievirus Infections ,Inflammation ,Article ,03 medical and health sciences ,Chemokine receptor ,Mice ,0302 clinical medicine ,Pancreatic cancer ,medicine ,Animals ,lcsh:Science ,Pancreatic disease ,Mice, Knockout ,Multidisciplinary ,Chemistry ,lcsh:R ,Mucin ,Mucin-1 ,Gastroenterology ,medicine.disease ,3. Good health ,Enterovirus B, Human ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Pancreatitis ,Cancer research ,lcsh:Q ,Bone marrow ,medicine.symptom ,Pancreas ,030217 neurology & neurosurgery - Abstract
The Muc-1 oncoprotein is a tumor-associated mucin often overexpressed in pancreatic cancer. We report that knockout of Muc-1 reduced the degree of pancreatic inflammation that resulted from infection with Coxsackievirus B3 (CVB3) in a mouse model. CVB3-infected Muc-1-deficient (Muc-1KO) mice had significantly reduced infiltration of macrophages into the murine pancreas. We found that Muc-1 signaling through NF-κB increased expression of ICAM-1, a pro-inflammatory mediator that recruits macrophages. Further investigation revealed that bone marrow derived macrophages (BMDM) from the Muc-1KO mice exhibited defective migration properties, in part due to low expression of the C-C motif chemokine receptor (CCR2) and the integrin Very Late Antigen 4 (VLA-4). The results presented here provide novel insight into the role of Muc-1 in regulating the inflammatory response and the cellular microenvironment in pancreatitis.
- Published
- 2019
49. Mimicking and Manipulating Pancreatic Acinar-to-Ductal Metaplasia in 3-dimensional Cell Culture
- Author
-
Peter Storz and Alicia K. Fleming Martinez
- Subjects
0301 basic medicine ,Ductal cells ,General Chemical Engineering ,Cell Culture Techniques ,Acinar Cells ,Article ,General Biochemistry, Genetics and Molecular Biology ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Metaplasia ,Pancreatic cancer ,medicine ,Acinar cell ,Humans ,Pancreas ,Basement membrane ,Gene knockdown ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Pancreatic Ducts ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,medicine.symptom - Abstract
The differentiation of acinar cells to ductal cells during pancreatitis and in the early development of pancreatic cancer is a key process that requires further study. To understand the mechanisms regulating acinar-to-ductal metaplasia (ADM), ex vivo 3D culture and differentiation of primary acinar cells to ductal cells offers many advantages over other systems. With the technique herein, modulation of protein expression is simple and quick, requiring only one day to isolate, stimulate or virally infect, and begin culturing primary acinar cells to investigate the ADM process. In contrast to using basement membrane matrix, the seeding of acinar cell clusters in collagen I extracellular matrix, allows acinar cells to retain their acinar identity before manipulation. This is vital when testing the contribution of various components to the induction of ADM. Not only are the effects of cytokines or other ectopically administered factors testable through this technique, but the contribution of common mutations, increased protein expression, or knockdown of protein expression is testable via viral infection of primary acinar cells, using adenoviral or lentiviral vectors. Moreover, cells can be re-isolated from collagen or basement membrane matrix at the endpoint and analyzed for protein expression.
- Published
- 2019
50. Targeting reactive oxygen species in development and progression of pancreatic cancer
- Author
-
Nisha Durand and Peter Storz
- Subjects
0301 basic medicine ,Senescence ,education ,Adenocarcinoma ,Biology ,Mitochondrion ,medicine.disease_cause ,Antioxidants ,Article ,Proto-Oncogene Proteins p21(ras) ,Gene product ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,medicine ,Animals ,Humans ,Pharmacology (medical) ,chemistry.chemical_classification ,Reactive oxygen species ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Immunology ,Disease Progression ,Cancer research ,KRAS ,Reactive Oxygen Species ,Carcinogenesis ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDA) is characterized by expression of oncogenic KRas which drives all aspects of tumorigenesis. Oncogenic KRas induces the formation of reactive oxygen species (ROS) which have been implicated in initiation and progression of PDA. To facilitate tumor promoting levels and to avoid oncogene-induced senescence or cytotoxicity, ROS homeostasis in PDA cells is balanced by additional up-regulation of antioxidant systems. Areas covered: We examine the sources of ROS in PDA, the mechanisms by which ROS homeostasis is maintained, and the biological consequences of ROS in PDA. Additionally, we discuss the potential mechanisms for targeting ROS homoeostasis as a point of therapeutic intervention. An extensive review of the relevant literature as it relates to the topic was conducted using PubMed. Expert commentary: Even though oncogenic mutations in the KRAS gene have been detected in over 95% of human pancreatic adenocarcinoma, targeting its gene product, KRas, has been difficult. The dependency of PDA cells on balancing ROS homeostasis could be an angle for new prevention or treatment strategies. These include use of antioxidants to prevent formation or progression of precancerous lesions, or methods to increase ROS in tumor cells to toxic levels.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.