Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Adv Drug Deliv Rev ; 200: 115027, 2023 09.
Article in English | MEDLINE | ID: mdl-37517779

ABSTRACT

The rising incidence and persistent thrombosis in multiple cancers including those that are immunosuppressive highlight the need for understanding the tumor coagulome system and its role beyond hemostatic complications. Immunotherapy has shown significant benefits in solid organ tumors but has been disappointing in the treatment of hypercoagulable cancers, such as glioblastoma and pancreatic ductal adenocarcinomas. Thus, targeting thrombosis to prevent immunosuppression seems a clinically viable approach in cancer treatment. Hypercoagulable tumors often develop fibrin clots within the tumor microenvironment (TME) that dictates the biophysical characteristics of the tumor tissue. The application of systems biology and single-cell approaches highlight the potential role of coagulome or thrombocytosis in shaping the tumor immune microenvironment (TIME). In-depth knowledge of the tumor coagulome would provide unprecedented opportunities to better predict the hemostatic complications, explore how thrombotic stroma modulates tumor immunity, reexamine the significance of clinical biomarkers, and enable steering the stromal versus systemic immune response for boosting the effectiveness of immune checkpoint inhibitors in cancer treatment. We focus on the role of coagulation factors in priming a suppressive TIME and the huge potential of existing anticoagulant drugs in the clinical settings of cancer immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tumor Microenvironment , Immunotherapy/adverse effects , Pancreatic Neoplasms/pathology , Immunosuppression Therapy/adverse effects , Carcinoma, Pancreatic Ductal/pathology
2.
Cancers (Basel) ; 15(11)2023 May 31.
Article in English | MEDLINE | ID: mdl-37296966

ABSTRACT

We have shown that activin A (activin), a TGF-ß superfamily member, has pro-metastatic effects in colorectal cancer (CRC). In lung cancer, activin activates pro-metastatic pathways to enhance tumor cell survival and migration while augmenting CD4+ to CD8+ communications to promote cytotoxicity. Here, we hypothesized that activin exerts cell-specific effects in the tumor microenvironment (TME) of CRC to promote anti-tumoral activity of immune cells and the pro-metastatic behavior of tumor cells in a cell-specific and context-dependent manner. We generated an Smad4 epithelial cell specific knockout (Smad4-/-) which was crossed with TS4-Cre mice to identify SMAD-specific changes in CRC. We also performed IHC and digital spatial profiling (DSP) of tissue microarrays (TMAs) obtained from 1055 stage II and III CRC patients in the QUASAR 2 clinical trial. We transfected the CRC cells to reduce their activin production and injected them into mice with intermittent tumor measurements to determine how cancer-derived activin alters tumor growth in vivo. In vivo, Smad4-/- mice displayed elevated colonic activin and pAKT expression and increased mortality. IHC analysis of the TMA samples revealed increased activin was required for TGF-ß-associated improved outcomes in CRC. DSP analysis identified that activin co-localization in the stroma was coupled with increases in T-cell exhaustion markers, activation markers of antigen presenting cells (APCs), and effectors of the PI3K/AKT pathway. Activin-stimulated PI3K-dependent CRC transwell migration, and the in vivo loss of activin lead to smaller CRC tumors. Taken together, activin is a targetable, highly context-dependent molecule with effects on CRC growth, migration, and TME immune plasticity.

3.
Clin Transl Gastroenterol ; 14(7): e00597, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37162146

ABSTRACT

INTRODUCTION: Diet and decreased gut microbiome diversity has been associated with acute pancreatitis (AP) risk. However, differences in dietary intake, gut microbiome, and their impact on microbial end metabolites have not been studied in AP. We aimed to determine differences in (i) dietary intake (ii) gut microbiome diversity and sulfidogenic bacterial abundance, and (iii) serum short-chain fatty acid (SCFA) and hydrogen sulfide (H 2 S) concentrations in AP and control subjects. METHODS: This case-control study recruited 54 AP and 46 control subjects during hospitalization. Clinical and diet data and stool and blood samples were collected. 16S rDNA sequencing was used to determine gut microbiome alpha diversity and composition. Serum SCFA and H 2 S levels were measured. Machine learning (ML) model was used to identify microbial targets associated with AP. RESULTS: AP patients had a decreased intake of vitamin D 3 , whole grains, fish, and beneficial eicosapentaenoic, docosapentaenoic, and docosahexaenoic acids. AP patients also had lower gut microbiome diversity ( P = 0.021) and a higher abundance of sulfidogenic bacteria including Veillonella sp. and Haemophilus sp., which were associated with AP risk. Serum acetate and H 2 S concentrations were significantly higher in the AP group ( P < 0.001 and P = 0.043, respectively). ML model had 96% predictive ability to distinguish AP patients from controls. DISCUSSION: AP patients have decreased beneficial nutrient intake and gut microbiome diversity. An increased abundance of H 2 S-producing genera in the AP and SCFA-producing genera in the control group and predictive ability of ML model to distinguish AP patients indicates that diet, gut microbiota, and their end metabolites play a key role in AP.


Subject(s)
Gastrointestinal Microbiome , Pancreatitis , Animals , Humans , Pancreatitis/etiology , Case-Control Studies , Acute Disease , Diet , Fatty Acids, Volatile
4.
Pancreas ; 51(6): 580-585, 2022 07 01.
Article in English | MEDLINE | ID: mdl-36206462

ABSTRACT

ABSTRACT: The association between acute pancreatitis (AP) and diabetes mellitus (DM) has long been established, with the initial descriptions of AP patients presenting with DM after a bout of AP published in the 1940s and 50s. However, the potential mechanisms involved, particularly those components related to the immune system, have not been well defined. The Diabetes RElated to Acute pancreatitis and its Mechanisms (DREAM) study is a multicenter clinical study designed to understand the frequency and phenotype of DM developing after AP. This article describes one objective of the DREAM study: to determine the immunologic mechanisms of DM after AP, including the contribution of ß-cell autoimmunity. This component of the study will assess the presence of islet autoimmunity, as well as the magnitude and kinetics of the innate and adaptive immune response at enrollment and during longitudinal follow-up after 1 or more episodes of AP. Finally, DREAM will evaluate the relationship between immune features, DM development, and pancreatitis etiology and severity.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Pancreatitis , Acute Disease , Diabetes Mellitus, Type 1/complications , Humans , Pancreatitis/complications
5.
Oncogene ; 40(43): 6153-6165, 2021 10.
Article in English | MEDLINE | ID: mdl-34511598

ABSTRACT

MAP4K4 is a Ste20 member and reported to play important roles in various pathologies, including in cancer. However, the mechanism by which MAP4K4 promotes pancreatic cancer is not fully understood. It is suggested that MAP4K4 might function as a cancer promoter via specific downstream target(s) in an organ-specific manner. Here we identified MLK3 as a direct downstream target of MAP4K4. The MAP4K4 and MLK3 associates with each other, and MAP4K4 phosphorylates MLK3 on Thr738 and increases MLK3 kinase activity and downstream signaling. The phosphorylation of MLK3 by MAP4K4 promotes pancreatic cancer cell proliferation, migration, and colony formation. Moreover, MAP4K4 is overexpressed in human pancreatic tumors and directly correlates with the disease progression. The MAP4K4-specific pharmacological inhibitor, GNE-495, impedes pancreatic cancer cell growth, migration, induces cell death, and arrests cell cycle progression. Additionally, the GNE-495 reduced the tumor burden and extended survival of the KPC mice with pancreatic cancer. The MAP4K4 inhibitor also reduced MAP4K4 protein expression, tumor stroma, and induced cell death in murine pancreatic tumors. These findings collectively suggest that MLK3 phosphorylation by MAP4K4 promotes pancreatic cancer, and therefore therapies targeting MAP4K4 might alleviate the pancreatic cancer tumor burden in patients.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Threonine/chemistry , Mitogen-Activated Protein Kinase Kinase Kinase 11
6.
Cancer Lett ; 515: 1-13, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34052323

ABSTRACT

The transcription factor Glioma-Associated Oncogene Homolog 1 (GLI1) is activated by sonic hedgehog (SHH) cascade and is an established driver of pancreatic ductal adenocarcinoma (PDAC). However, therapies targeting upstream hedgehog signaling have shown little to no efficacy in clinical trials. Here, we identify Mixed Lineage Kinase 3 (MLK3) as a druggable regulator of oncogenic GLI1. Earlier, we reported that MLK3 phosphorylated a peptidyl-prolyl isomerase PIN1 on the S138 site, and the PIN1-pS138 translocated to the nucleus. In this report, we identify GLI1 as one of the targets of PIN1-pS138 and demonstrate that PIN1-pS138 is upregulated in human PDAC and strongly associates with the upregulation of GLI1 and MLK3 expression. Moreover, we also identified two new phosphorylation sites on GLI1, T394, and S1089, which are directly phosphorylated by MLK3 to promote GLI1 nuclear translocation, transcriptional activity, and cell proliferation. Additionally, pharmacological inhibition of MLK3 by CEP-1347 promoted apoptosis in PDAC cell lines, reduced tumor burden, extended survival, and reduced GLI1 expression in the Pdx1-Cre x LSL-KRASG12D x LSL-TP53R172H (KPC) mouse model of PDAC. These findings collectively suggest that MLK3 is an important regulator of oncogenic GLI1 and that therapies targeting MLK3 warrant consideration in the management of PDAC patients.


Subject(s)
MAP Kinase Kinase Kinases/genetics , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Pancreatic Neoplasms/genetics , Zinc Finger Protein GLI1/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Humans , Mice , Pancreatic Neoplasms/pathology , Phosphorylation/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
Sci Rep ; 11(1): 7986, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846512

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has extensive stromal involvement and remains one of the cancers with the highest mortality rates. Activin A has been implicated in colon cancer and its stroma but its role in the stroma of PDAC has not been elucidated. Activin A expression in cancer and stroma was assessed in human PDAC tissue microarrays (TMA). Activin A expression in human TMA is significantly higher in cancer samples, with expression in stroma correlated with shorter survival. Cultured pancreatic stellate cells (PSC) were found to secrete high levels of activin A resulting in PDAC cell migration that is abolished by anti-activin A neutralizing antibody. KPC mice treated with anti-activin A neutralizing antibody were evaluated for tumors, lesions and metastases quantified by immunohistochemistry. KPC mice with increased tumor burden express high plasma activin A. Treating KPC mice with an activin A neutralizing antibody does not reduce primary tumor size but decreases tumor metastases. From these data we conclude that PDAC patients with high activin A expression in stroma have a worse prognosis. PSCs secrete activin A, promoting increased PDAC migration. Inhibition of activin A in mice decreased metastases. Hence, stroma-rich PDAC patients might benefit from activin A inhibition.


Subject(s)
Activins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Activins/blood , Adenocarcinoma/blood , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Prognosis , Stromal Cells/metabolism , Survival Analysis , Tumor Burden , Up-Regulation/genetics
8.
Sci Rep ; 11(1): 378, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33432089

ABSTRACT

Quantitative assessment of soft tissue elasticity is crucial to a broad range of applications, such as biomechanical modeling, physiological monitoring, and tissue diseases diagnosing. However, the modulus measurement of soft tissues, particularly in vivo, has proved challenging since the instrument has to reach the site of soft tissue and be able to measure in a very short time. Here, we present a simple method to measure the elastic modulus of soft tissues on site by exploiting buckling of a long slender bar to quantify the applied force and a spherical indentation to extract the elastic modulus. The method is realized by developing a portable pen-sized instrument (EPen: Elastic modulus pen). The measurement accuracies are verified by independent modulus measures using commercial nanoindenter. Quantitative measurements of the elastic modulus of mouse pancreas, healthy and cancerous, surgically exposed but attached to the body further confirm the potential clinical utility of the EPen.


Subject(s)
Animal Structures/physiology , Biomechanical Phenomena/physiology , Elasticity/physiology , Fiber Optic Technology/instrumentation , Animals , Biophysics/instrumentation , Elastic Modulus , Female , Fiber Optic Technology/methods , Materials Testing , Mice , Mice, Transgenic , Microtechnology/instrumentation , Mobile Applications , Muscle Tonus/physiology , Musculoskeletal Physiological Phenomena , Needles , Stress, Mechanical
10.
Asian J Pharm Sci ; 15(2): 237-251, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32373202

ABSTRACT

Based on the evidence that hemochromatosis, an iron-overload disease, drives hepatocellular carcinoma, we hypothesized that chronic exposure to excess iron, either due to genetic or environmental causes, predisposes an individual to cancer. Using pancreatic cancer as our primary focus, we employed cell culture studies to interrogate the connection between excess iron and cancer, and combined in vitro and in vivo studies to explore the connection further. Ferric ammonium citrate was used as an exogenous iron source. Chronic exposure to excess iron induced epithelial-mesenchymal transition (EMT) in normal and cancer cell lines, loss of p53, and suppression of p53 transcriptional activity evidenced from decreased expression of p53 target genes (p21, cyclin D1, Bax, SLC7A11). To further extrapolate our cell culture data, we generated EL-KrasG12D (EL-Kras) mouse (pancreatic neoplastic mouse model) expressing Hfe+/+ and Hfe-/- genetic background. p53 target gene expression decreased in EL-Kras/Hfe-/- mouse pancreas compared to EL-Kras/Hfe+/+ mouse pancreas. Interestingly, the incidence of acinar-to-ductal metaplasia and cystic pancreatic neoplasms (CPN) decreased in EL-Kras/Hfe-/- mice, but the CPNs that did develop were larger in these mice than in EL-Kras/Hfe+/+ mice. In conclusion, these in vitro and in vivo studies support a potential role for chronic exposure to excess iron as a promoter of more aggressive disease via p53 loss and SLC7A11 upregulation within pancreatic epithelial cells.

11.
Cancer Res ; 80(15): 3101-3115, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32238357

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death with a median survival time of 6-12 months. Most patients present with disseminated disease and the majority are offered palliative chemotherapy. With no approved treatment modalities for patients who progress on chemotherapy, we explored the effects of long-term gemcitabine administration on the tumor microenvironment to identify potential therapeutic options for chemorefractory PDAC. Using a combination of mouse models, primary cell line-derived xenografts, and established tumor cell lines, we first evaluated chemotherapy-induced alterations in the tumor secretome and immune surface proteins by high throughput proteomic arrays. In addition to enhancing antigen presentation and immune checkpoint expression, gemcitabine consistently increased the synthesis of CCL/CXCL chemokines and TGFß-associated signals. These secreted factors altered the composition of the tumor stroma, conferring gemcitabine resistance to cancer-associated fibroblasts in vitro and further enhancing TGFß1 biosynthesis. Combined gemcitabine and anti-PD-1 treatment in transgenic models of murine PDAC failed to alter disease course unless mice also underwent genetic or pharmacologic ablation of TGFß signaling. In the setting of TGFß signaling deficiency, gemcitabine and anti-PD-1 led to a robust CD8+ T-cell response and decrease in tumor burden, markedly enhancing overall survival. These results suggest that gemcitabine successfully primes PDAC tumors for immune checkpoint inhibition by enhancing antigen presentation only following disruption of the immunosuppressive cytokine barrier. Given the current lack of third-line treatment options, this approach warrants consideration in the clinical management of gemcitabine-refractory PDAC. SIGNIFICANCE: These data suggest that long-term treatment with gemcitabine leads to extensive reprogramming of the pancreatic tumor microenvironment and that patients who progress on gemcitabine-based regimens may benefit from multidrug immunotherapy.See related commentary by Carpenter et al., p. 3070 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/15/3101/F1.large.jpg.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Humans , Immunotherapy , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Proteomics , Tumor Microenvironment , Gemcitabine
13.
Sci Rep ; 9(1): 11147, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31367007

ABSTRACT

The pancreatic acinar-enriched miR-216a, miR-216b and miR-217 are encoded within the miR217HG. These miRNAs have been purported to play a tumor suppressive role as their expression is reduced in both human and mouse pancreatic ductal adenocarcinoma (PDAC). To examine this possibility, we generated individual, germline knockout (KO) mice of miR-216a, miR-216b or miR-217. Unlike our previous study showing germline deletion of the miR217HG was embryonic lethal, CRISPR-Cas9 deleted portions of the 5' seed region of the miRNAs produced live births. To investigate possible phenotypes during pancreatic acinar ductal metaplasia (ADM), pancreatic acini from wild type and KO mice were plated on collagen and allowed to transdifferentiate over 4 days. Acini from each of the three miRNA KO mice produced greater numbers of ducts compared to controls. Evaluation of the gene expression during in vitro ADM demonstrated an increase in Krt19 and a reduction in acinar genes (Carboxypeptidase A1, Amylase2a) on day 4 of the transdifferentiation. Recovery was delayed for the miR-216a and miR-216b KOs following caerulein-induced acute pancreatitis. Also predominate in the caerulein treated miR-216a and miR-216b KO mice was the presence of pancreatic duct glands (PDGs). To further establish a phenotype, miRNA KO mice were crossed with EL-KRASG12D (EK) mice and followed up to 13 months of age. While all mice developed severe dysplasia and cystic papillary neoplasms, there existed no apparent phenotypic difference in the miRNA KO/EK mice compared to EK mice. Our data does not support a tumor suppressor role for miR-216a, miR-216b or miR-217 in PDAC and emphasizes the need for phenotypic evaluation of miRNAs in complex in vivo models beyond that performed using cell culture.


Subject(s)
Acinar Cells/pathology , Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , MicroRNAs/genetics , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Transdifferentiation/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/pathology , Pancreatitis/genetics , Pancreatitis/pathology , Signal Transduction/genetics , Pancreatic Neoplasms
14.
Prog Community Health Partnersh ; 13(5): 113-122, 2019.
Article in English | MEDLINE | ID: mdl-31378741

ABSTRACT

BACKGROUND: Health care career pipeline training programs are one solution to increasing the number of minority and underrepresented health care providers. The Chicago Cancer Health Equity Collaborative (ChicagoCHEC) Research Fellows Program, a tri-institutional effort between the University of Illinois at Chicago (UIC), Northeastern Illinois University (NEIU), and Northwestern University (NU), provides a holistic, 8-week summer research fellowship that facilitates self-reflection, professional development, and exposes and guides the novice undergraduate and postbaccalaureate student toward a health care career inclusive of research and scientific discovery. OBJECTIVES: The number of underrepresented students achieving health care careers is minimal. We outline curriculum development, innovation, lessons learned, and selected outcomes from the first three cohorts of the ChicagoCHEC Research Fellows program. METHODS: A tri-institutional, collaborative curricular team was formed consisting of research faculty and staff at NEIU, UIC and NU. Once accepted, fellows experience a cohort model curriculum with particular emphasis to mindful inclusion of nontraditional students. The ChicagoCHEC Research Fellows Program uses evidence-based mentorship models, group reflection, and extensive program evaluation to continuously improve its program model. CONCLUSIONS: The 48 fellow alumni from the first 3 years reported high satisfaction with the program and will continued to be tracked for academic success. The ChicagoCHEC Research Fellows program will continue to provide academic and professional tools, sponsorship, and mentorship opportunities to underrepresented students as they progress toward health care careers. A program such as the ChicagoCHEC Fellows Program can serve as a useful model for increasing the number of minority researchers in health care careers.


Subject(s)
Health Occupations/education , Minority Groups , Universities/organization & administration , Career Choice , Community-Institutional Relations , Humans , Interinstitutional Relations , Mentors , Program Development , Program Evaluation
15.
Proc Natl Acad Sci U S A ; 116(29): 14724-14733, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31266893

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is notorious for its poor survival and resistance to conventional therapies. PI3K signaling is implicated in both disease initiation and progression, and specific inhibitors of selected PI3K p110 isoforms for managing solid tumors are emerging. We demonstrate that increased activation of PI3K signals cooperates with oncogenic Kras to promote aggressive PDAC in vivo. The p110γ isoform is overexpressed in tumor tissue and promotes carcinogenesis via canonical AKT signaling. Its selective blockade sensitizes tumor cells to gemcitabine in vitro, and genetic ablation of p110γ protects against Kras-induced tumorigenesis. Diet/obesity was identified as a crucial means of p110 subunit up-regulation, and in the setting of a high-fat diet, p110γ ablation failed to protect against tumor development, showing increased activation of pAKT and hepatic damage. These observations suggest that a careful and judicious approach should be considered when targeting p110γ for therapy, particularly in obese patients.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Class Ib Phosphatidylinositol 3-Kinase/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Pancreatic Neoplasms/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Diet, High-Fat/adverse effects , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fatty Acids, Omega-6/adverse effects , Female , Glucose/metabolism , Humans , Lipid Metabolism , Liver/pathology , Male , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/metabolism , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation , Gemcitabine
16.
Pancreas ; 48(6): 759-779, 2019 07.
Article in English | MEDLINE | ID: mdl-31206467

ABSTRACT

At the 2018 PancreasFest meeting, experts participating in basic research met to discuss the plethora of available animal models for studying exocrine pancreatic disease. In particular, the discussion focused on the challenges currently facing the field and potential solutions. That meeting culminated in this review, which describes the advantages and limitations of both common and infrequently used models of exocrine pancreatic disease, namely, pancreatitis and exocrine pancreatic cancer. The objective is to provide a comprehensive description of the available models but also to provide investigators with guidance in the application of these models to investigate both environmental and genetic contributions to exocrine pancreatic disease. The content covers both nongenic and genetically engineered models across multiple species (large and small). Recommendations for choosing the appropriate model as well as how to conduct and present results are provided.


Subject(s)
Disease Models, Animal , Genetic Engineering/methods , Pancreas, Exocrine/pathology , Pancreatic Neoplasms/therapy , Pancreatitis/therapy , Acute Disease , Animals , Humans , Mice , Pancreas, Exocrine/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatitis/diagnosis , Pancreatitis/genetics , Rats
17.
PLoS One ; 14(3): e0211711, 2019.
Article in English | MEDLINE | ID: mdl-30897168

ABSTRACT

Current therapies for pancreatic ductal adenocarcinoma (PDAC) only modestly impact survival and can be highly toxic. A greater understanding of the molecules regulating this disease is critical for identifying new drug targets and developing more effective therapies. The L6 family of proteins are known to be positive regulators of tumor growth and metastasis among various cancers. However, little is known about the L6 family member TM4SF18. We investigated the expression and localization of the TM4SF18 protein in normal human pancreas and in PDAC tissue. Utilizing immunohistochemistry (IHC) and western blot analysis, our studies for the first time demonstrate that TM4SF18 is highly expressed in PDAC tumor epithelium. Furthermore, we identified TM4SF18 to be expressed in normal acinar tissue and weakly expressed in normal ducts. Although there is minimal expression in normal ducts, we observed increased TM4SF18 levels in preneoplastic ducts and tumor epithelium. To investigate a functional role of TM4SF18 in PDAC we developed stably-expressing inducible shRNA pancreatic cancer cell lines. Knockdown of the TM4SF18 protein led to a significant decrease in Capan-1 cell growth as measured by the MTT assay, demonstrating this molecule to be a novel regulator of PDAC. Uniquely there is no ortholog of the TM4SF18 gene in mouse or rat prompting us to seek other in vivo experimental models. Using IHC and western blot analysis, expression of TM4SF18 was confirmed in the porcine PDAC model, thus we establish an alternative model to investigate this gene. TM4SF18 represents a promising novel biomarker and therapeutic target for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Tetraspanins/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Animals, Genetically Modified , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Survival/physiology , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression , Gene Knockdown Techniques , Humans , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Sus scrofa
18.
Transl Cancer Res ; 8(Suppl 2): S133-S134, 2019 Mar.
Article in English | MEDLINE | ID: mdl-35117083
19.
Mol Cancer Ther ; 18(3): 613-620, 2019 03.
Article in English | MEDLINE | ID: mdl-30587556

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains remarkably lethal with a 5-year survival rate of 8%. This is mainly attributed to the late stage of presentation, as well as widespread resistance to conventional therapy. In addition, PDAC tumors are largely nonimmunogenic, and most patients have displayed incomplete responses to cancer immunotherapies. Our group has previously identified TGFß as a crucial repressor of antitumor immune function in PDAC, particularly with respect to cytotoxic T lymphocytes. However, pharmacologic inhibition of TGFß signaling has had limited efficacy in clinical trials, failing to promote a significant antitumor immune response. Hence, in this work, we extend our analysis to identify and circumvent the mechanisms of resistance to TGFß signal inhibition in PDAC. Consistent with our previous observations, adoptive transfer of TGFß-insensitive CD8+ T cells led to the near complete regression of neoplastic disease in vivo However, we demonstrate that this cannot be recapitulated via global reduction in TGFß signaling, through either genetic ablation or pharmacologic inhibition of TGFBR1. In fact, tumors with TGFß signal inhibition displayed increased PD-L1 expression and had no observable change in antitumor immunity. Using genetic models of advanced PDAC, we then determined that concomitant inhibition of both TGFß and PD-L1 receptors led to a reduction in the neoplastic phenotype, improving survival and reducing disease-associated morbidity in vivo Combined, these data strongly suggest that TGFß and PD-L1 pathway inhibitors may synergize in PDAC, and this approach warrants clinical consideration.


Subject(s)
Adenocarcinoma/drug therapy , B7-H1 Antigen/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Transforming Growth Factor beta/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunotherapy , Mice , Mice, Transgenic , Receptor, Transforming Growth Factor-beta Type I/genetics , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Microenvironment/drug effects
20.
Sci Rep ; 8(1): 16501, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405152

ABSTRACT

Mice lacking Sirt2 spontaneously develop tumors in multiple organs, as well as when expressed in combination with oncogenic KrasG12D, leading to pancreatic tumors. Here, we report that after caerulein-induced pancreatitis, Sirt2-deficient mice exhibited an increased inflammatory phenotype and delayed pancreatic tissue recovery. Seven days post injury, the pancreas of Sirt2-/- mice display active inflammation, whereas wild-type mice had mostly recovered. In addition, the pancreas from the Sirt2-/- mice exhibited extensive tissue fibrosis, which was still present at six weeks after exposure. The mice lacking Sirt2 also demonstrated an enhanced whole body pro-inflammatory phenotype that was most obvious with increasing age. Importantly, an accumulation of a cell population with spontaneous cancerous KrasG12D mutations was observed in the Sirt2-/- mice that is enhanced in the recovering pancreas after exposure to caerulein. Finally, transcriptome analysis of the pancreas of the Sirt2-/- mice exhibited a pro-inflammatory genomic signature. These results suggest that loss of Sirt2, as well as increased age, enhanced the immune response to pancreatic injury and induced an inflammatory phenotype permissive for the accumulation of cells carrying oncogenic Kras mutations.


Subject(s)
Ceruletide/adverse effects , Mutation , Pancreatitis/etiology , Pancreatitis/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Sirtuin 2/genetics , Animals , Disease Models, Animal , Disease Susceptibility/immunology , Female , Genetic Predisposition to Disease , Immunohistochemistry , Male , Mice , Mice, Knockout , Pancreatitis/pathology , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL
...