Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 13(17): e2301941, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38471128

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense and stiff extracellular matrix (ECM) associated with tumor progression and therapy resistance. To further the understanding of how stiffening of the tumor microenvironment (TME) contributes to aggressiveness, a three-dimensional (3D) self-assembling hydrogel disease model is developed based on peptide amphiphiles (PAs, PA-E3Y) designed to tailor stiffness. The model displays nanofibrous architectures reminiscent of native TME and enables the study of the invasive behavior of PDAC cells. Enhanced tuneability of stiffness is demonstrated by interacting thermally annealed aqueous solutions of PA-E3Y (PA-E3Yh) with divalent cations to create hydrogels with mechanical properties and ultrastructure similar to native tumor ECM. It is shown that stiffening of PA-E3Yh hydrogels to levels found in PDAC induces ECM deposition, promotes epithelial-to-mesenchymal transition (EMT), enriches CD133+/CXCR4+ cancer stem cells (CSCs), and subsequently enhances drug resistance. The findings reveal how a stiff 3D environment renders PDAC cells more aggressive and therefore more faithfully recapitulates in vivo tumors.


Subject(s)
Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Extracellular Matrix , Hydrogels , Neoplastic Stem Cells , Pancreatic Neoplasms , Tumor Microenvironment , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Hydrogels/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Extracellular Matrix/metabolism , Tumor Microenvironment/drug effects , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Peptides/chemistry , Peptides/pharmacology , Phenotype , Receptors, CXCR4/metabolism
2.
Cell Rep ; 38(4): 110227, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081338

ABSTRACT

In pancreatic ductal adenocarcinoma (PDAC), differentiation of pancreatic stellate cells (PSCs) into myofibroblast-like cancer-associated fibroblasts (CAFs) can both promote and suppress tumor progression. Here, we show that the Rho effector protein kinase N2 (PKN2) is critical for PSC myofibroblast differentiation. Loss of PKN2 is associated with reduced PSC proliferation, contractility, and alpha-smooth muscle actin (α-SMA) stress fibers. In spheroid co-cultures with PDAC cells, loss of PKN2 prevents PSC invasion but, counter-intuitively, promotes invasive cancer cell outgrowth. PKN2 deletion induces a myofibroblast to inflammatory CAF switch in the PSC matrisome signature both in vitro and in vivo. Further, deletion of PKN2 in the pancreatic stroma induces more locally invasive, orthotopic pancreatic tumors. Finally, we demonstrate that a PKN2KO matrisome signature predicts poor outcome in pancreatic and other solid human cancers. Our data indicate that suppressing PSC myofibroblast function can limit important stromal tumor-suppressive mechanisms, while promoting a switch to a cancer-supporting CAF phenotype.


Subject(s)
Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Animals , Humans , Mice , Pancreatic Stellate Cells/metabolism , Phenotype , Protein Kinase C/metabolism , Tumor Microenvironment/physiology
3.
Nat Commun ; 12(1): 5623, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34561461

ABSTRACT

Patient-derived in vivo models of human cancer have become a reality, yet their turnaround time is inadequate for clinical applications. Therefore, tailored ex vivo models that faithfully recapitulate in vivo tumour biology are urgently needed. These may especially benefit the management of pancreatic ductal adenocarcinoma (PDAC), where therapy failure has been ascribed to its high cancer stem cell (CSC) content and high density of stromal cells and extracellular matrix (ECM). To date, these features are only partially reproduced ex vivo using organoid and sphere cultures. We have now developed a more comprehensive and highly tuneable ex vivo model of PDAC based on the 3D co-assembly of peptide amphiphiles (PAs) with custom ECM components (PA-ECM). These cultures maintain patient-specific transcriptional profiles and exhibit CSC functionality, including strong in vivo tumourigenicity. User-defined modification of the system enables control over niche-dependent phenotypes such as epithelial-to-mesenchymal transition and matrix deposition. Indeed, proteomic analysis of these cultures reveals improved matrisome recapitulation compared to organoids. Most importantly, patient-specific in vivo drug responses are better reproduced in self-assembled cultures than in other models. These findings support the use of tuneable self-assembling platforms in cancer research and pave the way for future precision medicine approaches.


Subject(s)
Bioengineering/methods , Carcinoma, Pancreatic Ductal/pathology , Models, Biological , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Stromal Cells/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Culture Techniques/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Reproducibility of Results , Stromal Cells/metabolism , Tumor Cells, Cultured
4.
Cell Chem Biol ; 28(3): 283-299, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740433

ABSTRACT

The use of phenotypic screening was central to the discovery and development of novel thalidomide analogs, the IMiDs (immunomodulatory drugs) agents. With the discovery that these agents bind the E3 ligase, CRL4CRBN, and alter its substrate specificity, there has been a great deal of endeavor to discover other small molecules that can modulate alternative E3 ligases. Furthermore, the chemical properties necessary for drug discovery and the rules by which neo-substrates are selected for degradation are being defined in the context of phenotypic alterations in specific cellular systems. This review gives a detailed summary of these recent advances and the methodologies being exploited to understand the mechanism of action of emerging protein degradation therapies.


Subject(s)
Drug Development , Immunologic Factors/pharmacology , Ubiquitin-Protein Ligases/metabolism , Drug Evaluation, Preclinical , Humans , Immunologic Factors/chemistry , Phenotype
5.
Gut ; 64(12): 1921-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25841238

ABSTRACT

OBJECTIVES: The tumour stroma/microenvironment not only provides structural support for tumour development, but more importantly it provides cues to cancer stem cells (CSCs) that regulate their self-renewal and metastatic potential. This is certainly true for pancreatic ductal adenocarcinomas (PDAC), where tumour-associated fibroblasts, pancreatic stellate cells and immune cells create an abundant paracrine niche for CSCs via microenvironment-secreted factors. Thus understanding the role that tumour stroma cells play in PDAC development and CSC biology is of utmost importance. DESIGN: Microarray analyses, tumour microarray immunohistochemical assays, in vitro co-culture experiments, recombinant protein treatment approaches and in vivo intervention studies were performed to understand the role that the immunomodulatory cationic antimicrobial peptide 18/LL-37 (hCAP-18/LL-37) plays in PDAC biology. RESULTS: We found that hCAP-18/LL-37 was strongly expressed in the stroma of advanced primary and secondary PDAC tumours and is secreted by immune cells of the stroma (eg, tumour-associated macrophages) in response to tumour growth factor-ß1 and particularly CSC-secreted Nodal/ActivinA. Treatment of pancreatic CSCs with recombinant LL-37 increased pluripotency-associated gene expression, self-renewal, invasion and tumourigenicity via formyl peptide receptor 2 (FPR2)- and P2X purinoceptor 7 receptor (P2X7R)-dependent mechanisms, which could be reversed by inhibiting these receptors. Importantly, in a genetically engineered mouse model of K-Ras-driven pancreatic tumourigenesis, we also showed that tumour formation was inhibited by either reconstituting these mice with bone marrow from cathelicidin-related antimicrobial peptide (ie, murine homologue of hCAP-18/LL-37) knockout mice or by pharmacologically inhibiting FPR2 and P2X7R. CONCLUSIONS: Thus, hCAP-18/LL-37 represents a previously unrecognised PDAC microenvironment factor that plays a critical role in pancreatic CSC-mediated tumourigenesis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Activins/metabolism , Animals , Antimicrobial Cationic Peptides/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Self Renewal/drug effects , Gene Expression/drug effects , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/genetics , Protein Array Analysis , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects , Tissue Array Analysis , Transforming Growth Factor beta1/pharmacology , Cathelicidins
6.
Cancer Res ; 74(24): 7309-20, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25368022

ABSTRACT

Cancer stem cells (CSC) are thought to play a major role in the development and metastatic progression of pancreatic ductal adenocarcinoma (PDAC), one of the deadliest solid tumors. Likewise, the tumor microenvironment contributes critical support in this setting, including from tumor stromal cells and tumor-associated macrophages (TAM) that contribute structural and paracrine-mediated supports, respectively. Here, we show that TAMs secrete the IFN-stimulated factor ISG15, which enhances CSC phenotypes in PDAC in vitro and in vivo. ISG15 was preferentially and highly expressed by TAM present in primary PDAC tumors resected from patients. ISG15 was secreted by macrophages in response to secretion of IFNß by CSC, thereby reinforcing CSC self-renewal, invasive capacity, and tumorigenic potential. Overall, our work demonstrates that ISG15 is a previously unrecognized support factor for CSC in the PDAC microenvironment with a key role in pathogenesis and progression.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Cytokines/genetics , Neoplastic Stem Cells/pathology , Ubiquitins/genetics , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/pathology , Cytokines/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Macrophages/metabolism , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Tumor Microenvironment/genetics , Ubiquitins/biosynthesis
7.
Cell Mol Life Sci ; 71(18): 3599-609, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24573695

ABSTRACT

Epithelial homeostasis within the epidermis is maintained by means of multiple cell-cell adhesion complexes such as adherens junctions, tight junctions, gap junctions, and desmosomes. These complexes co-operate in the formation and the regulation of the epidermal barrier. Disruption of the epidermal barrier through the deregulation of the above complexes is the cause behind a number of skin disorders such as psoriasis, dermatitis, keratosis, and others. During epithelial-to-mesenchymal transition (EMT), epithelial cells lose their adhesive capacities and gain mesenchymal properties. ZEB transcription factors are key inducers of EMT. In order to gain a better understanding of the functional role of ZEB2 in epidermal homeostasis, we generated a mouse model with conditional overexpression of Zeb2 in the epidermis. Our analysis revealed that Zeb2 expression in the epidermis leads to hyperproliferation due to the combined downregulation of different tight junction proteins compromising the epidermal barrier. Using two epidermis-specific in vivo models and in vitro promoter assays, we identified occludin as a new Zeb2 target gene. Immunohistological analysis performed on human skin biopsies covering various pathogeneses revealed ZEB2 expression in the epidermis of pemphigus vulgaris. Collectively, our data support the notion for a potential role of ZEB2 in intracellular signaling of this disease.


Subject(s)
Homeodomain Proteins/physiology , Repressor Proteins/physiology , Skin/metabolism , Tight Junctions/metabolism , Animals , Cell Line , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Mice , Occludin/metabolism , Pemphigus/genetics , Pemphigus/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Skin/pathology , Zinc Finger E-box Binding Homeobox 2
8.
Semin Cancer Biol ; 22(3): 194-207, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22406545

ABSTRACT

Epithelial cancers make up the vast majority of cancer types and, during the transition from benign adenoma to malignant carcinoma and metastasis, epithelial tumor cells acquire a de-differentiated, migratory and invasive behavior. This process of epithelial-mesenchymal transition (EMT) goes along with dramatic changes in cellular morphology, the loss and remodeling of cell-cell and cell-matrix adhesions, and the gain of migratory and invasive capabilities. EMT itself is a multistage process, involving a high degree of cellular plasticity and a large number of distinct genetic and epigenetic alterations, as fully differentiated epithelial cells convert into poorly differentiated, migratory and invasive mesenchymal cells. In the past years, a plethora of genes have been identified that are critical for EMT and metastasis formation. Notably, the EMT process not only induces increased cancer cell motility and invasiveness but also allows cancer cells to avoid apoptosis, anoikis, oncogene addiction, cellular, senescence and general immune defense. Notably, EMT seems to play a critical role in the generation and maintenance of cancer stem cells, highly consistent with the notion that metastatic cells carry the ability to initiate new tumors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell-Matrix Junctions/pathology , Epithelial-Mesenchymal Transition/genetics , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/pathology , Cadherins/genetics , Cadherins/physiology , Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Cell-Matrix Junctions/genetics , Disease Progression , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology , Neoplastic Stem Cells/metabolism , Signal Transduction/physiology , Transcription Factors/physiology
9.
PLoS One ; 5(4): e10162, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20418949

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small RNAs that recognize and regulate mRNA target genes. Multiple lines of evidence indicate that they are key regulators of numerous critical functions in development and disease, including cancer. However, defining the place and function of miRNAs in complex regulatory networks is not straightforward. Systems approaches, like the inference of a module network from expression data, can help to achieve this goal. METHODOLOGY/PRINCIPAL FINDINGS: During the last decade, much progress has been made in the development of robust and powerful module network inference algorithms. In this study, we analyze and assess experimentally a module network inferred from both miRNA and mRNA expression data, using our recently developed module network inference algorithm based on probabilistic optimization techniques. We show that several miRNAs are predicted as statistically significant regulators for various modules of tightly co-expressed genes. A detailed analysis of three of those modules demonstrates that the specific assignment of miRNAs is functionally coherent and supported by literature. We further designed a set of experiments to test the assignment of miR-200a as the top regulator of a small module of nine genes. The results strongly suggest that miR-200a is regulating the module genes via the transcription factor ZEB1. Interestingly, this module is most likely involved in epithelial homeostasis and its dysregulation might contribute to the malignant process in cancer cells. CONCLUSIONS/SIGNIFICANCE: Our results show that a robust module network analysis of expression data can provide novel insights of miRNA function in important cellular processes. Such a computational approach, starting from expression data alone, can be helpful in the process of identifying the function of miRNAs by suggesting modules of co-expressed genes in which they play a regulatory role. As shown in this study, those modules can then be tested experimentally to further investigate and refine the function of the miRNA in the regulatory network.


Subject(s)
Computational Biology/methods , Gene Expression Profiling , Gene Regulatory Networks , MicroRNAs , Neoplasms/genetics , Algorithms , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Humans , RNA, Messenger
10.
Arch Insect Biochem Physiol ; 67(1): 20-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18064699

ABSTRACT

In order to define the regulatory elements responsible for the expression of the medfly hsp83 (Cchsp83) gene, we determined the sequence of a genomic region of the gene that included 3,536 bp upstream of the transcription initiation site, the first untranslated exon of 144 bp, a 275-bp intron, and 516 bp of the second coding exon. Structural analysis of the 5' flanking region revealed the presence of a typical TATA box, 28 bp upstream of the transcription start site, and seven putative heat shock elements (HSEs) further upstream. The 5' untranslated region of the Cchsp83 mRNA was found to contain extensive secondary structure in the first 126 nucleotides. We carried out deletion functional analysis of the proximal promoter region (-380/+139) in vivo by germ line transformation using the lacZ as a reporter gene. We found that sequences in the -380/-86 region are essential for the constitutive expression of the Cchsp83 gene. Under normal conditions, the -380/+139 region was able to drive significant levels of transgene expression in all developmental stages of the medfly as well as in the ovaries and testis. In most stages, the temporal expression pattern of the reporter gene was similar to the respective pattern of the endogenous Cchsp83 gene. Although the -380/+139 promoter region contained two putative HSEs, it was found unable to confer any heat-induced expression in the reporter gene.


Subject(s)
Ceratitis capitata/genetics , Germ Cells/metabolism , Heat-Shock Proteins/genetics , Insect Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Animals, Genetically Modified , Base Sequence , Ceratitis capitata/cytology , Lac Operon , Molecular Sequence Data , Transformation, Genetic
11.
J Cell Biochem ; 101(2): 331-47, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17177294

ABSTRACT

Focal adhesion kinase (FAK) and its downstream signaling targets are implicated in the process of apoptosis induced by external stimuli, in several mammalian systems. In this report, we demonstrate, that medfly (Ceratitis capitata) hemocytes do undergo apoptosis during larval development. In particular, we show using Western blot, ELISA and flow cytometry analysis, that FAK expression silencing in transfected by FAK double-stranded RNA (dsRNA) hemocytes, enhances twofold hemocyte apoptosis, by signaling through Src, MEK/ERK, and PI-3K/Akt signaling pathways. FAK expression silencing, in response to FAK dsRNA treatment, blocks partially the phosphorylation of its downstream targets. Pre-incubation of hemocytes, with specific inhibitors of FAK downstream signaling molecules, demonstrated that all these inhibitors reduced hemocyte viability and enhanced the magnitude of apoptosis about threefold. This data suggest that these pathways contribute to hemocyte survival and/or death during development. The expression and phosphorylation of FAK, Src, PI-3K p85a, Akt, and ERK signaling molecules appear to be dependent upon developmental stages. The expression and phosphorylation of the above signaling molecules, in annexin-positive and annexin-negative hemocytes is also distinct. The maximum expression and phosphorylation of FAK, Src, PI-3K p85a, Akt, and ERK appeared in annexin-positive hemocytes, in both early and late apoptotic hemocytes. The novel aspect of this report is based on the fact that hemocytes attempt to suppress apoptosis, by increasing the expression/phosphorylation of FAK and, hence its downstream targets signaling molecules Src, ERK, PI-3K p85a, and Akt. Evidently, the basic survival pathways among insects and mammals appear to remain unchanged, during evolution.


Subject(s)
Apoptosis/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Hemocytes/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Annexins/metabolism , Cell Survival , Ceratitis capitata/cytology , Ceratitis capitata/embryology , Ceratitis capitata/metabolism , Focal Adhesion Kinase 1/genetics , Gene Silencing , Hemocytes/cytology , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Phosphorylation , RNA, Double-Stranded/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...