Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res ; 83(10): 1560-1562, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37183657

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) contains a desmoplastic stroma that limits blood perfusion and thus the delivery of nutrients, oxygen, and even therapeutics, creating a hypoxic microenvironment that resists nearly all forms of treatment, including immunomodulating therapy. Cancer-associated fibroblasts (CAF) are the main cellular components and producers of stroma in PDAC. Interestingly, CAFs exist as functionally diverse subpopulations derived from distinct lineages, some of which can be either inflammatory (iCAF) or myofibroblastic (myCAF). While previous work has suggested a link between hypoxia and the iCAF phenotype, direct experimental evidence is lacking. In this issue of Cancer Research, Schwörer and colleagues investigate the role of hypoxia and hypoxia-inducible factor-1α (HIF1α) in maintaining fibroblast heterogeneity and promoting tumor progression in PDAC. The authors use a combination of in vitro and orthotopic techniques to identify a strong role for hypoxia in combination with tumor-derived cytokines in maintaining an iCAF phenotype highlighted by IL6 expression. The authors use an innovative in vitro system to simulate oxygen gradients and use these methods to support their assertions regarding hypoxia as a proinflammatory state. These findings suggest that HIF1α promotes the generation of iCAFs, providing novel insight into CAF heterogeneity. See related article by Schwörer et al., p. 1596.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cytokines/metabolism , Oxygen/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/metabolism , Cancer-Associated Fibroblasts/metabolism , Phenotype , Hypoxia/metabolism , Inflammation/pathology , Tumor Microenvironment , Pancreatic Neoplasms
2.
Gastroenterology ; 162(7): 2018-2031, 2022 06.
Article in English | MEDLINE | ID: mdl-35216965

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) has a hypoxic, immunosuppressive stroma that contributes to its resistance to immune checkpoint blockade therapies. The hypoxia-inducible factors (HIFs) mediate the cellular response to hypoxia, but their role within the PDAC tumor microenvironment remains unknown. METHODS: We used a dual recombinase mouse model to delete Hif1α or Hif2α in α-smooth muscle actin-expressing cancer-associated fibroblasts (CAFs) arising within spontaneous pancreatic tumors. The effects of CAF HIF2α expression on tumor progression and composition of the tumor microenvironment were evaluated by Kaplan-Meier analysis, reverse transcription quantitative real-time polymerase chain reaction, histology, immunostaining, and by both bulk and single-cell RNA sequencing. CAF-macrophage crosstalk was modeled ex vivo using conditioned media from CAFs after treatment with hypoxia and PT2399, an HIF2 inhibitor currently in clinical trials. Syngeneic flank and orthotopic PDAC models were used to assess whether HIF2 inhibition improves response to immune checkpoint blockade. RESULTS: CAF-specific deletion of Hif2α, but not Hif1α, suppressed PDAC tumor progression and growth, and improved survival of mice by 50% (n = 21-23 mice/group, Log-rank P = .0009). Deletion of CAF-HIF2 modestly reduced tumor fibrosis and significantly decreased the intratumoral recruitment of immunosuppressive M2 macrophages and regulatory T cells. Treatment with the clinical HIF2 inhibitor PT2399 significantly reduced in vitro macrophage chemotaxis and M2 polarization, and improved tumor responses to immunotherapy in both syngeneic PDAC mouse models. CONCLUSIONS: Together, these data suggest that stromal HIF2 is an essential component of PDAC pathobiology and is a druggable therapeutic target that could relieve tumor microenvironment immunosuppression and enhance immune responses in this disease.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/pathology , Humans , Hypoxia/metabolism , Immune Checkpoint Inhibitors , Immunosuppression Therapy , Mice , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Pancreatic Neoplasms
3.
Methods Mol Biol ; 2262: 251-258, 2021.
Article in English | MEDLINE | ID: mdl-33977481

ABSTRACT

Phospholipid fatty acid (FA) composition influences the biophysical properties of the plasma membrane and plays an important role in cellular signaling. Our previous work has demonstrated that plasma membrane fatty acid composition is an important determinant of oncogenic Ras signaling and that dietary (exogenous) modulation of membrane composition may underlie the chemoprotective benefits of long chain n-3 polyunsaturated fatty acids (PUFA). In this chapter, we describe in vitro methods to modulate membrane phospholipid fatty acid composition of cultured cells using fatty acids complexed to bovine serum albumin (BSA). Furthermore, we describe a method to quantify the biophysical properties of plasma membranes in live cells using Di-4-ANEPPDHQ (Di4) and image-based flow cytometry.


Subject(s)
Cell Membrane/metabolism , Fatty Acids/metabolism , Membrane Fluidity , Phospholipids/metabolism , Serum Albumin, Bovine/metabolism , Animals , Cattle , Flow Cytometry
4.
J Lipid Res ; 62: 100026, 2021.
Article in English | MEDLINE | ID: mdl-33515553

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling drives the formation of many types of cancer, including colon cancer. Docosahexaenoic acid (DHA, 22∶6Δ4,7,10,13,16,19), a chemoprotective long-chain n-3 polyunsaturated fatty acid suppresses EGFR signaling. However, the mechanism underlying this phenotype remains unclear. Therefore, we used super-resolution microscopy techniques to investigate the mechanistic link between EGFR function and DHA-induced alterations to plasma membrane nanodomains. Using isogenic in vitro (YAMC and IMCE mouse colonic cell lines) and in vivo (Drosophila, wild type and Fat-1 mice) models, cellular DHA enrichment via therapeutic nanoparticle delivery, endogenous synthesis, or dietary supplementation reduced EGFR-mediated cell proliferation and downstream Ras/ERK signaling. Phospholipid incorporation of DHA reduced membrane rigidity and the size of EGFR nanoclusters. Similarly, pharmacological reduction of plasma membrane phosphatidic acid (PA), phosphatidylinositol-4,5-bisphosphate (PIP2) or cholesterol was associated with a decrease in EGFR nanocluster size. Furthermore, in DHA-treated cells only the addition of cholesterol, unlike PA or PIP2, restored EGFR nanoscale clustering. These findings reveal that DHA reduces EGFR signaling in part by reshaping EGFR proteolipid nanodomains, supporting the feasibility of using membrane therapy, i.e., dietary/drug-related strategies to target plasma membrane organization, to reduce EGFR signaling and cancer risk.


Subject(s)
Docosahexaenoic Acids
5.
Cancer Lett ; 489: 50-55, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32512024

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is currently the third leading cause of cancer-related deaths and has a 5-year survival rate of less than 10%, far below the ~70% national average for all cancers. This poor prognosis is driven by an extreme resistance to nearly all known cancer treatments, which has long been attributed to hypoxia driven interactions between tumor cells and the supporting stromal microenvironment. The cellular response to hypoxia is driven by the transcription factors known as the hypoxia inducible factors (HIFs), which have been hypothesized to play a role in the pathobiology of PDAC as well as a potential therapeutic target based on years of cell culture data. Attempts to validate the oncogenic role of HIF in PDAC through rigorous spontaneous tumor models have paradoxically shown that the HIFs may act as a tumor suppressor in epithelial cells. Here, we seek to resolve this paradox by discussing the roles of HIFs both in cancer cells and the supporting microenvironment and place them into context of current model systems that could be used to interrogate these interactions. We suggest that HIF may exert its oncogenic influences by modulating the form and function of the stroma rather than direct effects on cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Hypoxia-Inducible Factor 1/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/physiology , Humans , Pancreatic Neoplasms/pathology
6.
Biophys J ; 118(4): 885-897, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31630812

ABSTRACT

The increasing prevalence of adult and adolescent obesity and its associated risk of colorectal cancer reinforces the urgent need to elucidate the underlying mechanisms contributing to the promotion of colon cancer in obese individuals. Adiponectin is an adipose tissue-derived adipokine, whose levels are reduced during obesity. Both epidemiological and preclinical data indicate that adiponectin suppresses colon tumorigenesis. We have previously demonstrated that both adiponectin and AdipoRon, a small-molecule adiponectin receptor agonist, suppress colon cancer risk in part by reducing the number of Lgr5+ stem cells in mouse colonic organoids. However, the mechanism by which the adiponectin signaling pathway attenuates colon cancer risk remains to be addressed. Here, we have hypothesized that adiponectin signaling supports colonic stem cell maintenance through modulation of the biophysical properties of the plasma membrane (PM). Specifically, we investigated the effects of adiponectin receptor activation by AdipoRon on the biophysical perturbations linked to the attenuation of Wnt-driven signaling and cell proliferation as determined by LEF luciferase reporter assay and colonic organoid proliferation, respectively. Using physicochemical sensitive dyes, Di-4-ANEPPDHQ and C-laurdan, we demonstrated that AdipoRon decreased the rigidity of the colonic cell PM. The decrease in membrane rigidity was associated with a reduction in PM free cholesterol levels and the intracellular accumulation of free cholesterol in lysosomes. These results suggest that adiponectin signaling plays a role in modulating cellular cholesterol homeostasis, PM biophysical properties, and Wnt-driven signaling. These findings are noteworthy because they may in part explain how obesity drives colon cancer progression.


Subject(s)
Receptors, Adiponectin , Wnt Signaling Pathway , Animals , Cell Membrane , Cholesterol , Mice , Piperidines
7.
Cancer Metastasis Rev ; 37(2-3): 519-544, 2018 09.
Article in English | MEDLINE | ID: mdl-29860560

ABSTRACT

The cell plasma membrane serves as a nexus integrating extra- and intracellular components, which together enable many of the fundamental cellular signaling processes that sustain life. In order to perform this key function, plasma membrane components assemble into well-defined domains exhibiting distinct biochemical and biophysical properties that modulate various signaling events. Dysregulation of these highly dynamic membrane domains can promote oncogenic signaling. Recently, it has been demonstrated that select membrane-targeted dietary bioactives (MTDBs) have the ability to remodel plasma membrane domains and subsequently reduce cancer risk. In this review, we focus on the importance of plasma membrane domain structural and signaling functionalities as well as how loss of membrane homeostasis can drive aberrant signaling. Additionally, we discuss the intricacies associated with the investigation of these membrane domain features and their associations with cancer biology. Lastly, we describe the current literature focusing on MTDBs, including mechanisms of chemoprevention and therapeutics in order to establish a functional link between these membrane-altering biomolecules, tuning of plasma membrane hierarchal organization, and their implications in cancer prevention.


Subject(s)
Cell Membrane/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biochemical Phenomena , Biomarkers , Biophysical Phenomena , Cell Membrane/chemistry , Cell Membrane/drug effects , Chemoprevention , Dietary Supplements , Humans , Lipid Metabolism , Membrane Lipids/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction
8.
Cancer Res ; 78(14): 3899-3912, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29769200

ABSTRACT

Ras signaling originates from transient nanoscale compartmentalized regions of the plasma membrane composed of specific proteins and lipids. The highly specific lipid composition of these nanodomains, termed nanoclusters, facilitates effector recruitment and therefore influences signal transduction. This suggests that Ras nanocluster proteolipid composition could represent a novel target for future chemoprevention interventions. There is evidence that consumption of fish oil containing long-chain n-3 polyunsaturated fatty acids (n-3 PUFA) such as eicosapentaenoic acid (EPA, 20:5Δ5,8,11,14,17) and docosahexaenoic acid (DHA, 22:6Δ4,7,10,13,16,19) may reduce colon cancer risk in humans, yet the mechanism underlying this effect is unknown. Here, we demonstrate that dietary n-3 PUFA reduce the lateral segregation of cholesterol-dependent and -independent nanoclusters, suppressing phosphatidic acid-dependent oncogenic KRas effector interactions, via their physical incorporation into plasma membrane phospholipids. This results in attenuation of oncogenic Ras-driven colonic hyperproliferation in both Drosophila and murine models. These findings demonstrate the unique properties of dietary n-3 PUFA in the shaping of Ras nanoscale proteolipid complexes and support the emerging role of plasma membrane-targeted therapies.Significance: The influence of dietary long chain n-3 polyunsaturated fatty acids on plasma membrane protein nanoscale organization and KRas signaling supports development of plasma membrane-targeted therapies in colon cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3899/F1.large.jpg Cancer Res; 78(14); 3899-912. ©2018 AACR.


Subject(s)
Cell Membrane/drug effects , Cell Proliferation/drug effects , Fatty Acids, Omega-3/pharmacology , Proteolipids/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/metabolism , Diet , Docosahexaenoic Acids/pharmacology , Drosophila/metabolism , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Fish Oils , Mice , Phospholipids/metabolism
9.
Mol Aspects Med ; 64: 79-91, 2018 12.
Article in English | MEDLINE | ID: mdl-29627343

ABSTRACT

Proteins are often credited as the macromolecule responsible for performing critical cellular functions, however lipids have recently garnered more attention as our understanding of their role in cell function and human health becomes more apparent. Although cellular membranes are the lipid environment in which many proteins function, it is now apparent that protein and lipid assemblies can be organized to form distinct micro- or nanodomains that facilitate signaling events. Indeed, it is now appreciated that cellular function is partly regulated by the specific spatiotemporal lipid composition of the membrane, down to the nanosecond and nanometer scale. Furthermore, membrane composition is altered during human disease processes such as cancer and obesity. For example, an increased rate of lipid/cholesterol synthesis in cancerous tissues has long been recognized as an important aspect of the rewired metabolism of transformed cells. However, the contribution of lipids/cholesterol to cellular function in disease models is not yet fully understood. Furthermore, an important consideration in regard to human health is that diet is a major modulator of cell membrane composition. This can occur directly through incorporation of membrane substrates, such as fatty acids, e.g., n-3 polyunsaturated fatty acids (n-3 PUFA) and cholesterol. In this review, we describe scenarios in which changes in membrane composition impact human health. Particular focus is placed on the importance of intrinsic lipid/cholesterol biosynthesis and metabolism and extrinsic dietary modification in cancer and its effect on plasma membrane properties.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Fatty Acids, Omega-3/metabolism , Neoplasms/prevention & control , Cell Membrane/pathology , Cholesterol/chemistry , Diet , Fatty Acids, Omega-3/chemistry , Humans , Lipid Metabolism/genetics , Neoplasms/diet therapy , Neoplasms/metabolism , Neoplasms/pathology , Obesity/metabolism , Obesity/pathology
10.
Br J Nutr ; 119(2): 163-175, 2018 01.
Article in English | MEDLINE | ID: mdl-29249211

ABSTRACT

Cell membrane fatty acids influence fundamental properties of the plasma membrane, including membrane fluidity, protein functionality, and lipid raft signalling. Evidence suggests that dietary n-3 PUFA may target the plasma membrane of immune cells by altering plasma membrane lipid dynamics, thereby regulating the attenuation of immune cell activation and suppression of inflammation. As lipid-based immunotherapy might be a promising new clinical strategy for the treatment of inflammatory disorders, we conducted in vitro and in vivo experiments to examine the effects of n-3 PUFA on CD4+ T cell membrane order, mitochondrial bioenergetics and lymphoproliferation. n-3 PUFA were incorporated into human primary CD4+ T cells phospholipids in vitro in a dose-dependent manner, resulting in a reduction in whole cell membrane order, oxidative phosphorylation and proliferation. At higher doses, n-3 PUFA induced unique phase separation in T cell-derived giant plasma membrane vesicles. Similarly, in a short-term human pilot study, supplementation of fish oil (4 g n-3 PUFA/d) for 6 weeks in healthy subjects significantly elevated EPA (20 : 5n-3) levels in CD4+ T cell membrane phospholipids, and reduced membrane lipid order. These results demonstrate that the dynamic reshaping of human CD4+ T cell plasma membrane organisation by n-3 PUFA may modulate down-stream clonal expansion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Cell Membrane/drug effects , Fatty Acids, Omega-3/pharmacology , Aged , Aged, 80 and over , Cell Membrane/chemistry , Cell Membrane/physiology , Dietary Fats/administration & dosage , Dietary Supplements , Eicosapentaenoic Acid/blood , Energy Metabolism/drug effects , Fatty Acids/blood , Female , Fish Oils/administration & dosage , Humans , Lymphocyte Activation/drug effects , Male , Membrane Lipids/blood , Membrane Lipids/chemistry , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Phospholipids/blood , Phospholipids/chemistry , Pilot Projects
11.
Biochim Biophys Acta Biomembr ; 1859(9 Pt B): 1668-1678, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28342710

ABSTRACT

In the context of an organism, epithelial cells by nature are designed to be the defining barrier between self and the outside world. This is especially true for the epithelial cells that form the lining of the digestive tract, which absorb nutrients and serve as a barrier against harmful substances. These cells are constantly bathed by a complex mixture of endogenous (bile acids, mucus, microbial metabolites) and exogenous (food, nutrients, drugs) bioactive compounds. From a cell biology perspective, this type of exposure would directly impact the plasma membrane, which consists of a myriad of complex lipids and proteins. The plasma membrane not only functions as a barrier but also as the medium in which cellular signaling complexes form and function. This property is mediated by the organization of the plasma membrane, which is exquisitely temporally (nanoseconds to minutes) and spatially (nanometers to micrometers) regulated. Since numerous bioactive compounds found in the intestinal lumen can directly interact with lipid membranes, we hypothesize that the dynamic reshaping of plasma membrane organization underlies the chemoprotective effect of select membrane targeted dietary bioactives (MTDBs). This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/chemistry , Colorectal Neoplasms/prevention & control , Diet , Humans , Neoplastic Stem Cells/drug effects
12.
Annu Rev Nutr ; 36: 543-70, 2016 07 17.
Article in English | MEDLINE | ID: mdl-27431370

ABSTRACT

The International Agency for Research on Cancer recently released an assessment classifying red and processed meat as "carcinogenic to humans" on the basis of the positive association between increased consumption and risk for colorectal cancer. Diet, however, can also decrease the risk for colorectal cancer and be used as a chemopreventive strategy. Bioactive dietary molecules, such as n-3 polyunsaturated fatty acids, curcumin, and fermentable fiber, have been proposed to exert chemoprotective effects, and their molecular mechanisms have been the focus of research in the dietary/chemoprevention field. Using these bioactives as examples, this review surveys the proposed mechanisms by which they exert their effects, from the nucleus to the cellular membrane. In addition, we discuss emerging technologies involving the culturing of colonic organoids to study the physiological effects of dietary bioactives. Finally, we address future challenges to the field regarding the identification of additional molecular mechanisms and other bioactive dietary molecules that can be utilized in our fight to reduce the incidence of colorectal cancer.


Subject(s)
Colonic Neoplasms/prevention & control , Diet, Healthy , Gene Expression Regulation , Models, Biological , Nutrigenomics/methods , Animals , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/therapeutic use , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/microbiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/prevention & control , Curcumin/metabolism , Curcumin/therapeutic use , DNA Methylation , Dietary Fiber/metabolism , Dietary Fiber/therapeutic use , Epigenesis, Genetic , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/therapeutic use , Fermentation , Gastrointestinal Microbiome , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , MicroRNAs/metabolism , Nutrigenomics/trends , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...