Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 173
Filter
Add more filters

Publication year range
1.
Immunity ; 49(2): 326-341.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30054204

ABSTRACT

The maintenance of appropriate arterial tone is critically important for normal physiological arterial function. However, the cellular and molecular mechanisms remain poorly defined. Here, we have shown that in the mouse aorta, resident macrophages prevented arterial stiffness and collagen deposition in the steady state. Using phenotyping, transcriptional profiling, and targeted deletion of Csf1r, we have demonstrated that these macrophages-which are a feature of blood vessels invested with smooth muscle cells (SMCs) in both mouse and human tissues-expressed the hyaluronan (HA) receptor LYVE-l. Furthermore, we have shown they possessed the unique ability to modulate collagen expression in SMCs by matrix metalloproteinase MMP-9-dependent proteolysis through engagement of LYVE-1 with the HA pericellular matrix of SMCs. Our study has unveiled a hitherto unknown homeostatic contribution of arterial LYVE-1+ macrophages through the control of collagen production by SMCs and has identified a function of LYVE-1 in leukocytes.


Subject(s)
Collagen/metabolism , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Vascular Stiffness/physiology , Animals , Aorta/physiology , Female , Glycoproteins/genetics , Humans , Hyaluronic Acid/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
2.
Proc Natl Acad Sci U S A ; 119(11): e2113074119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35254894

ABSTRACT

SignificanceWith obesity on the rise, there is a growing appreciation for intracellular lipid droplet (LD) regulation. Here, we show how saturated fatty acids (SFAs) reduce fat storage-inducing transmembrane protein 2 (FIT2)-facilitated, pancreatic ß cell LD biogenesis, which in turn induces ß cell dysfunction and death, leading to diabetes. This mechanism involves direct acylation of FIT2 cysteine residues, which then marks the FIT2 protein for endoplasmic reticulum (ER)-associated degradation. Loss of ß cell FIT2 and LDs reduces insulin secretion, increases intracellular ceramides, stimulates ER stress, and exacerbates diet-induced diabetes in mice. While palmitate and stearate degrade FIT2, unsaturated fatty acids such as palmitoleate and oleate do not, results of which extend to nutrition and diabetes.


Subject(s)
Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Insulin-Secreting Cells/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Membrane Proteins/genetics , Animals , Cell Line , Endoplasmic Reticulum Stress , Fatty Acids/metabolism , Glucose/metabolism , Glucose Intolerance , Membrane Proteins/metabolism , Mice , Mutation , Palmitates/metabolism , Stearates/metabolism
3.
EMBO Rep ; 23(6): e54271, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35403791

ABSTRACT

The top cause of mortality in patients with nonalcoholic fatty liver disease (NAFLD) is cardiovascular complications. However, mechanisms of NAFLD-associated vasculopathy remain understudied. Here, we show that blood outgrowth endothelial cells (BOECs) from NAFLD subjects exhibit global transcriptional upregulation of chemokines and human leukocyte antigens. In mouse models of diet-induced NAFLD, we confirm heightened endothelial expressions of CXCL12 in the aortas and the liver vasculatures, and increased retention of infiltrated leukocytes within the vessel walls. To elucidate endothelial-immune crosstalk, we performed immunoprofiling by single-cell analysis, uncovering T cell intensification in NAFLD patients. Functionally, treatment with a CXCL12-neutralizing antibody is effective at moderating the enhanced chemotactic effect of NAFLD BOECs in recruiting CD8+ T lymphocytes. Interference with the CXCL12-CXCR4 axis using a CXCR4 antagonist also averts the impact of immune cell transendothelial migration and restores endothelial barrier integrity. Clinically, we detect threefold more circulating damaged endothelial cells in NAFLD patients than in healthy controls. Our work provides insight into the modulation of interactions with effector immune cells to mitigate endothelial injury in NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Cell Movement , Endothelial Cells/metabolism , Humans , Liver/metabolism , Lymphocytes/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction
4.
Am J Respir Crit Care Med ; 207(7): 908-920, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36288294

ABSTRACT

Rationale: Emerging data support the existence of a microbial "gut-lung" axis that remains unexplored in bronchiectasis. Methods: Prospective and concurrent sampling of gut (stool) and lung (sputum) was performed in a cohort of n = 57 individuals with bronchiectasis and subjected to bacteriome (16S rRNA) and mycobiome (18S Internal Transcribed Spacer) sequencing (total, 228 microbiomes). Shotgun metagenomics was performed in a subset (n = 15; 30 microbiomes). Data from gut and lung compartments were integrated by weighted similarity network fusion, clustered, and subjected to co-occurrence analysis to evaluate gut-lung networks. Murine experiments were undertaken to validate specific Pseudomonas-driven gut-lung interactions. Results: Microbial communities in stable bronchiectasis demonstrate a significant gut-lung interaction. Multibiome integration followed by unsupervised clustering reveals two patient clusters, differing by gut-lung interactions and with contrasting clinical phenotypes. A high gut-lung interaction cluster, characterized by lung Pseudomonas, gut Bacteroides, and gut Saccharomyces, is associated with increased exacerbations and greater radiological and overall bronchiectasis severity, whereas the low gut-lung interaction cluster demonstrates an overrepresentation of lung commensals, including Prevotella, Fusobacterium, and Porphyromonas with gut Candida. The lung Pseudomonas-gut Bacteroides relationship, observed in the high gut-lung interaction bronchiectasis cluster, was validated in a murine model of lung Pseudomonas aeruginosa infection. This interaction was abrogated after antibiotic (imipenem) pretreatment in mice confirming the relevance and therapeutic potential of targeting the gut microbiome to influence the gut-lung axis. Metagenomics in a subset of individuals with bronchiectasis corroborated our findings from targeted analyses. Conclusions: A dysregulated gut-lung axis, driven by lung Pseudomonas, associates with poorer clinical outcomes in bronchiectasis.


Subject(s)
Bronchiectasis , Microbiota , Animals , Mice , Prospective Studies , RNA, Ribosomal, 16S/genetics , Lung/microbiology , Bronchiectasis/drug therapy
5.
Int J Mol Sci ; 24(6)2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36982702

ABSTRACT

Blood is conventionally thought to be sterile. However, emerging evidence on the blood microbiome has started to challenge this notion. Recent reports have revealed the presence of genetic materials of microbes or pathogens in the blood circulation, leading to the conceptualization of a blood microbiome that is vital for physical wellbeing. Dysbiosis of the blood microbial profile has been implicated in a wide range of health conditions. Our review aims to consolidate recent findings about the blood microbiome in human health and to highlight the existing controversies, prospects, and challenges around this topic. Current evidence does not seem to support the presence of a core healthy blood microbiome. Common microbial taxa have been identified in some diseases, for instance, Legionella and Devosia in kidney impairment, Bacteroides in cirrhosis, Escherichia/Shigella and Staphylococcus in inflammatory diseases, and Janthinobacterium in mood disorders. While the presence of culturable blood microbes remains debatable, their genetic materials in the blood could potentially be exploited to improve precision medicine for cancers, pregnancy-related complications, and asthma by augmenting patient stratification. Key controversies in blood microbiome research are the susceptibility of low-biomass samples to exogenous contamination and undetermined microbial viability from NGS-based microbial profiling, however, ongoing initiatives are attempting to mitigate these issues. We also envisage future blood microbiome research to adopt more robust and standardized approaches, to delve into the origins of these multibiome genetic materials and to focus on host-microbe interactions through the elaboration of causative and mechanistic relationships with the aid of more accurate and powerful analytical tools.


Subject(s)
Legionella , Microbiota , Humans , Host Microbial Interactions , Dysbiosis/microbiology , Forecasting
7.
Int J Mol Sci ; 23(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36499091

ABSTRACT

Fatty liver disease is an emerging contributor to disease burden worldwide. The past decades of work established the heterogeneous nature of non-alcoholic fatty liver disease (NAFLD) etiology and systemic contributions to the pathogenesis of the disease. This called for the proposal of a redefinition in 2020 to that of metabolic dysfunction-associated fatty liver disease (MAFLD) to better reflect the current understanding of the disease. To date, several clinical cohort studies comparing NAFLD and MAFLD hint at the relevancy of the new nomenclature in enriching for patients with more severe hepatic injury and extrahepatic comorbidities. However, the underlying systemic pathogenesis is still not fully understood. Preclinical animal models have been imperative in elucidating key biological mechanisms in various contexts, including intrahepatic disease progression, interorgan crosstalk and systemic dysregulation. Furthermore, they are integral in developing novel therapeutics against MAFLD. However, substantial contextual variabilities exist across different models due to the lack of standardization in several aspects. As such, it is crucial to understand the strengths and weaknesses of existing models to better align them to the human condition. In this review, we consolidate the implications arising from the change in nomenclature and summarize MAFLD pathogenesis. Subsequently, we provide an updated evaluation of existing MAFLD preclinical models in alignment with the new definitions and perspectives to improve their translational relevance.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Humans , Models, Animal , Disease Progression , Cross Reactions
8.
Int J Mol Sci ; 24(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36613602

ABSTRACT

NAFLD is the most common chronic liver disease worldwide, occurring in both obese and lean patients. It can lead to life-threatening liver diseases and nonhepatic complications, such as cirrhosis and cardiovascular diseases, that burden public health and the health care system. Current care is weight loss through diet and exercise, which is a challenging goal to achieve. However, there are no FDA-approved pharmacotherapies for NAFLD. This review thoroughly examines the clinical trial findings from 22 drugs (Phase 2 and above) and evaluates the future direction that trials should take for further drug development. These trialed drugs can broadly be categorized into five groups-hypoglycemic, lipid-lowering, bile-pathway, anti-inflammatory, and others, which include nutraceuticals. The multitude of challenges faced in these yet-to-be-approved NAFLD drug trials provided insight into a few areas of improvement worth considering. These include drug repurposing, combinations, noninvasive outcomes, standardization, adverse event alleviation, and the need for precision medicine with more extensive consideration of NAFLD heterogenicity in drug trials. Understandably, every evolution of the drug development landscape lies with its own set of challenges. However, this paper believes in the importance of always learning from lessons of the past, with each potential improvement pushing clinical trials an additional step forward toward discovering appropriate drugs for effective NAFLD management.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Hypoglycemic Agents/therapeutic use , Liver Cirrhosis/drug therapy , Obesity/drug therapy , Dietary Supplements
9.
Haematologica ; 106(6): 1671-1683, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33538151

ABSTRACT

The mobilization efficiency of hematopoietic stem/progenitor cells from bone marrow (BM) to circulation by granulocyte colony-stimulating factor (G-CSF) is dramatically dispersed in humans and mice with no mechanistic lead for poor mobilizers. The regulatory mechanism for mobilization efficiency by dietary fat was assessed in mice. Fat-free diet (FFD) for 2 weeks greatly increased mobilization compared to normal diet (ND). The BM mRNA level of peroxisome proliferator-activated receptor δ (PPARδ), a receptor for lipid mediators, was markedly up-regulated by G-CSF in mice fed with ND and displayed strong positive correlation with widely scattered mobilization efficiency. It was hypothesized that BM fat ligand for PPARδ might inhibit mobilization. The PPARδ agonist inhibited mobilization in mice fed with ND and enhanced mobilization by FFD. Treatment with the PPARδ antagonist and chimeric mice with PPARδ+/- BM showed enhanced mobilization. Immunohistochemical staining and flow cytometry revealed that BM PPARδ expression was enhanced by G-CSF mainly in mature/immature neutrophils. BM lipid mediator analysis revealed that G-CSF treatment and FFD resulted in the exhaustion of ω3-polyunsaturated fatty acids such as eicosapentaenoic acid (EPA). EPA induced the up-regulation of genes downstream of PPARδ, such as carnitine palmitoyltransferase-1α and angiopoietin-like protein 4 (Angptl4), in mature/immature neutrophils in vitro and inhibited enhanced mobilization in mice fed with FFD in vivo. Treatment of wild-type mice with the anti-Angptl4 antibody enhanced mobilization together with BM vascular permeability. Collectively, PPARδ signaling in BM mature/immature neutrophils induced by dietary fatty acids negatively regulates mobilization, at least partially, via Angptl4 production.


Subject(s)
Bone Marrow , PPAR delta , Animals , Bone Marrow Cells , Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Mice , PPAR delta/genetics
10.
Inorg Chem ; 60(22): 17276-17287, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34709031

ABSTRACT

A series of activated vinyl azoles was hydrophosphinated in the presence of a chiral palladacycle catalyst under mild conditions to give enantioenriched phosphine azoles with moderate enantioselectivities and yields. The racemic phosphine azoles were transformed into eleven novel chelating phosphine-N-heterocyclic carbene (NHC) platinum complexes. The drug efficacies of nine selected phosphine-NHC platinum(II) chlorides in two cancer cell lines (MKN74 and MCF7) were evaluated, and two were found to exhibit activities comparable to that of cisplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Chelating Agents/pharmacology , Methane/analogs & derivatives , Organoplatinum Compounds/pharmacology , Phosphines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Methane/chemistry , Methane/pharmacology , Molecular Structure , Organoplatinum Compounds/chemistry , Phosphines/chemistry , Tumor Cells, Cultured
11.
Small ; 16(34): e2003757, 2020 08.
Article in English | MEDLINE | ID: mdl-32686344

ABSTRACT

Exogenous sources of amino acids are essential nutrients to fuel cancer growth. Here, the increased demand for amino acid displayed by cancer cells is unconventionally exploited as a design principle to replete cancer cells with apoptosis inducing nanoscopic porous amino acid mimics (Nano-PAAM). A small library consisting of nine essential amino acids nanoconjugates (30 nm) are synthesized, and the in vitro anticancer activity is evaluated. Among the Nano-PAAMs, l-phenylalanine functionalized Nano-PAAM (Nano-pPAAM) has emerged as a novel nanotherapeutics with excellent intrinsic anticancer and cancer-selective properties. The therapeutic efficacy of Nano-pPAAM against a panel of human breast, gastric, and skin cancer cells could be ascribed to the specific targeting of the overexpressed human large neutral amino acid transporter SLC7A5 (LAT-1) in cancer cells, and its intracellular reactive oxygen species (ROS) inducing properties of the nanoporous core. At the mechanistic level, it is revealed that Nano-pPAAM could activate both the extrinsic and intrinsic apoptosis pathways to exert a potent "double-whammy" anticancer effect. The potential clinical utility of Nano-pPAAM is further investigated using an MDA-MB-231 xenograft in NOD scid gamma mice, where an overall suppression of tumor growth by 60% is achieved without the aid of any drugs or application of external stimuli.


Subject(s)
Antineoplastic Agents , Amino Acids , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Mice , Nanoconjugates , Porosity
12.
Environ Sci Technol ; 54(17): 10722-10734, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32786581

ABSTRACT

Humans are exposed to many xenobiotics simultaneously, but little is known about the toxic effects based on chemical-chemical interactions. This study aims at evaluating the binary interactions between 13 common environmental organic compounds (resulting in 78 pairs) by observing their cytotoxicity on HepG2 cells. Among all of the tested pairs, the combination of flame-retardant triphenyl phosphate (TPP) and tris(1,3-dichloro-2-propyl)phosphate (TDCPP) exhibited one of the most significant synergistic effects. We further characterized the transcriptome and metabolome after combined exposure to TPP and TDCPP and individual exposure. The results suggested that the coexposure caused many more changes in gene expressions and cellular activities. The transcriptome data showed that the coexposure triggered significant pathway changes including "cholesterol biosynthesis" and "ATF6-Alpha activated chaperone genes", together with distinct gene ontology (GO) terms such as the "negative regulation of the ERK1 and ERK2 cascade". Additionally, coexposure enhanced the biological activity of liver X receptors and nuclear factor erythroid 2-related factor 2 (Nrf2). The metabolome data showed that coexposure significantly elevated oxidative stress and affected the purine and pyrimidine metabolism. Overall, this study showed that interactions, which may enhance or suppress the biological processes, are common among environmental chemicals, although their environmental relevance should be studied in the future.


Subject(s)
Biological Products , Flame Retardants , Humans , Organophosphates/toxicity , Organophosphorus Compounds , Phosphates
13.
Macromol Rapid Commun ; 41(21): e2000275, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32815257

ABSTRACT

Transplantation of microencapsulated islet cells holds great potential for the treatment of type 1 diabetes mellitus. However, its clinical translation is hampered by the peri-transplantation loss of islet viability and functionality in the microcapsules. In this work, a novel islet cells biomimetic microencapsulant material that is based on the interpenetrating networks of alginate and extracellular matrix (ECM) hydrogel composite (AEC) is presented. The ECM component is derived from human lipoaspirate. In situ encapsulation of pancreatic ß islet cells (MIN6 ß-cells) can be achieved via ionotropic gelation of the alginate matrix and thermal-induced gelation of the pepsin-solubilized ECM pre-gel. Due to the enhanced cell-matrix interaction, islets encapsulated within the AEC microcapsules (≈640 µm) display sevenfold increase in cell growth over 1 week of culture and characteristic glucose-stimulated insulin response in vitro. The results show that the AEC microcapsule is a potent platform to bioaugment the performance of islet cells.


Subject(s)
Alginates , Islets of Langerhans , Extracellular Matrix/metabolism , Humans , Hydrogels/metabolism , Insulin , Insulin Secretion , Islets of Langerhans/metabolism
14.
Int J Mol Sci ; 21(5)2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32143325

ABSTRACT

Daily activities expose muscles to innumerable impacts, causing accumulated tissue damage and inflammation that impairs muscle recovery and function, yet the mechanism modulating the inflammatory response in muscles remains unclear. Our study suggests that Forkhead box A2 (FoxA2), a pioneer transcription factor, has a predominant role in the inflammatory response during skeletal muscle injury. FoxA2 expression in skeletal muscle is upregulated by fatty acids and peroxisome proliferator-activated receptors (PPARs) but is refractory to insulin and glucocorticoids. Using PPARß/δ agonist GW501516 upregulates FoxA2, which in turn, attenuates the production of proinflammatory cytokines and reduces the infiltration of CD45+ immune cells in two mouse models of muscle inflammation, systemic LPS and intramuscular injection of carrageenan, which mimic localized exercise-induced inflammation. This reduced local inflammatory response limits tissue damage and restores muscle tetanic contraction. In line with these results, a deficiency in either PPARß/δ or FoxA2 diminishes the action of the PPARß/δ agonist GW501516 to suppress an aggravated inflammatory response. Our study suggests that FoxA2 in skeletal muscle helps maintain homeostasis, acting as a gatekeeper to maintain key inflammation parameters at the desired level upon injury. Therefore, it is conceivable that certain myositis disorders or other forms of painful musculoskeletal diseases may benefit from approaches that increase FoxA2 activity in skeletal muscle.


Subject(s)
Hepatocyte Nuclear Factor 3-beta/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , PPAR delta/agonists , PPAR-beta/agonists , Animals , Cytokines/metabolism , Gene Expression Regulation , Glucocorticoids/metabolism , HEK293 Cells , Homeostasis , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Thiazoles/pharmacology , Transcriptional Activation , Up-Regulation
15.
Int J Mol Sci ; 21(12)2020 Jun 20.
Article in English | MEDLINE | ID: mdl-32575793

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR)ß/δ is a member of the nuclear receptor superfamily of transcription factors, which plays fundamental roles in cell proliferation and differentiation, inflammation, adipogenesis, and energy homeostasis. Previous studies demonstrated a reduced choroidal neovascularization (CNV) in Pparß/δ-deficient mice. However, PPARß/δ's role in physiological blood vessel formation and vessel remodeling in the retina has yet to be established. Our study showed that PPARß/δ is specifically required for disordered blood vessel formation in the retina. We further demonstrated an increased arteriovenous crossover and wider venous caliber in Pparß/δ-haplodeficient mice. In summary, these results indicated a critical role of PPARß/δ in pathological angiogenesis and blood vessel remodeling in the retina.


Subject(s)
Choroidal Neovascularization/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Vascular Remodeling/genetics , Animals , Cells, Cultured , Disease Models, Animal , Haploinsufficiency , Humans , Lasers/adverse effects , Mice , Retinal Vessels/cytology , Retinal Vessels/metabolism
16.
Mol Cancer ; 18(1): 65, 2019 03 30.
Article in English | MEDLINE | ID: mdl-30927919

ABSTRACT

Field cancerization and metastasis are the leading causes for cancer recurrence and mortality in cancer patients. The formation of primary, secondary tumors or metastasis is greatly influenced by multifaceted tumor-stroma interactions, in which stromal components of the tumor microenvironment (TME) can affect the behavior of the cancer cells. Many studies have identified cytokines and growth factors as cell signaling molecules that aid cell to cell communication. However, the functional contribution of reactive oxygen species (ROS), a family of volatile chemicals, as communication molecules are less understood. Cancer cells and various tumor-associated stromal cells produce and secrete a copious amount of ROS into the TME. Intracellular ROS modulate cell signaling cascades that aid in the acquisition of several hallmarks of cancers. Extracellular ROS help to propagate, amplify, and effectively create a mutagenic and oncogenic field which facilitate the formation of multifoci tumors and act as a springboard for metastatic tumor cells. In this review, we summarize our current knowledge of ROS as atypical paracrine signaling molecules for field cancerization and metastasis. Field cancerization and metastasis are often discussed separately; we offer a model that placed these events with ROS as the focal instigating agent in a broader "seed-soil" hypothesis.


Subject(s)
Cell Communication , Neoplasm Metastasis , Neoplasms/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Humans , Neoplasms/metabolism , Signal Transduction
17.
Mol Cancer ; 18(1): 51, 2019 03 30.
Article in English | MEDLINE | ID: mdl-30925918

ABSTRACT

The tumor microenvironment is a complex and dynamic cellular community comprising the tumor epithelium and various tumor-supporting cells such as immune cells, fibroblasts, immunosuppressive cells, adipose cells, endothelial cells, and pericytes. The interplay between the tumor microenvironment and tumor cells represents a key contributor to immune evasiveness, physiological hardiness and the local and systemic invasiveness of malignant cells. Nuclear receptors are master regulators of physiological processes and are known to play pro-/anti-oncogenic activities in tumor cells. However, the actions of nuclear receptors in tumor-supporting cells have not been widely studied. Given the excellent druggability and extensive regulatory effects of nuclear receptors, understanding their biological functionality in the tumor microenvironment is of utmost importance. Therefore, the present review aims to summarize recent evidence about the roles of nuclear receptors in tumor-supporting cells and their implications for malignant processes such as tumor proliferation, evasion of immune surveillance, angiogenesis, chemotherapeutic resistance, and metastasis. Based on findings derived mostly from cell culture studies and a few in vivo animal cancer models, the functions of VDR, PPARs, AR, ER and GR in tumor-supporting cells are relatively well-characterized. Evidence for other receptors, such as RARß, RORγ, and FXR, is limited yet promising. Hence, the nuclear receptor signature in the tumor microenvironment may harbor prognostic value. The clinical prospects of a tumor microenvironment-oriented cancer therapy exploiting the nuclear receptors in different tumor-supporting cells are also encouraging. The major challenge, however, lies in the ability to develop a highly specific drug delivery system to facilitate precision medicine in cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Stromal Cells/drug effects , Tumor Microenvironment/drug effects , Animals , Humans , Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Stromal Cells/metabolism
18.
Cell Immunol ; 343: 103729, 2019 09.
Article in English | MEDLINE | ID: mdl-29397066

ABSTRACT

There is much cellular heterogeneity in the tumor microenvironment. The tumor epithelia and stromal cells co-evolve, and this reciprocal relationship dictates almost every step of cancer development and progression. Despite this, many anticancer therapies are designed around druggable features of tumor epithelia, ignoring the supportive role of stromal cells. Cancer-associated fibroblasts (CAFs) are the dominant cell type within the reactive stroma of many tumor types. Numerous previous studies have highlighted a pro-tumorigenic role for CAFs via secretion of various growth factors, cytokines, chemokines, and the degradation of extracellular matrix. Recent works showed that CAFs secrete H2O2 to effect stromal-mediated field cancerization, transform primary epithelial cells, and aggravate cancer cell aggressiveness, in addition to inflammatory and mitogenic factors. Molecular characterization of CAFs also underscores the importance of Notch and specific nuclear receptor signaling in the activation of CAFs. This review consolidates recent findings of CAFs and highlights areas for future investigations.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms/pathology , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/metabolism , Carcinogenesis , Humans , Neoplasms/immunology , Neoplasms/physiopathology
19.
Langmuir ; 35(23): 7487-7495, 2019 06 11.
Article in English | MEDLINE | ID: mdl-30480453

ABSTRACT

Tumor stromal residing cancer-associated fibroblasts (CAFs) are significant accomplices in the growth and development of malignant neoplasms. As cancer progresses, the stroma undergoes a dramatic remodeling and stiffening of its extracellular matrix (ECM). However, exactly how these biomechanical changes influence the CAF behavior and the functional paracrine crosstalk with the neighboring tumor cells in a 3-dimensional (3D) microenvironment remains elusive. Herein, a collagen and alginate interpenetrating network (CoAl-IPN) hydrogel system was employed as a 3D in vitro surrogate of the cancerous breast tissue stromal niche. In this study, the mechanical properties of CoAl-IPN were precisely fine-tuned with Young's modulus ( E) values of ∼108 and 898 Pa. The results revealed that the 3D polymeric network mechanics and microstructure are critical biophysical determinants of the human breast CAF (b-CAF) morphology, phenotype, and paracrine dialogue with MDA-MB-231 tumoroids. A compliant hydrogel network favors b-CAF spreading, nuclear translocation of the YAP/TAZ mechanosignaling protein, and upregulation of CAF hallmark transcripts. Conversely, a rigid and highly cross-linked hydrogel network imposed a physical entrapment effect on the b-CAFs that limited their spreading and phenotype in a manner that effectively muted their pro-tumorigenic paracrine activity. Collectively, the CoAl-IPN 3D culture system has proven to be a versatile platform in defining the 3D biophysical parameters that could either promote or restrain the protumorigenic activity of b-CAFs and sheds critical mechano-mediated light onto the phenotypic plasticity and corresponding specific bioactivity of b-CAFs in the 3D microenvironment.


Subject(s)
Fibroblasts/cytology , Fibroblasts/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Mechanical Phenomena/drug effects , Phenotype , Alginates/chemistry , Biomechanical Phenomena/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL