Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Immunol ; 14: 1174140, 2023.
Article in English | MEDLINE | ID: mdl-37638013

ABSTRACT

Introduction: The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods: Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results: Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion: These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.


Subject(s)
Endotoxemia , Radiation Injuries , alpha-Defensins , Humans , Adult , Animals , Mice , Paneth Cells , Dysbiosis , Endotoxemia/etiology , RNA, Ribosomal, 16S , Radiation Injuries/etiology , Cytokines , Inflammation
2.
Cell Signal ; 78: 109850, 2021 02.
Article in English | MEDLINE | ID: mdl-33253914

ABSTRACT

The TP53 gene has been widely studied for its roles in cell cycle control, maintaining genome stability, activating repair mechanisms upon DNA damage, and initiating apoptosis should repair mechanisms fail. Thus, it is not surprising that mutations of p53 are the most common genetic alterations found in human cancer. Emerging evidence indicates that dysregulation of lipid metabolism by p53 can have a profound impact not only on cancer cells but also cells of the tumor microenvironment (TME). In particular, intermediates of the sphingolipid and lysophospholipid pathways regulate many cellular responses common to p53 such as cell survival, migration, DNA damage repair and apoptosis. The majority of these cellular events become dysregulated in cancer as well as cell senescence. In this review, we will provide an account on the seminal contributions of Prof. Lina Obeid, who deciphered the crosstalk between p53 and the sphingolipid pathway particularly in modulating DNA damage repair and apoptosis in non-transformed as well as transformed cells. We will also provide insights on the integrative role of p53 with the lysophosphatidic acid (LPA) signaling pathway in cancer progression and TME regulation.


Subject(s)
Lysophospholipids/metabolism , Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment , Tumor Suppressor Protein p53/metabolism , Humans , Lysophospholipids/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
3.
Cancers (Basel) ; 12(5)2020 May 10.
Article in English | MEDLINE | ID: mdl-32397679

ABSTRACT

The tumor microenvironment (TME) may be best conceptualized as an ecosystem comprised of cancer cells interacting with a multitude of stromal components such as the extracellular matrix (ECM), blood and lymphatic networks, fibroblasts, adipocytes, and cells of the immune system. At the center of this crosstalk between cancer cells and their TME is the bioactive lipid lysophosphatidic acid (LPA). High levels of LPA and the enzyme generating it, termed autotaxin (ATX), are present in many cancers. It is also well documented that LPA drives tumor progression by promoting angiogenesis, proliferation, survival, invasion and metastasis. One of the hallmarks of cancer is the ability to modulate and escape immune detection and eradication. Despite the profound role of LPA in regulating immune functions and inflammation, its role in the context of tumor immunity has not received much attention until recently where emerging studies highlight that this signaling axis may be a means that cancer cells adopt to evade immune detection and eradication. The present review aims to look at the immunomodulatory actions of LPA in baseline immunity to provide a broad understanding of the subject with a special emphasis on LPA and cancer immunity, highlighting the latest progress in this area of research.

4.
Sci Transl Med ; 11(482)2019 03 06.
Article in English | MEDLINE | ID: mdl-30842314

ABSTRACT

The failure of anti-CD20 antibody (Rituximab) as therapy for lupus may be attributed to the transient and incomplete B cell depletion achieved in clinical trials. Here, using an alternative approach, we report that complete and sustained CD19+ B cell depletion is a highly effective therapy in lupus models. CD8+ T cells expressing CD19-targeted chimeric antigen receptors (CARs) persistently depleted CD19+ B cells, eliminated autoantibody production, reversed disease manifestations in target organs, and extended life spans well beyond normal in the (NZB × NZW) F1 and MRL fas/fas mouse models of lupus. CAR T cells were active for 1 year in vivo and were enriched in the CD44+CD62L+ T cell subset. Adoptively transferred splenic T cells from CAR T cell-treated mice depleted CD19+ B cells and reduced disease in naive autoimmune mice, indicating that disease control was cell-mediated. Sustained B cell depletion with CD19-targeted CAR T cell immunotherapy is a stable and effective strategy to treat murine lupus, and its effectiveness should be explored in clinical trials for lupus.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes/immunology , Immunotherapy, Adoptive , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , Lymphocyte Depletion , T-Lymphocytes/metabolism , Animals , Female , Lupus Erythematosus, Systemic/blood , Mice , Phenotype , Proteome/metabolism , Survival Analysis
5.
Adv Biol Regul ; 71: 183-193, 2019 01.
Article in English | MEDLINE | ID: mdl-30243984

ABSTRACT

The lipid mediator lysophosphatidic acid (LPA) in biological fluids is primarily produced by cleavage of lysophospholipids by the lysophospholipase D enzyme Autotaxin (ATX). LPA has been identified and abundantly detected in the culture medium of various cancer cell types, tumor effusates, and ascites fluid of cancer patients. Our current understanding of the physiological role of LPA established its role in fundamental biological responses that include cell proliferation, metabolism, neuronal differentiation, angiogenesis, cell migration, hematopoiesis, inflammation, immunity, wound healing, regulation of cell excitability, and the promotion of cell survival by protecting against apoptotic death. These essential biological responses elicited by LPA are seemingly hijacked by cancer cells in many ways; transcriptional upregulation of ATX leading to increased LPA levels, enhanced expression of multiple LPA GPCR subtypes, and the downregulation of its metabolic breakdown. Recent studies have shown that overexpression of ATX and LPA GPCR can lead to malignant transformation, enhanced proliferation of cancer stem cells, increased invasion and metastasis, reprogramming of the tumor microenvironment and the metastatic niche, and development of resistance to chemo-, immuno-, and radiation-therapy of cancer. The fundamental role of LPA in cancer progression and the therapeutic inhibition of the ATX-LPA axis, although highly appealing, remains unexploited as drug development to these targets has not reached into the clinic yet. The purpose of this brief review is to highlight some unique signaling mechanisms engaged by the ATX-LPA axis and emphasize the therapeutic potential that lies in blocking the molecular targets of the LPA system.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lysophospholipids/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Humans , Neoplasms/pathology
6.
Sci Rep ; 8(1): 16241, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389960

ABSTRACT

Alcohol consumption has been shown to cause dysbiosis, but the mechanism involved in it is unknown. Recurrent colitis is known to induce expression of α-defensins in the colon, but the effect of alcohol consumption on it is not known. We investigated the effect of ethanol on α-defensin expression in the small intestine and colitis-induced expression in colon in mice. Furthermore, we evaluated the effect of human defensin-5 (HD5) on ethanol and colitis-induced gut barrier dysfunction and mucosal damage. Recurrent colitis was induced by feeding dextran sulfate sodium (DSS), 3 cycles of 5-days each with 15 days intervals, followed by 30-days remission. Ethanol was fed during the intervals and recovery in a liquid diet with or without HD5. Expression of α-defensins, tight junction (TJ) integrity and cytokine/chemokine expression were analyzed. Chronic ethanol feeding reduced α-defensin expression in the small intestine and colitis-induced defensin expression in the colon. HD5 attenuated the growth of enterotoxigenic Bacteriodes fragilis and E. coli, but had no effect on non-toxigenic Bacteriodes fragilis or probiotics, the Lactobacilli. Ethanol and colitis elevated Enterobacteriaceae, Firmicutes and Firmicutes to Bacteriodetes ratio in colonic mucosa. HD5 feeding attenuated ethanol and colitis-induced dysbiosis, disruption of intestinal epithelial TJ, mucosal inflammation, expression of pro-inflammatory cytokines and chemokines in the small intestine and colon, and endotoxemia. These results demonstrate that ethanol suppresses intestinal α-defensin expression, leading to dysbiosis, barrier dysfunction, inflammation and endotoxemia. HD5 feeding attenuates intestinal injury caused by ethanol and colitis, indicating that defensin expression is a potential target for treatment of alcoholic tissue injury and colitis.


Subject(s)
Colitis, Ulcerative/drug therapy , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , alpha-Defensins/administration & dosage , Administration, Oral , Animals , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , DNA, Bacterial/isolation & purification , Dextran Sulfate/toxicity , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/microbiology , Dysbiosis/pathology , Ethanol/toxicity , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Tight Junctions/drug effects , Tight Junctions/pathology , Treatment Outcome , alpha-Defensins/chemical synthesis
7.
Cell Signal ; 27(9): 1751-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26027517

ABSTRACT

In this study we characterized the effects of radiation injury on the expression and function of the autotaxin (ATX)-LPA2 GPCR axis. In IEC-6 crypt cells and jejunum enteroids quantitative RT-PCR showed a time- and dose-dependent upregulation of lpa2 in response to γ-irradiation that was abolished by mutation of the NF-κB site in the lpa2 promoter or by inhibition of ATM/ATR kinases with CGK-733, suggesting that lpa2 is a DNA damage response gene upregulated by ATM via NF-κB. The resolution kinetics of the DNA damage marker γ-H2AX in LPA-treated IEC-6 cells exposed to γ-irradiation was accelerated compared to vehicle, whereas pharmacological inhibition of LPA2 delayed the resolution of γ-H2AX. In LPA2-reconstituted MEF cells lacking LPA1&3 the levels of γ-H2AX decreased rapidly, whereas in Vector MEF were high and remained sustained. Inhibition of ERK1&2 or PI3K/AKT signaling axis by pertussis toxin or the C311A/C314A/L351A mutation in the C-terminus of LPA2 abrogated the effect of LPA on DNA repair. LPA2 transcripts in Lin(-)Sca-1(+)c-Kit(+) enriched for bone marrow stem cells were 27- and 5-fold higher than in common myeloid or lymphoid progenitors, respectively. Furthermore, after irradiation higher residual γ-H2AX levels were detected in the bone marrow or jejunum of irradiated LPA2-KO mice compared to WT mice. We found that γ-irradiation increases plasma ATX activity and LPA level that is in part due to the previously established radiation-induced upregulation of TNFα. These findings identify ATX and LPA2 as radiation-regulated genes that appear to play a physiological role in DNA repair.


Subject(s)
DNA Damage , Gamma Rays , MAP Kinase Signaling System/radiation effects , Phosphoric Diester Hydrolases/metabolism , Radiation Injuries, Experimental/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cell Line , Jejunum/metabolism , Jejunum/pathology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/pathology , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Diester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/pathology , Rats , Receptors, Lysophosphatidic Acid/genetics , Response Elements
8.
Radiat Res ; 183(4): 465-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25807318

ABSTRACT

We have previously demonstrated that the small molecule octadecenyl thiophosphate (OTP), a synthetic mimic of the growth factor-like mediator lysophosphatidic acid (LPA), showed radioprotective activity in a mouse model of total-body irradiation (TBI) when given orally or intraperitoneally 30 min before exposure to 9 Gy γ radiation. In the current study, we evaluated the effects of OTP, delivered subcutaneously, for radioprotection or radiomitigation from -24 h before to up to +72 h postirradiation using a mouse TBI model with therapeutic doses at around 1 mg/kg. OTP was injected at 10 mg/kg without observable toxic side effects in mice, providing a comfortable safety margin. Treatment of C57BL/6 mice with a single dose of OTP over the time period from -12 h before to +26 h after a lethal dose of TBI reduced mortality by 50%. When administered at +48 h to +72 h postirradiation (LD50/30 to LD100/30), OTP reduced mortality by ≥34%. OTP administered at +24 h postirradiation significantly elevated peripheral white blood cell and platelet counts, increased crypt survival in the jejunum, enhanced intestinal glucose absorption and reduced endotoxin seepage into the blood. In the 6.4-8.6 Gy TBI range using LD50/10 as the end point, OTP yielded a dose modification factor of 1.2. The current data indicate that OTP is a potent radioprotector and radiomitigator ameliorating the mortality and tissue injury of acute hematopoietic as well as acute gastrointestinal radiation syndrome.


Subject(s)
Acute Radiation Syndrome/prevention & control , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Lysophospholipids/metabolism , Organophosphorus Compounds/pharmacology , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD34/metabolism , Biological Transport/drug effects , Biological Transport/radiation effects , Biomimetic Materials/adverse effects , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/radiation effects , Dose-Response Relationship, Drug , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Glucose/metabolism , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , LIM Domain Proteins/metabolism , Leukocyte Count , Mice , Mice, Inbred C57BL , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/pharmacokinetics , Phosphoproteins/metabolism , Platelet Count , Proteasome Endopeptidase Complex , Radiation-Protective Agents/adverse effects , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/pharmacology , Sodium-Hydrogen Exchangers/metabolism , Transcription Factors/metabolism , Whole-Body Irradiation/adverse effects
9.
Chem Biol ; 22(2): 206-16, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25619933

ABSTRACT

Pharmacological mitigation of injuries caused by high-dose ionizing radiation is an unsolved medical problem. A specific nonlipid agonist of the type 2 G protein coupled receptor for lysophosphatidic acid (LPA2) 2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl]benzoic acid (DBIBB) when administered with a postirradiation delay of up to 72 hr reduced mortality of C57BL/6 mice but not LPA2 knockout mice. DBIBB mitigated the gastrointestinal radiation syndrome, increased intestinal crypt survival and enterocyte proliferation, and reduced apoptosis. DBIBB enhanced DNA repair by augmenting the resolution of γ-H2AX foci, increased clonogenic survival of irradiated IEC-6 cells, attenuated the radiation-induced death of human CD34(+) hematopoietic progenitors and enhanced the survival of the granulocyte/macrophage lineage. DBIBB also increased the survival of mice suffering from the hematopoietic acute radiation syndrome after total-body irradiation. DBIBB represents a drug candidate capable of mitigating acute radiation syndrome caused by high-dose γ-radiation to the hematopoietic and gastrointestinal system.


Subject(s)
Apoptosis/drug effects , Lysophospholipids/pharmacology , Naphthalimides/pharmacology , Receptors, Lysophosphatidic Acid/agonists , Sulfonamides/pharmacology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Acute Radiation Syndrome/prevention & control , Animals , Apoptosis/radiation effects , Binding Sites , Caspase 8/metabolism , Cell Line , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Gamma Rays , Histones/metabolism , Humans , Lysophospholipids/chemistry , Lysophospholipids/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Naphthalimides/chemistry , Naphthalimides/therapeutic use , Protein Structure, Tertiary , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Sulfonamides/chemistry , Sulfonamides/therapeutic use
10.
Mol Cancer Res ; 13(1): 174-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25158955

ABSTRACT

UNLABELLED: Autotaxin (ENPP2/ATX) and lysophosphatidic acid (LPA) receptors represent two key players in regulating cancer progression. The present study sought to understand the mechanistic role of LPA G protein-coupled receptors (GPCR), not only in the tumor cells but also in stromal cells of the tumor microenvironment. B16F10 melanoma cells predominantly express LPA5 and LPA2 receptors but lack LPA1. LPA dose dependently inhibited invasion of cells across a Matrigel layer. RNAi-mediated knockdown of LPA5 relieved the inhibitory effect of LPA on invasion without affecting basal invasion. This suggests that LPA5 exerts an anti-invasive action in melanoma cells in response to LPA. In addition, both siRNA-mediated knockdown and pharmacologic inhibition of LPA2 reduced the basal rate invasion. Unexpectedly, when probing the role of this GPCR in host tissues, it was found that the incidence of melanoma-derived lung metastasis was greatly reduced in LPA5 knockout (KO) mice compared with wild-type (WT) mice. LPA1-KO but not LPA2-KO mice also showed diminished melanoma-derived lung metastasis, suggesting that host LPA1 and LPA5 receptors play critical roles in the seeding of metastasis. The decrease in tumor cell residence in the lungs of LPA1-KO and LPA5-KO animals was apparent 24 hours after injection. However, KO of LPA1, LPA2, or LPA5 did not affect the subcutaneous growth of melanoma tumors. IMPLICATIONS: These findings suggest that tumor and stromal LPA receptors, in particular LPA1 and LPA5, play different roles in invasion and the seeding of metastasis.


Subject(s)
Lung Neoplasms/genetics , Melanoma, Experimental/genetics , Receptors, Lysophosphatidic Acid/genetics , Animals , Carcinogenesis/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Phosphoric Diester Hydrolases/genetics , Signal Transduction/genetics , Tumor Microenvironment
11.
J Biol Chem ; 287(23): 19018-28, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22511778

ABSTRACT

DiGeorge Critical Region 8 (DGCR8) is a double-stranded RNA-binding protein that interacts with Drosha and facilitates microRNA (miRNA) maturation. However, the role of DGCR8 in vascular smooth muscle cells (VSMCs) is not well understood. To investigate whether DGCR8 contributes to miRNA maturation in VSMCs, we generated DGCR8 conditional knockout (cKO) mice by crossing VSMC-specific Cre mice (SM22-Cre) with DGCR8(loxp/loxp) mice. We found that loss of DGCR8 in VSMCs resulted in extensive liver hemorrhage and embryonic mortality between embryonic days (E) 12.5 and E13.5. DGCR8 cKO embryos displayed dilated blood vessels and disarrayed vascular architecture. Blood vessels were absent in the yolk sac of DGCR8 KOs after E12.5. Disruption of DGCR8 in VSMCs reduced VSMC proliferation and promoted apoptosis in vitro and in vivo. In DGCR8 cKO embryos and knockout VSMCs, differentiation marker genes, including αSMA, SM22, and CNN1, were significantly down-regulated, and the survival pathways of ERK1/2 mitogen-activated protein kinase and the phosphatidylinositol 3-kinase/AKT were attenuated. Knockout of DGCR8 in VSMCs has led to down-regulation of the miR-17/92 and miR-143/145 clusters. We further demonstrated that the miR-17/92 cluster promotes VSMC proliferation and enhances VSMC marker gene expression, which may contribute to the defects of DGCR8 cKO mutants. Our results indicate that the DGCR8 gene is required for vascular development through the regulation of VSMC proliferation, apoptosis, and differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , MicroRNAs/metabolism , Muscle, Smooth, Vascular/embryology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/physiology , Proteins/metabolism , Animals , Embryo Loss/genetics , Embryo Loss/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Proteins/genetics , RNA-Binding Proteins , Yolk Sac/blood supply , Yolk Sac/cytology , Yolk Sac/enzymology
12.
Biochem Soc Trans ; 40(1): 31-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260662

ABSTRACT

LPA (lysophosphatidic acid, 1-acyl-2-hydroxy-sn-glycero-3-phosphate), is a growth factor-like lipid mediator that regulates many cellular functions, many of which are unique to malignantly transformed cells. The simple chemical structure of LPA and its profound effects in cancer cells has attracted the attention of the cancer therapeutics field and drives the development of therapeutics based on the LPA scaffold. In biological fluids, LPA is generated by ATX (autotaxin), a lysophospholipase D that cleaves the choline/serine headgroup from lysophosphatidylcholine and lysophosphatidylserine to generate LPA. In the present article, we review some of the key findings that make the ATX-LPA signalling axis an emerging target for cancer therapy.


Subject(s)
Neoplasms/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Lysophospholipids/metabolism , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasms/drug therapy , Neoplasms/pathology , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/genetics , Signal Transduction , Xenograft Model Antitumor Assays
13.
Am J Respir Cell Mol Biol ; 46(4): 461-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22052879

ABSTRACT

Both hyperoxia and mechanical ventilation can independently cause lung injury. In combination, these insults produce accelerated and severe lung injury. We recently reported that pre-exposure to hyperoxia for 12 hours, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone. We also reported that such injury and apoptosis are inhibited by antioxidant treatment. In this study, we hypothesized that apoptosis signal-regulating kinase-1 (ASK-1), a redox-sensitive, mitogen-activated protein kinase kinase kinase, plays a role in lung injury and apoptosis in this model. To determine the role of ASK-1 in lung injury, the release of inflammatory mediators and apoptosis, attributable to 12 hours of hyperoxia, were followed by large tidal volume mechanical ventilation with hyperoxia. Wild-type and ASK-1 knockout mice were subjected to hyperoxia (Fi(O(2)) = 0.9) for 12 hours before 4 hours of large tidal mechanical ventilation (tidal volume = 25 µl/g) with hyperoxia, and were compared with nonventilated control mice. Lung injury, apoptosis, and cytokine release were measured. The deletion of ASK-1 significantly inhibited lung injury and apoptosis, but did not affect the release of inflammatory mediators, compared with the wild-type mice. ASK-1 is an important regulator of lung injury and apoptosis in this model. Further study is needed to determine the mechanism of lung injury and apoptosis by ASK-1 and its downstream mediators in the lung.


Subject(s)
MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/prevention & control , Animals , Apoptosis/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Epithelial Cells/pathology , Female , Hyperoxia/enzymology , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Pulmonary Alveoli/pathology , Ventilator-Induced Lung Injury/pathology
14.
J Appl Physiol (1985) ; 111(5): 1467-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21799126

ABSTRACT

Both prolonged exposure to hyperoxia and large tidal volume mechanical ventilation can each independently cause lung injury. However, the combined impact of these insults is poorly understood. We recently reported that preexposure to hyperoxia for 12 h, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone (Makena et al. Am J Physiol Lung Cell Mol Physiol 299: L711-L719, 2010). The upstream mechanisms of this lung injury and apoptosis have not been clearly elucidated. We hypothesized that lung injury in this model was dependent on oxidative signaling via the c-Jun NH(2)-terminal kinases (JNK). We, therefore, evaluated lung injury and apoptosis in the presence of N-acetyl-cysteine (NAC) in both mouse and cell culture models, and we provide evidence that NAC significantly inhibited lung injury and apoptosis by reducing the production of ROS, activation of JNK, and apoptosis. To confirm JNK involvement in apoptosis, cells treated with a specific JNK inhibitor, SP600125, and subjected to preexposure to hyperoxia, followed by mechanical stretch, exhibited significantly reduced evidence of apoptosis. In conclusion, lung injury and apoptosis caused by preexposure to hyperoxia, followed by high tidal volume mechanical ventilation, induces ROS-mediated activation of JNK and mitochondrial-mediated apoptosis. NAC protects lung injury and apoptosis by inhibiting ROS-mediated activation of JNK and downstream proapoptotic signaling.


Subject(s)
Hyperoxia/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Injury/metabolism , Oxidants/metabolism , Acetylcysteine/pharmacology , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase Inhibitors , Cell Line , Cytochromes c/antagonists & inhibitors , Cytochromes c/metabolism , Epithelial Cells/metabolism , Hyperoxia/etiology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Respiration, Artificial/adverse effects , Respiration, Artificial/methods , Tidal Volume
15.
Biochem Biophys Res Commun ; 408(3): 369-74, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21371421

ABSTRACT

Dicer is a RNAase III enzyme that cleaves double stranded RNA and generates small interfering RNA (siRNA) and microRNA (miRNA). The goal of this study is to examine the role of Dicer and miRNAs in vascular smooth muscle cells (VSMCs). We deleted Dicer in VSMCs of mice, which caused a developmental delay that manifested as early as embryonic day E12.5, leading to embryonic death between E14.5 and E15.5 due to extensive hemorrhage in the liver, brain, and skin. Dicer KO embryos showed dilated blood vessels and a disarray of vascular architecture between E14.5 and E15.5. VSMC proliferation was significantly inhibited in Dicer KOs. The expression of VSMC marker genes were significantly downregulated in Dicer cKO embryos. The vascular structure of the yolk sac and embryo in Dicer KOs was lost to an extent that no blood vessels could be identified after E15.5. Expression of most miRNAs examined was compromised in VSMCs of Dicer KO. Our results indicate that Dicer is required for vascular development and regulates vascular remodeling by modulating VSMC proliferation and differentiation.


Subject(s)
DEAD-box RNA Helicases/genetics , Developmental Disabilities/genetics , Embryo Loss/genetics , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neovascularization, Physiologic/genetics , Ribonuclease III/genetics , Animals , Cell Proliferation , Gene Deletion , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology
16.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L711-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833778

ABSTRACT

Both high tidal volume mechanical ventilation (HV) and hyperoxia (HO) have been implicated in ventilator-induced lung injury. However, patients with acute lung injury are often exposed to HO before the application of mechanical ventilation. The potential priming of the lungs for subsequent injury by exposure to HO has not been extensively studied. We provide evidence that HO (90%) for 12 h followed by HV (25 µl/g) combined with HO for 2 or 4 h (HO-12h+HVHO-2h or -4h) induced severe lung injury in mice. Analysis of lung homogenates showed that lung injury was associated with cleavage of executioner caspases, caspases-3 and -7, and their downstream substrate poly(ADP-ribose) polymerase-1 (PARP-1). No significant lung injury or caspase cleavage was seen with either HO for 16 h or HV for up to 4 h. Ventilation for 4 h with HO (HVHO) did not cause significant lung injury without preexposure to HO. Twelve-hour HO followed by lower tidal volume (6 µl/g) mechanical ventilation failed to produce significant injury or caspase cleavage. We also evaluated the initiator caspases, caspases-8 and -9, to determine whether the death receptor or mitochondrial-mediated pathways were involved. Caspase-9 cleavage was observed in HO-12h+HVHO-2h and -4h as well as HO for 16 h. Caspase-8 activation was observed only in HO-12h+HVHO-4h, indicating the involvement of both pathways. Immunohistochemistry and in vitro stretch studies showed caspase cleavage in alveolar epithelial cells. In conclusion, preexposure to HO followed by HV produced severe lung injury associated with alveolar epithelial cell apoptosis.


Subject(s)
Apoptosis/physiology , Epithelial Cells/pathology , Epithelial Cells/physiology , Hyperoxia/complications , Tidal Volume , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/pathology , Animals , Caspases/metabolism , Cell Line , Enzyme Activation , Epithelial Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Poly Adenosine Diphosphate Ribose/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiration, Artificial/adverse effects , Stress, Mechanical
17.
Mol Cell ; 39(3): 421-32, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20705243

ABSTRACT

Cyclic phosphatidic acid (1-acyl-2,3-cyclic-glycerophosphate, CPA), one of nature's simplest phospholipids, is found in cells from slime mold to humans and has a largely unknown function. We find here that CPA is generated in mammalian cells in a stimulus-coupled manner by phospholipase D2 (PLD2) and binds to and inhibits the nuclear hormone receptor PPARgamma with nanomolar affinity and high specificity through stabilizing its interaction with the corepressor SMRT. CPA production inhibits the PPARgamma target-gene transcription that normally drives adipocytic differentiation of 3T3-L1 cells, lipid accumulation in RAW264.7 cells and primary mouse macrophages, and arterial wall remodeling in a rat model in vivo. Inhibition of PLD2 by shRNA, a dominant-negative mutant, or a small molecule inhibitor blocks CPA production and relieves PPARgamma inhibition. We conclude that CPA is a second messenger and a physiological inhibitor of PPARgamma, revealing that PPARgamma is regulated by endogenous agonists as well as by antagonists.


Subject(s)
Adipocytes/metabolism , Macrophages/metabolism , PPAR gamma/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , 3T3-L1 Cells , Animals , Cell Differentiation/physiology , Mice , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , PPAR gamma/genetics , Phosphatidic Acids/genetics , Phospholipase D/genetics , Rats , Transcription, Genetic/physiology
18.
Am J Physiol Lung Cell Mol Physiol ; 298(6): L819-29, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20348281

ABSTRACT

Pulmonary fibroblasts regulate extracellular matrix production and degradation and are critical in maintenance of lung structure, function, and repair, but they also play a central role in lung fibrosis. cAMP-elevating agents inhibit cytokine- and growth factor-stimulated myofibroblast differentiation and collagen synthesis in pulmonary fibroblasts. In the present study, we overexpressed adenylyl cyclase 6 (AC6) in pulmonary fibroblasts and measured cAMP production and collagen synthesis. AC6 overexpression enhanced cAMP production and the inhibition of collagen synthesis mediated by isoproterenol and beraprost, but not the responses to butaprost or PGE(2). To examine if increased AC6 expression would impact the development of fibrosis in an animal model, we generated transgenic mice that overexpress AC6 under a fibroblast-specific promoter, FTS1. Lung fibrosis was induced in FTS1-AC6(+/-) mice and littermate controls by intratracheal instillation of saline or bleomycin. Wild-type mice treated with bleomycin showed extensive peribronchial and interstitial fibrosis and collagen deposition. By contrast, FTS1-AC6(+/-) mice displayed decreased fibrotic development, lymphocyte infiltration (as determined by pathological scoring), and lung collagen content. Thus, AC6 overexpression inhibits fibrogenesis in the lung by reducing pulmonary fibroblast-mediated collagen synthesis and myofibroblast differentiation. Because AC6 overexpression does not lead to enhanced basal or PGE(2)-stimulated levels of cAMP, we conclude that endogenous catecholamines or prostacyclin is produced during bleomycin-induced lung fibrosis and that these signals have antifibrotic potential.


Subject(s)
Adenylyl Cyclases/biosynthesis , Fibroblasts/physiology , Pulmonary Fibrosis/physiopathology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Bleomycin , Calcium-Binding Proteins/physiology , Catecholamines/physiology , Colforsin/pharmacology , Collagen/biosynthesis , Cyclic AMP/biosynthesis , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Fibroblasts/drug effects , Isoproterenol/pharmacology , Lung/metabolism , Lung/pathology , Membrane Microdomains/enzymology , Mice , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , S100 Calcium-Binding Protein A4 , S100 Proteins , Signal Transduction
19.
Cell Signal ; 21(12): 1874-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19709640

ABSTRACT

Lysophosphatidic acid (LPA) and its ether analog alkyl-glycerophosphate (AGP) elicit arterial wall remodeling when applied intralumenally into the uninjured carotid artery. LPA is the ligand of eight GPCRs and the peroxisome proliferator-activated receptor gamma (PPARgamma). We pursued a gene knockout strategy to identify the LPA receptor subtypes necessary for the neointimal response in a non-injury model of carotid remodeling and also compared the effects of AGP and the PPARgamma agonist rosiglitazone (ROSI) on balloon injury-elicited neointima development. In the balloon injury model AGP significantly increased neointima; however, rosiglitazone application attenuated it. AGP and ROSI were also applied intralumenally for 1h without injury into the carotid arteries of LPA(1), LPA(2), LPA(1&2) double knockout, and Mx1Cre-inducible conditional PPARgamma knockout mice targeted to vascular smooth muscle cells, macrophages, and endothelial cells. The neointima was quantified and also stained for CD31, CD68, CD11b, and alpha-smooth muscle actin markers. In LPA(1), LPA(2), LPA(1&2) GPCR knockout, Mx1Cre transgenic, PPARgamma(fl/-), and uninduced Mx1CrexPPARgamma(fl/-) mice AGP- and ROSI-elicited neointima was indistinguishable in its progression and cytological features from that of WT C57BL/6 mice. In PPARgamma(-/-) knockout mice, generated by activation of Mx1Cre-mediated recombination, AGP and ROSI failed to elicit neointima and vascular wall remodeling. Our findings point to a difference in the effects of AGP and ROSI between the balloon injury- and the non-injury chemically-induced neointima. The present data provide genetic evidence for the requirement of PPARgamma in AGP- and ROSI-elicited neointimal thickening in the non-injury model and reveal that the overwhelming majority of the cells in the neointimal layer express alpha-smooth muscle actin.


Subject(s)
Carotid Artery Injuries/drug therapy , Carotid Artery, Common/drug effects , Lysophospholipids/therapeutic use , PPAR gamma/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carotid Artery Injuries/pathology , Carotid Artery, Common/pathology , Carotid Artery, Common/ultrastructure , Gene Knockdown Techniques , Glycerophosphates/therapeutic use , Hypoglycemic Agents/therapeutic use , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/agonists , PPAR gamma/genetics , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Rosiglitazone , Thiazolidinediones/therapeutic use
20.
Diagn Cytopathol ; 36(11): 823-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18831021

ABSTRACT

We report a 29-year-old woman who underwent routine gynecologic evaluation at a community clinic and had a cervical sample drawn for liquid-based cytologic evaluation. At cytology, many hyperchromatic crowded groups (HCG) were present, but a consensus could not be established whether the abnormal cells were primarily glandular or squamous with secondary endocervical glandular involvement. An interpretation of atypical endocervical cells, favor neoplastic, was rendered and biopsy advised if clinically appropriate. At biopsy, the cervix contained synchronous squamous cell carcinoma in situ, secondarily involving endocervical glands, and neighboring adenocarcinoma in situ. Immunohistochemistry for Ki-67 and p16(INK4A) crisply and precisely stained both the lesions, clearly separating them from the adjacent uninvolved mucosa. This case re-emphasizes the challenge associated with accurate evaluation of HCG at cytology, the significance of ancillary testing for surrogate markers of high-risk HPV (HR-HPV) infection, the need for adjunct testing for HPV-DNA in the setting of HCG at cervical cytology, and a recommendation to set up studies to evaluate the role of surrogate markers of HR-HPV infection in cytologic samples with HCG.


Subject(s)
Adenocarcinoma/pathology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/pathology , Adult , Biopsy , Cell Nucleus/pathology , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...