Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Burns ; 50(4): 947-956, 2024 May.
Article in English | MEDLINE | ID: mdl-38336496

ABSTRACT

Burn wound conversion is the observed process where superficial partial thickness burns convert into deep partial or full thickness burn injuries. This conversion process often involves surgical excision to achieve timely wound healing. Unfortunately, the pathophysiology of this phenomenon is multifactorial and poorly understood. Thus, a therapeutic intervention that may prevent secondary progression and cell death in burn-injured tissue is desirable. Recent work by our group and others has established that tranexamic acid (TXA) has significant anti-inflammatory properties in addition to its well-known anti-fibrinolytic effects. This study investigates TXA as a novel therapeutic treatment to mitigate burn wound conversion and reduce systemic inflammation. Sprague-Dawley rats were subjected to a hot comb burn contact injury. A subset of animals underwent a similar comb burn with an adjacent 30%TBSA contact injury. The interspaces represent the ischemic zones simulating the zone of stasis. The treatment group received injections of TXA (100 mg/kg) immediately after injury and once daily until euthanasia. Animals were harvested for analyses at 6 h and 7 days after injury. Full-thickness biopsies from the ischemic zones and lung tissue were assessed with established histological techniques. Plasma was collected for measurement of damage associated molecular patterns (DAMPs), and liver samples were used to study inflammatory cytokines expression. Treatment with TXA was associated with reduced burn wound conversion and decreased burn-induced systemic inflammatory response syndrome (SIRS). Lung inflammation and capillary leak were also significantly reduced in TXA treated animals. Future research will elucidate the underlying anti-inflammatory properties of TXA responsible for these findings.


Subject(s)
Antifibrinolytic Agents , Burns , Disease Models, Animal , Edema , Inflammation , Rats, Sprague-Dawley , Tranexamic Acid , Animals , Tranexamic Acid/pharmacology , Tranexamic Acid/therapeutic use , Burns/drug therapy , Burns/complications , Burns/pathology , Rats , Antifibrinolytic Agents/pharmacology , Antifibrinolytic Agents/therapeutic use , Inflammation/drug therapy , Edema/drug therapy , Male , Wound Healing/drug effects , Skin/drug effects , Skin/pathology , Skin/injuries , Liver/drug effects , Liver/pathology , Lung/pathology , Lung/drug effects , Lung/metabolism
2.
Vascul Pharmacol ; 153: 107246, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38040222

ABSTRACT

CTHRC1 is transiently expressed by activated fibroblasts during tissue repair and in certain cancers, and CTHRC1 derived from osteocytes is detectable in circulation. Because its biological activity is poorly understood, we investigated whether the N terminus of CTHRC1 encodes a propeptide requiring cleavage to become activated. The effects of full-length versus cleaved recombinant CTHRC1 on endothelial cell metabolism and gene expression were examined in vitro. Respirometry was performed on Cthrc1 null and wildtype mice to obtain evidence for biological activity of CTHRC1 in vivo. Cleavage of the propeptide observed in vitro was attenuated in the presence of protease inhibitors, and cleaved CTHRC1 significantly promoted glycolysis whereas full-length CTHRC1 was less effective. The respiratory exchange ratio was significantly higher in wildtype mice compared to Cthrc1 null mice, supporting the findings of CTHRC1 promoting glycolysis in vivo. Key enzymes involved in glycolysis were significantly upregulated in endothelial cells in response to treatment with CTHRC1. In healthy human subjects, 58% of the cohort had detectable levels of circulating full-length CTHRC1, whereas all subjects with undetectable levels of full-length CTHRC1 (with one exception) had measurable levels of truncated CTHRC1 (88 pg/ml to >400 ng/ml). Our findings support a concept where CTHRC1 induction in activated fibroblasts at sites of ischemia such as tissue injury or cancer functions to increase glycolysis for ATP production under hypoxic conditions, thereby promoting cell survival and tissue repair. By promoting glycolysis under normoxic conditions, CTHRC1 may also be a contributor to the Warburg effect characteristically observed in many cancers.


Subject(s)
Extracellular Matrix Proteins , Neoplasms , Animals , Humans , Mice , Angiogenesis , Endothelial Cells/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Mice, Knockout
3.
Transfusion ; 63 Suppl 3: S168-S176, 2023 05.
Article in English | MEDLINE | ID: mdl-37070378

ABSTRACT

INTRODUCTION: Tranexamic acid (TXA) is widely used as an antifibrinolytic agent in hemorrhagic trauma patients. The beneficial effects of TXA exceed the suppression of blood loss and include the ability to decrease inflammation and edema. We found that TXA suppresses the release of mitochondrial DNA and enhances mitochondrial respiration. These results allude that TXA could operate through plasmin-independent mechanisms. To address this hypothesis, we compared the effects of TXA on lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines in plasminogen (Plg) null and Plg heterozygous mice. METHODS: Plg null and Plg heterozygous mice were injected with LPS and TXA or LPS only. Four hours later, mice were sacrificed and total RNA was prepared from livers and hearts. Real time quantitative polymerase chain reaction with specific primers was used to assess the effects of LPS and TXA on the expression of pro-inflammatory cytokines. RESULTS: LPS enhanced the expression of Tnfα in the livers and hearts of recipient mice. The co-injection of TXA significantly decreased the effect of LPS both in Plg null and heterozygous mice. A similar trend was observed with LPS-induced Il1α expression in hearts and livers. CONCLUSIONS: The effects of TXA on the endotoxin-stimulated expression of Tnfα and Il1α in mice do not depend on the inhibition of plasmin generation. These results indicate that TXA has other biologically important target(s) besides plasminogen/plasmin. Fully understanding the molecular mechanisms behind the extensive beneficial effects of TXA and future identification of its targets may lead to improvement in the use of TXA in trauma, cardiac, and orthopedic surgical patients.


Subject(s)
Antifibrinolytic Agents , Tranexamic Acid , Mice , Animals , Tranexamic Acid/pharmacology , Fibrinolysin , Fibrinolytic Agents , Endotoxins , Tumor Necrosis Factor-alpha/genetics , Lipopolysaccharides/pharmacology , Antifibrinolytic Agents/pharmacology , Plasminogen/genetics , Plasminogen/metabolism , Hemorrhage
4.
Transfusion ; 62 Suppl 1: S301-S312, 2022 08.
Article in English | MEDLINE | ID: mdl-35834488

ABSTRACT

Tranexamic acid (TXA) is a popular antifibrinolytic drug widely used in hemorrhagic trauma patients and cardiovascular, orthopedic, and gynecological surgical patients. TXA binds plasminogen and prevents its maturation to the fibrinolytic enzyme plasmin. A number of studies have demonstrated the broad life-saving effects of TXA in trauma, superior to those of other antifibrinolytic agents. Besides preventing fibrinolysis and blood loss, TXA has been reported to suppress posttraumatic inflammation and edema. Although the efficiency of TXA transcends simple inhibition of fibrinolysis, little is known about its mechanisms of action besides the suppression of plasmin maturation. Understanding the broader effects of TXA at the cell, organ, and organism levels are required to elucidate its potential mechanisms of action transcending antifibrinolytic activity. In this article, we provide a brief review of the current clinical use of TXA and then focus on the effects of TXA beyond antifibrinolytics such as its anti-inflammatory activity, protection of the endothelial and epithelial monolayers, stimulation of mitochondrial respiration, and suppression of melanogenesis.


Subject(s)
Antifibrinolytic Agents , Blood Coagulation Disorders , Tranexamic Acid , Antifibrinolytic Agents/pharmacology , Antifibrinolytic Agents/therapeutic use , Fibrinolysin/pharmacology , Fibrinolysin/therapeutic use , Fibrinolysis , Hemorrhage , Humans , Tranexamic Acid/pharmacology , Tranexamic Acid/therapeutic use
5.
J Surg Res ; 251: 287-295, 2020 07.
Article in English | MEDLINE | ID: mdl-32199337

ABSTRACT

BACKGROUND: The endothelial glycocalyx (EG) is involved in critical regulatory mechanisms that maintain endothelial vascular integrity. We hypothesized that prolonged cardiopulmonary bypass (CPB) may be associated with EG degradation. We performed an analysis of soluble syndecan-1 levels in relation to duration of CPB, as well as factors associated with cell stress and damage, such as mitochondrial DNA (mtDNA) and inflammation. METHODS: Blood samples from subjects undergoing cardiac surgery with CPB (n = 54) were obtained before and during surgery, 4-8 h and 24 h after completion of CPB, and on postoperative day 4. Flow cytometry was used to determine subpopulations of white blood cells. Plasma levels of mtDNA were determined using quantitative polymerase chain reaction and plasma content of shed syndecan-1 was measured. To determine whether syndecan-1 was signaling white blood cells, the effect of recombinant syndecan-1 on mobilization of neutrophils from bone marrow was tested in mice. RESULTS: CPB is associated with increased mtDNA during surgery, increased syndecan-1 blood levels at 4-8 h, and increased white blood cell count at 4-8 h and 24 h. Correlation analysis revealed significant positive associations between time on CPB and syndecan-1 (rs = 0.488, P < 0.001) and level of syndecan-1 and neutrophil count (rs = 0.351, P = 0.038) at 4-8 h. Intravenous administration of recombinant syndecan-1 in mice resulted in a 2.5-fold increase in the number of circulating neutrophils, concurrent with decreased bone marrow neutrophil number. CONCLUSIONS: Longer duration of CPB is associated with increased plasma levels of soluble syndecan-1, a signal for EG degradation, which can induce neutrophil egress from the bone marrow. Development of therapy targeting EG shedding may be beneficial in patients with prolonged CPB.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Endothelium/ultrastructure , Glycocalyx/physiology , Operative Time , Aged , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cardiopulmonary Bypass/methods , DNA, Mitochondrial/blood , Female , Humans , Interleukin-6/blood , Leukocyte Count , Male , Mice , Middle Aged , Neutrophils/pathology , Recombinant Proteins/pharmacology , Syndecan-1/blood , Syndecan-1/pharmacology
6.
J Cell Physiol ; 234(11): 19121-19129, 2019 11.
Article in English | MEDLINE | ID: mdl-30941770

ABSTRACT

Damage-associated molecular patterns, including mitochondrial DNA (mtDNA) are released during hemorrhage resulting in the development of endotheliopathy. Tranexamic acid (TXA), an antifibrinolytic drug used in hemorrhaging patients, enhances their survival despite the lack of a comprehensive understanding of its cellular mechanisms of action. The present study is aimed to elucidate these mechanisms, with a focus on mitochondria. We found that TXA inhibits the release of endogenous mtDNA from granulocytes and endothelial cells. Furthermore, TXA attenuates the loss of the endothelial monolayer integrity induced by exogenous mtDNA. Using the Seahorse XF technology, it was demonstrated that TXA strongly stimulates mitochondrial respiration. Studies using Mitotracker dye, cells derived from mito-QC mice, and the ActivSignal IPAD assay, indicate that TXA stimulates biogenesis of mitochondria and inhibits mitophagy. These findings open the potential for improvement of the strategies of TXA applications in trauma patients and the development of more efficient TXA derivatives.


Subject(s)
DNA, Mitochondrial/drug effects , Hemorrhage/drug therapy , Tranexamic Acid/pharmacology , Wounds and Injuries/drug therapy , Animals , DNA Damage/drug effects , Endothelial Cells/drug effects , Granulocytes/drug effects , Hemorrhage/genetics , Hemorrhage/pathology , Humans , Mice , Mitochondria/drug effects , Mitophagy/drug effects , Oxidative Phosphorylation/drug effects , Wounds and Injuries/genetics , Wounds and Injuries/pathology
7.
J Trauma Acute Care Surg ; 86(4): 617-624, 2019 04.
Article in English | MEDLINE | ID: mdl-30589751

ABSTRACT

BACKGROUND: Severe burn injuries are known to initiate a profound systemic inflammatory response (SIRS) that may lead to burn shock and other SIRS-related complications. Damage-associated molecular patterns (DAMPs) are important early signaling molecules that initiate SIRS after burn injury. Previous work in a rodent model has shown that application of a topical immune modulator (p38MAPK inhibitor) applied directly to the burn wound decreases cytokine expression, reduces pulmonary inflammation and edema. Our group has demonstrated that tranexamic acid (TXA)-in addition to its use as an antifibrinolytic-has cell protective in vitro effects. We hypothesized that administration of TXA after burn injury would attenuate DAMP release and reduce lung inflammation. METHODS: C57/BL6 male mice underwent a 40% Total Body Surface Area (TBSA) scald burn. Sham animals underwent the same procedure in room temperature water. One treatment group received the topical application of p38MAPK inhibitor after burn injury. The other treatment group received an intraperitoneal administration of TXA after burn injury. Animals were sacrificed at 5 hours. Plasma was collected by cardiac puncture. MtDNA levels in plasma were determined by quantitative Polymerase Chain Reaction (qPCR). Syndecan-1 levels in plasma were measured by ELISA. Lungs were harvested, fixed, and paraffin-embedded. Sections of lungs were stained for antigen to detect macrophages. RESULTS: Topical p38MAPK inhibitor and TXA significantly attenuated mtDNA release. Both TXA and the topical p38MAPK inhibitor reduced lung inflammation as represented by decreased macrophage infiltration. Syndecan-1 levels showed no difference between burn and treatment groups. CONCLUSION: Both p38 MAPK inhibitor and TXA demonstrated the ability to attenuate burn-induced DAMP release and lung inflammation. Beyond its role as an antifibrinolytic, TXA may have significant anti-inflammatory effects pertinent to burn resuscitation. Further study is required; however, TXA may be a useful adjunct in burn resuscitation.


Subject(s)
Alarmins/drug effects , Burns/drug therapy , Burns/physiopathology , Disease Models, Animal , Mitochondria/drug effects , Pneumonia/drug therapy , Tranexamic Acid/pharmacology , Administration, Topical , Animals , DNA, Mitochondrial/antagonists & inhibitors , DNA, Mitochondrial/metabolism , Male , Mice , Mice, Inbred C57BL , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
8.
J Cell Biochem ; 119(3): 2636-2645, 2018 03.
Article in English | MEDLINE | ID: mdl-29058774

ABSTRACT

Highly variable expression of mesoderm-specific transcript (Mest) in adipose tissue among genetically homogeneous mice fed an obesogenic diet, and its positive association with fat mass expansion, suggests that Mest is an epigenetic determinant for the development of obesity. Although the mechanisms by which MEST augments fat accumulation in adipocytes have not been elucidated, it has sequence homology and catalytic peptide motifs which suggests that it functions as an epoxide hydrolase or as a glycerol- or acylglycerol-3-phosphate acyltransferase. To better understand MEST function, detailed studies were performed to precisely define the intracellular organelle localization of MEST using immunofluorescence confocal microscopy. Lentiviral-mediated expression of a C-terminus Myc-DDK-tagged MEST fusion protein expressed in 3T3-L1 preadipocytes/adipocytes, and ear-derived mesenchymal stem cells (EMSC) from mice was observed in the endoplasmic reticulum (ER) membranes and is consistent with previous studies showing endogenous MEST in the membrane fraction of adipose tissue. MEST was not associated with the Golgi apparatus or mitochondria; however, frequent contacts were observed between MEST-positive ER and mitochondria. MEST-positive domains were also shown on the plasma membrane (PM) of non-permeabilized cells but they did not co-localize with ER-PM bridges. Post-adipogenic differentiated 3T3-L1 adipocytes and EMSC showed significant co-localization of MEST with the lipid droplet surface marker perilipin at contact points between the ER and lipid droplet. Identification of MEST as an ER-specific protein that co-localizes with lipid droplets in cells undergoing adipogenic differentiation supports a function for MEST in the facilitation of lipid accumulation and storage in adipocytes.


Subject(s)
Adipocytes/metabolism , Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Proteins/metabolism , 3T3-L1 Cells , Adipocytes/pathology , Animals , Cell Differentiation/physiology , Hypertrophy/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Obesity/metabolism
9.
Biochemistry ; 55(7): 1159-67, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26836284

ABSTRACT

Fibroblast growth factor 1 (FGF1), a ubiquitously expressed pro-angiogenic protein that is involved in tissue repair, carcinogenesis, and maintenance of vasculature stability, is released from the cells via a stress-dependent nonclassical secretory pathway. FGF1 secretion is a result of transmembrane translocation of this protein. It correlates with the ability of FGF1 to permeabilize membranes composed of acidic phospholipids. Like several other nonclassically exported proteins, FGF1 exhibits ß-barrel folding. To assess the role of folding of FGF1 in its secretion, we applied targeted mutagenesis in combination with a complex of biophysical methods and molecular dynamics studies, followed by artificial membrane permeabilization and stress-induced release experiments. It has been demonstrated that a mutation of proline 135 located in the C-terminus of FGF1 results in (i) partial unfolding of FGF1, (ii) a decrease in FGF1's ability to permeabilize bilayers composed of phosphatidylserine, and (iii) drastic inhibition of stress-induced FGF1 export. Thus, folding of FGF1 is critical for its nonclassical secretion.


Subject(s)
Cell Membrane Permeability , Fibroblast Growth Factor 1/chemistry , Models, Molecular , Protein Folding , Amino Acid Substitution , Animals , Calorimetry, Differential Scanning , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , HEK293 Cells , Humans , Kinetics , Lipid Bilayers/chemistry , Membranes, Artificial , Mice , Molecular Dynamics Simulation , Mutation , NIH 3T3 Cells , Permeability , Phosphatidylserines/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
10.
J Cell Physiol ; 231(3): 650-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26218437

ABSTRACT

Transient FGF stimulation of various cell types results in FGF memory--a sustained blockage of efficient proliferative response to FGF and other growth factors. FGF memory establishment requires HDAC activity, indicating its epigenetic character. FGF treatment stimulates proinflammatory NFκB signaling, which is also critical for FGF memory formation. The search for FGF-induced mediators of FGF memory revealed that FGF stimulates HDAC-dependent expression of the inflammatory cytokine IL1α. Similarly to FGF, transient cell treatment with recombinant IL1α inhibits the proliferative response to further FGF and EGF stimulation, but does not prevent FGF receptor-mediated signaling. Interestingly, like cells pretreated with FGF1, cells pretreated with IL1α exhibit enhanced restructuring of actin cytoskeleton and increased migration in response to FGF stimulation. IRAP, a specific inhibitor of IL 1 receptor, and a neutralizing anti-IL1α antibody prevent the formation of FGF memory and rescue an efficient proliferative response to FGF restimulation. A similar effect results following treatment with the anti-inflammatory agents aspirin and dexamethasone. Thus, FGF memory is mediated by proinflammatory IL1 signaling. It may play a role in the limitation of proliferative response to tissue damage and prevention of wound-induced hyperplasia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects , Fibroblast Growth Factors/pharmacology , Interleukin-1/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Fibroblast Growth Factors/metabolism , Mice , NF-kappa B/metabolism
11.
Obesity (Silver Spring) ; 23(8): 1633-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26148471

ABSTRACT

OBJECTIVE: This study investigated the effects of loss of Cthrc1 on adipogenesis, body composition, metabolism, physical activity, and muscle physiology. METHODS: Complete metabolic and activity monitoring as well as grip strength measurements and muscle myography was performed in Cthrc1 null and wildtype mice. RESULTS: Compared to wildtypes, Cthrc1 null mice had similar body weights but significantly reduced energy expenditure, decreased lean mass, and increased fat mass, especially visceral fat. In vitro studies demonstrated that Cthrc1 inhibited adipocyte differentiation as well as PPAR and CREB reporter activity, while preadipocytes isolated from Cthrc1 null mice exhibited enhanced adipogenic differentiation. Voluntary physical activity in Cthrc1 null mice as assessed by wheel running was reduced to approximately half the distance covered by wildtypes. Reduced grip strength was observed in Cthrc1 null mice at the age of 15 weeks or older with reduced performance and mass of hyphenate muscle. In the brain, Cthrc1 expression was most prominent in neurons of thalamic and hypothalamic nuclei with evidence for secretion into the circulation in the median eminence. CONCLUSIONS: Our data indicate that Cthrc1 regulates body composition through inhibition of adipogenesis. In addition, central Cthrc1 may be a mediator of muscle function and physical activity.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/metabolism , Extracellular Matrix Proteins/chemistry , Motor Activity/physiology , Adipogenesis/physiology , Animals , Body Composition , Cell Differentiation , Male , Mice , Mice, Knockout
12.
J Cell Biochem ; 116(8): 1522-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25560297

ABSTRACT

FGF1 is a nonclassically released growth factor that regulates carcinogenesis, angiogenesis, and inflammation. In vitro and in vivo, FGF1 export is stimulated by cell stress. Upon stress, FGF1 is transported to the plasma membrane where it localizes prior to transmembrane translocation. To determine which proteins participate in the submembrane localization of FGF1 and its export, we used immunoprecipitation mass spectrometry to identify novel proteins that associate with FGF1 during heat shock. The heat shock-dependent association of FGF1 with the large protein AHNAK2 was observed. Heat shock induced the translocation of FGF1 and AHNAK2 to the cytoskeletal fraction. In heat-shocked cells, FGF1 and the C-terminal fragment of AHNAK2 colocalized with F-actin in the vicinity of the cell membrane. Depletion of AHNAK2 resulted in a drastic decrease of stress-induced FGF1 export but did not affect spontaneous FGF2 export and FGF1 release induced by the inhibition of Notch signaling. Thus, AHNAK2 is an important element of the FGF1 nonclassical export pathway.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Fibroblast Growth Factor 1/metabolism , Secretory Pathway , Stress, Physiological , Actins/metabolism , Animals , Cell Membrane/metabolism , Cytoskeletal Proteins/chemistry , Humans , Mass Spectrometry , Mice , NIH 3T3 Cells , Temperature
13.
J Cell Biochem ; 115(5): 874-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24375433

ABSTRACT

FGF applied as a single growth factor to quiescent mouse fibroblasts induces a round of DNA replication, however continuous stimulation results in arrest in the G1 phase of the next cell cycle. We hypothesized that FGF stimulation induces the establishment of cell memory, which prevents the proliferative response to repeated or continuous FGF application. When a 2-5 days quiescence period was introduced between primary and repeated FGF treatments, fibroblasts failed to efficiently replicate in response to secondary FGF application. The establishment of "FGF memory" during the first FGF stimulation did not require DNA synthesis, but was dependent on the activity of FGF receptors, MEK, p38 MAPK and NFκB signaling, and protein synthesis. While secondary stimulation resulted in strongly decreased replication rate, we did not observe any attenuation of morphological changes, Erk1/2 phosphorylation and cyclin D1 induction. However, secondary FGF stimulation failed to induce the expression of cyclin A, which is critical for the progression from G1 to S phase. Treatment of cells with a broad range histone deacetylase inhibitor during the primary FGF stimulation rescued the proliferative response to the secondary FGF treatment suggesting that the establishment of "FGF memory" may be based on epigenetic changes. We suggest that "FGF memory" can prevent the hyperplastic response to cell damage and inflammation, which are associated with an enhanced FGF production and secretion. "FGF memory" may present a natural obstacle to the efficient application of recombinant FGFs for the treatment of ulcers, ischemias, and wounds.


Subject(s)
Cell Cycle/genetics , Cell Movement/genetics , Cell Proliferation , Fibroblast Growth Factors/metabolism , Animals , Cyclin D1/genetics , DNA Replication/drug effects , DNA Replication/genetics , Fibroblast Growth Factors/administration & dosage , G1 Phase/genetics , Histone Deacetylases/metabolism , MAP Kinase Signaling System/genetics , Mice , NF-kappa B/metabolism , Signal Transduction/drug effects
14.
Mol Cancer Res ; 10(2): 230-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22172337

ABSTRACT

UNLABELLED: Prostate carcinoma is among the most common causes of cancer-related death in men, representing 15% of all male malignancies in developed countries. Neuroendocrine differentiation (NED) has been associated with tumor progression, poor prognosis, and with the androgen-independent status. Currently, no successful therapy exists for advanced, castration-resistant disease. Because hypoxia has been linked to prostate cancer progression and unfavorable outcome, we sought to determine whether hypoxia would impact the degree of neuroendocrine differentiation of prostate cancer cells in vitro. RESULTS: Exposure of LNCaP cells to low oxygen tension induced a neuroendocrine phenotype, associated with an increased expression of the transcription factor neurogenin3 and neuroendocrine markers, such as neuron-specific enolase, chromogranin A, and ß3-tubulin. Moreover, hypoxia triggered a significant decrease of Notch 1 and Notch 2 mRNA and protein expression, with subsequent downregulation of Notch-mediated signaling, as shown by reduced levels of the Notch target genes, Hes1 and Hey1. NED was promoted by attenuation of Hes1 transcription, as cells expressing a dominant-negative form of Hes1 displayed increased levels of neuroendocrine markers under normoxic conditions. Although hypoxia downregulated Notch 1 and Notch 2 mRNA transcription and receptor activation also in the androgen-independent cell lines, PC-3 and Du145, it did not change the extent of NED in these cultures, suggesting that androgen sensitivity may be required for transdifferentiation to occur. CONCLUSIONS: Hypoxia induces NED of LNCaP cells in vitro, which seems to be driven by the inhibition of Notch signaling with subsequent downregulation of Hes1 transcription.


Subject(s)
Androgens/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Nerve Tissue Proteins/genetics , Neuroendocrine Tumors/metabolism , Oxygen/administration & dosage , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Transcription Factor HES-1
15.
J Cell Physiol ; 226(11): 3064-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21302306

ABSTRACT

FGF1, a widely expressed proangiogenic factor involved in tissue repair and carcinogenesis, is released from cells through a non-classical pathway independent of endoplasmic reticulum and Golgi. Although several proteins participating in FGF1 export were identified, genetic mechanisms regulating this process remained obscure. We found that FGF1 export and expression are regulated through Notch signaling mediated by transcription factor CBF1 and its partner MAML. The expression of a dominant negative (dn) form of CBF1 in 3T3 cells induces transcription of FGF1 and sphingosine kinase 1 (SphK1), which is a component of FGF1 export pathway. dnCBF1 expression stimulates the stress-independent release of transduced FGF1 from NIH 3T3 cells and endogenous FGF1 from A375 melanoma cells. NIH 3T3 cells transfected with dnCBF1 form colonies in soft agar and produce rapidly growing highly angiogenic tumors in nude mice. The transformed phenotype of dnCBF1 transfected cells is efficiently blocked by dn forms of FGF receptor 1 and S100A13, which is a component of FGF1 export pathway. FGF1 export and acceleration of cell growth induced by dnCBF1 depend on SphK1. Similar to dnCBF1, dnMAML transfection induces FGF1 expression and release, and accelerates cell proliferation. The latter effect is strongly decreased in FGF1 null cells. We suggest that the regulation of FGF1 expression and release by CBF1-mediated Notch signaling can play an important role in tumor formation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblast Growth Factor 1/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , S100 Proteins/pharmacology , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection
16.
Mol Biol Cell ; 19(11): 4863-74, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18784255

ABSTRACT

Angiogenesis is controlled by several regulatory mechanisms, including the Notch and fibroblast growth factor (FGF) signaling pathways. FGF1, a prototype member of FGF family, lacks a signal peptide and is released through an endoplasmic reticulum-Golgi-independent mechanism. A soluble extracellular domain of the Notch ligand Jagged1 (sJ1) inhibits Notch signaling and induces FGF1 release. Thrombin, a key protease of the blood coagulation cascade and a potent inducer of angiogenesis, stimulates rapid FGF1 release through a mechanism dependent on the major thrombin receptor protease-activated receptor (PAR) 1. This study demonstrates that thrombin cleaves Jagged1 in its extracellular domain. The sJ1 form produced as a result of thrombin cleavage inhibits Notch-mediated CBF1/Suppressor of Hairless [(Su(H)]/Lag-1-dependent transcription and induces FGF1 expression and release. The overexpression of Jagged1 in PAR1 null cells results in a rapid thrombin-induced export of FGF1. These data demonstrate the existence of novel cross-talk between thrombin, FGF, and Notch signaling pathways, which play important roles in vascular formation and remodeling.


Subject(s)
Calcium-Binding Proteins/metabolism , Cardiovascular Agents/metabolism , Fibroblast Growth Factor 1/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Thrombin/pharmacology , Animals , Calcium-Binding Proteins/chemistry , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 1/genetics , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Jagged-1 Protein , Membrane Proteins/chemistry , Mice , Molecular Weight , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Neural Crest/cytology , Protein Structure, Tertiary , Protein Transport/drug effects , Receptor, PAR-1/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Notch/metabolism , Receptors, Thrombin , Serrate-Jagged Proteins , Signal Transduction/drug effects , Transcription, Genetic/drug effects
17.
Am J Pathol ; 173(3): 865-78, 2008 09.
Article in English | MEDLINE | ID: mdl-18688026

ABSTRACT

We previously found that soluble forms of the Notch ligands Jagged1 and Delta1 induced fibroblast growth factor receptor-dependent cell transformation in NIH3T3 fibroblasts. However, the phenotypes of these lines differed, indicating distinct functional differences among these Notch ligands. In the present study, we used allografts to test the hypothesis that NIH3T3 fibroblasts that express soluble forms of Delta1 and Jagged1 accelerate tumorigenicity in vivo. With the exception of the full-length Jagged1 transfectant, all other cell lines, including the control, generated tumors when injected subcutaneously in athymic mice. Suppression of Notch signaling by the soluble ligands significantly increased tumor onset and growth, whereas full-length Jagged1 completely suppressed tumor development. In addition, there were striking differences in tumor pathology with respect to growth kinetics, vascularization, collagen content, size and number of necrotic foci, and invasiveness into the underlying tissue. Further, the production of angiogenic factors, including vascular endothelial growth factor, also differed among the tumor types. Lastly, both Jagged1- and Delta1-derived tumors contained phenotypically distinct populations of lipid-filled cells that corresponded with increased expression of adipocyte markers. The divergence of tumor phenotype may be attributed to ligand-specific alterations in Notch receptor responses in exogenous and endogenous cell populations within the allographs. Our findings demonstrate distinct functional properties for these Notch ligands in the promotion of tumorigenicity in vivo.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neoplasms, Experimental/metabolism , Phenotype , Animals , Blotting, Western , Calcium-Binding Proteins/genetics , Cell Line , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Magnetic Resonance Imaging , Male , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Transfection
18.
Exp Cell Res ; 313(15): 3308-18, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17643421

ABSTRACT

Sphingosine kinase 1 catalyzes the formation of sphingosine-1-phosphate, a lipid mediator involved in the regulation of angiogenesis. Sphingosine kinase 1 is constitutively released from cells, even though it lacks a classical signal peptide sequence. Because copper-dependent non-classical stress-induced release of FGF1 also regulates angiogenesis, we questioned whether sphingosine kinase 1 is involved in the FGF1 release pathway. We report that (i) the coexpression of sphingosine kinase 1 with FGF1 inhibited the release of sphingosine kinase 1 at 37 degrees C; (ii) sphingosine kinase 1 was released at 42 degrees C in complex with FGF1; (iii) sphingosine kinase 1 null cells failed to release FGF1 at stress; (iv) sphingosine kinase 1 is a high affinity copper-binding protein which formed a complex with FGF1 in a cell-free system, and (v) sphingosine kinase 1 over expression rescued the release of FGF1 from inhibition by the copper chelator, tetrathiomolybdate. We propose that sphingosine kinase 1 is a component of the copper-dependent FGF1 release pathway.


Subject(s)
Copper/metabolism , Fibroblast Growth Factor 1/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cells, Cultured , Chelating Agents/pharmacology , Cloning, Molecular , Fibroblasts/metabolism , Mice , Mice, Knockout , Molybdenum/pharmacology , NIH 3T3 Cells , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Transport , Temperature
19.
Biochem Biophys Res Commun ; 360(3): 604-8, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17618910

ABSTRACT

Interleukin-1 alpha (IL-1alpha) regulates a wide range of important cellular processes. In this study for the first time, we report the cloning, expression, biophysical, and biological characterization of the human interleukin-1alpha. Human IL-1alpha has been expressed in Escherichia coli in high yields ( approximately 4mg per liter of the bacterial culture). The protein was purified to homogeneity ( approximately 98% purity) using affinity chromatography and size exclusion chromatography. Results of the steady-state fluorescence and 2D NMR experiments show that the recombinant IL-1alpha is in a folded conformation. Far-UV circular dichroism (CD) data suggest that IL-1alpha is an all beta-sheet protein with a beta-barrel architecture. Isothermal titration calorimetry (ITC) experiments show that the recombinant IL-1alpha binds strongly (K(d) approximately 5.6 x 10(-7) M) to S100A13, a calcium binding protein that chaperones the in vivo release of IL-1alpha into the extracellular compartment. Recombinant IL-1alpha was observed to exhibit strong cytostatic effect on human umbilical vascular endothelial cells. The findings of the present study not only pave way for an in-depth structural investigation of the molecular mechanism(s) underlying the non-classical release of IL-1alpha but also provide avenues for the rational design of potent inhibitors against IL-1alpha mediated pathogenesis.


Subject(s)
Interleukin-1alpha/biosynthesis , Interleukin-1alpha/genetics , Calorimetry/methods , Cell Proliferation/drug effects , Cells, Cultured , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/drug effects , Humans , Interleukin-1alpha/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , S100 Proteins/metabolism
20.
Biochem Biophys Res Commun ; 350(3): 604-9, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17027650

ABSTRACT

Thrombin induces cell proliferation and migration during vascular injury. We report that thrombin rapidly stimulated expression and release of the pro-angiogenic polypeptide fibroblast growth factor 1 (FGF1). Thrombin failed to induce FGF1 release from protease-activated receptor 1 (PAR1) null fibroblasts, indicating that this effect was dependent on PAR1. Similarly to thrombin, FGF1 expression and release were induced by TRAP, a specific oligopeptide agonist of PAR1. These results identify a novel aspect of the crosstalk between FGF and thrombin signaling pathways which both play important roles in tissue repair and angiogenesis.


Subject(s)
Fibroblast Growth Factor 1/metabolism , Receptor, PAR-1/metabolism , Signal Transduction/physiology , Thrombin/administration & dosage , Animals , Dose-Response Relationship, Drug , Mice , NIH 3T3 Cells , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...