Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
2.
J Biomark ; 2016: 1274603, 2016.
Article in English | MEDLINE | ID: mdl-27247826

ABSTRACT

Objective. Use of tyramide signal amplification (TSA) to detect autophagy biomarkers in formalin fixed and paraffin embedded (FFPE) xenograft tissue. Materials and Methods. Autophagy marker regulation was studied in xenograft tissues using Amp HQ IHC and standard IHC methods. Results. The data demonstrate the feasibility of using high sensitivity TSA IHC assays to measure low abundant autophagy markers in FFPE xenograft tissue.

3.
J Clin Invest ; 121(9): 3564-77, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21821913

ABSTRACT

IgE has a key role in the pathogenesis of allergic responses through its ability to activate mast cells via the receptor FcεR1. In addition to mast cells, many cell types implicated in atherogenesis express FcεR1, but whether IgE has a role in this disease has not been determined. Here, we demonstrate that serum IgE levels are elevated in patients with myocardial infarction or unstable angina pectoris. We found that IgE and the FcεR1 subunit FcεR1α were present in human atherosclerotic lesions and that they localized particularly to macrophage-rich areas. In mice, absence of FcεR1α reduced inflammation and apoptosis in atherosclerotic plaques and reduced the burden of disease. In cultured macrophages, the presence of TLR4 was required for FcεR1 activity. IgE stimulated the interaction between FcεR1 and TLR4, thereby inducing macrophage signal transduction, inflammatory molecule expression, and apoptosis. These IgE activities were reduced in the absence of FcεR1 or TLR4. Furthermore, IgE activated macrophages by enhancing Na+/H+ exchanger 1 (NHE1) activity. Inactivation of NHE1 blocked IgE-induced macrophage production of inflammatory molecules and apoptosis. Cultured human aortic SMCs (HuSMCs) and ECs also exhibited IgE-induced signal transduction, cytokine expression, and apoptosis. In human atherosclerotic lesions, SMCs and ECs colocalized with IgE and TUNEL staining. This study reveals what we believe to be several previously unrecognized IgE activities that affect arterial cell biology and likely other IgE-associated pathologies in human diseases.


Subject(s)
Apolipoproteins E/metabolism , Apoptosis/physiology , Atherosclerosis/physiopathology , Cytokines/metabolism , Immunoglobulin E/metabolism , Angina Pectoris/blood , Angina Pectoris/immunology , Angina Pectoris/pathology , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Cells, Cultured , China , Dietary Fats/adverse effects , Humans , Macrophages/cytology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Plaque, Atherosclerotic/chemistry , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Receptors, IgE/genetics , Receptors, IgE/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
4.
BMC Cancer ; 11: 331, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21812995

ABSTRACT

BACKGROUND: Hypoxia has been identified as a major negative factor for tumor progression in clinical observations and in animal studies. However, the precise role of hypoxia in tumor progression has not been fully explained. In this study, we extensively investigated the effect of long-term exposure to hypoxia on tumor progression in vivo. METHODS: Rats bearing transplanted tumors consisting of A549 human lung cancer cells (lung cancer tumor) were exposed to hypoxia for different durations and different levels of oxygen. The tumor growth and metastasis were evaluated. We also treated A549 lung cancer cells (A549 cells) with chronic hypoxia and then implanted the hypoxia-pretreated cancer cells into mice. The effect of exposure to hypoxia on metastasis of Lewis lung carcinoma in mice was also investigated. RESULTS: We found that long-term exposure to hypoxia a) significantly inhibited lung cancer tumor growth in xenograft and orthotopic models in rats, b) significantly reduced lymphatic metastasis of the lung cancer in rats and decreased lung metastasis of Lewis lung carcinoma in mice, c) reduced lung cancer cell proliferation and cell cycle progression in vitro, d) decreased growth of the tumors from hypoxia-pretreated A549 cells, e) decreased Na+-K+ ATPase α1 expression in hypoxic lung cancer tumors, and f) increased expression of hypoxia inducible factors (HIF1α and HIF2α) but decreased microvessel density in the lung cancer tumors. In contrast to lung cancer, the growth of tumor from HCT116 human colon cancer cells (colon cancer tumor) was a) significantly enhanced in the same hypoxia conditions, accompanied by b) no significant change in expression of Na+-K+ ATPase α1, c) increased HIF1α expression (no HIF2α was detected) and d) increased microvessel density in the tumor tissues. CONCLUSIONS: This study demonstrated that long-term exposure to hypoxia repressed tumor progression of the lung cancer from A549 cells and that decreased expression of Na+-K+ ATPase was involved in hypoxic inhibition of tumor progression. The results from this study provide new insights into the role of hypoxia in tumor progression and therapeutic strategies for cancer treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Hypoxia , Lung Neoplasms/pathology , Neoplasms, Experimental/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Hypoxia , Cell Line, Tumor , Disease Progression , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats , Rats, Nude , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors , Transplantation, Heterologous
5.
Glycoconj J ; 28(6): 419-26, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21773727

ABSTRACT

Heparin (HP) inhibits the growth of several cell types in vitro including bovine pulmonary artery (BPA) smooth muscle cells (SMCs). In initial studies we discovered that an O-hexanoylated low-molecular-weight (LMW) HP derivative having acyl groups with 6-carbon chain length was more potent inhibitor of BPA-SMCs than the starting HP. We prepared several O-acylated LMWHP derivatives having 4-, 6-, 8-, 10-, 12-, and 18- carbon acyl chain lengths to determine the optimal acyl chain length for maximum anti-proliferative properties of BPA-SMCs. The starting LMWHP was prepared from unfractionated HP by sodium periodate treatment followed by sodium borohydride reduction. The tri-n-butylammonium salt of this LMWHP was O-acylated with butanoic, hexanoic, octanoic, decanoic, dodecanoic, and stearyl anhydrides separately to give respective O-acylated LMWHP derivatives. Gradient polyacrylamide gel electrophoresis (PAGE) was used to examine the average molecular weights of those O-acylated LMWHP derivatives. NMR analysis indicated the presence of one O-acyl group per disaccharide residue. Measurement of the inhibition of BPA-SMCS as a function of O-acyl chain length shows two optima, at a carbon chain length of 6 (O-hexanoylated LMWHP) and at a carbon chain length 12-18 (O-dodecanoyl and O-stearyl LMWHPs). A solution competition SPR study was performed to test the ability of different O-acylated LMWHP derivatives to inhibit fibroblast growth factor (FGF) 1 and FGF2 binding to surface-immobilized heparin. All the LMWHP derivatives bound to FGF1 and FGF2 but each exhibited slightly different binding affinity.


Subject(s)
Fibrinolytic Agents/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/cytology , Animals , Carbohydrate Sequence , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Fibrinolytic Agents/chemistry , Heparin, Low-Molecular-Weight/chemistry , Inhibitory Concentration 50 , Molecular Sequence Data , Molecular Structure , Pulmonary Artery/drug effects
6.
J Vasc Res ; 48(6): 465-75, 2011.
Article in English | MEDLINE | ID: mdl-21691120

ABSTRACT

BACKGROUND: Hypoxia results in pulmonary hypertension and vascular remodeling due to induction of pulmonary artery cell proliferation. Besides pulmonary artery smooth muscle cells, pulmonary artery endothelial cells (PAECs) are also involved in the development of pulmonary hypertension, but the effect of hypoxia on PAEC proliferation has not been completely understood. METHODS: We investigated PAEC proliferation in mice and rats with hypoxia-induced pulmonary hypertension and vascular remodeling as well as in human PAECs under hypoxia. RESULTS AND CONCLUSION: We did not find significant PAEC proliferation in chronically hypoxic rats or mice. There was a slight decrease in proliferation in mice and rats with pulmonary hypertension and vascular remodeling. We also did not find significant human PAEC proliferation and cell cycle progression under different levels of oxygen (1, 2, 3, 5 and 10%) for one day, although the same conditions of hypoxia induced significant proliferation and cell cycle progression in pulmonary artery smooth muscle cells and pulmonary artery fibroblasts. Exposure to hypoxia for 7 days also did not increase PAEC proliferation. These results demonstrated that hypoxia alone is not a stimulus to PAEC proliferation in vivo and in vitro. The present study provides a novel role for PAECs in hypoxia-induced pulmonary hypertension and vascular remodeling.


Subject(s)
Endothelial Cells/cytology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Pulmonary Artery/cytology , Pulmonary Circulation/physiology , Animals , Cell Cycle/physiology , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Endothelial Cells/physiology , Humans , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley
7.
Am J Respir Cell Mol Biol ; 45(5): 923-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21454803

ABSTRACT

We previously found that deficiency of the sodium-hydrogen exchanger 1 (NHE1) gene prevented hypoxia-induced pulmonary hypertension and vascular remodeling in mice, which were accompanied by a significantly reduced proliferation of pulmonary artery smooth muscle cells (PASMCs), and which decreased the medial-wall thickness of pulmonary arteries. That finding indicated the involvement of NHE1 in the proliferation and hypertrophy of PASMCs, but the underlying mechanism was not fully understood. To define the mechanism by which the inhibition of NHE1 decreases hypoxic pulmonary hypertension and vascular remodeling, we investigated the role of E2F1, a nuclear transcription factor, in silencing the NHE1 gene-induced inhibition of the proliferation, hypertrophy, and migration of human PASMCs. We found that: (1) silencing of NHE1 by short, interfering RNA (siRNA) significantly inhibited PASMC proliferation and cell cycle progression, decreased hypoxia-induced hypertrophy (in terms of cell size and protein/DNA ratio) and migration (in terms of the wound-healing and migration chamber assays); (2) hypoxia induced the expression of E2F1, which was reversed by NHE1 siRNA; and (3) the overexpression of E2F1 blocked the inhibitory effect of NHE1 siRNA on the proliferation, hypertrophy, and migration of PASMCs. The present study determined that silencing the NHE1 gene significantly inhibited the hypoxia-induced proliferation, hypertrophy, and migration of human PASMCs via repression of the nuclear transcription factor E2F1. This study revealed a novel mechanism underlying the regulation of hypoxic pulmonary hypertension and vascular remodeling via NHE1.


Subject(s)
Cation Transport Proteins/metabolism , E2F1 Transcription Factor/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Sodium-Hydrogen Exchangers/metabolism , Cation Transport Proteins/genetics , Cell Hypoxia , Cell Line , Cell Movement , Cell Proliferation , Cell Size , Gene Silencing , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , RNA, Small Interfering/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics
8.
Respir Res ; 12: 21, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21294880

ABSTRACT

BACKGROUND: CXCR4 is the receptor for chemokine CXCL12 and reportedly plays an important role in systemic vascular repair and remodeling, but the role of CXCR4 in development of pulmonary hypertension and vascular remodeling has not been fully understood. METHODS: In this study we investigated the role of CXCR4 in the development of pulmonary hypertension and vascular remodeling by using a CXCR4 inhibitor AMD3100 and by electroporation of CXCR4 shRNA into bone marrow cells and then transplantation of the bone marrow cells into rats. RESULTS: We found that the CXCR4 inhibitor significantly decreased chronic hypoxia-induced pulmonary hypertension and vascular remodeling in rats and, most importantly, we found that the rats that were transplanted with the bone marrow cells electroporated with CXCR4 shRNA had significantly lower mean pulmonary pressure (mPAP), ratio of right ventricular weight to left ventricular plus septal weight (RV/(LV+S)) and wall thickness of pulmonary artery induced by chronic hypoxia as compared with control rats. CONCLUSIONS: The hypothesis that CXCR4 is critical in hypoxic pulmonary hypertension in rats has been demonstrated. The present study not only has shown an inhibitory effect caused by systemic inhibition of CXCR4 activity on pulmonary hypertension, but more importantly also has revealed that specific inhibition of the CXCR4 in bone marrow cells can reduce pulmonary hypertension and vascular remodeling via decreasing bone marrow derived cell recruitment to the lung in hypoxia. This study suggests a novel therapeutic approach for pulmonary hypertension by inhibiting bone marrow derived cell recruitment.


Subject(s)
Blood Pressure , Hypertension, Pulmonary/immunology , Hypoxia/immunology , Pulmonary Artery/immunology , Receptors, CXCR4/metabolism , Animals , Benzylamines , Blood Pressure/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Movement , Cyclams , Disease Models, Animal , Electroporation , Green Fluorescent Proteins/genetics , Heterocyclic Compounds/pharmacology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/drug therapy , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/physiopathology , Male , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , RNA Interference , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Stem Cells/drug effects , Stem Cells/immunology , Transfection
9.
Am J Respir Cell Mol Biol ; 44(4): 524-30, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20558775

ABSTRACT

Ras homolog gene family member A (RhoA) through Rho kinase kinase (ROCK), one of its downstream effectors, regulates a wide range of cell physiological functions, including vascular smooth muscle cell (SMC) proliferation, by degrading cyclin-dependent kinase inhibitor, p27. Our previous studies found that heparin inhibition of pulmonary artery SMC (PASMC) proliferation and pulmonary hypertension was dependent on p27 up-regulation. To investigate whether ROCK, a regulator of p27, is involved in regulation of heparin inhibition of PASMC proliferation, we analyzed ROCK expression in the lungs from mice and from human PASMCs exposed to hypoxia, and investigated the effect of ROCK expression in vitro by RhoA cDNA transfection. We also investigated the effect of guanine nucleotide exchange factor (GEF)-H1, an upstream regulator of RhoA, on heparin inhibition of PASMC proliferation by GEF-H1 cDNA transfection. We found that: (1) hypoxia increased ROCK expression in mice and PASMCs; (2) overexpression of RhoA diminished the inhibitory effect of heparin on PASMC proliferation and down-regulated p27 expression; and (3) overexpression of GEF-H1 negated heparin inhibition of PASMC proliferation, which was accompanied by increased GTP-RhoA and decreased p27. This study demonstrates that the RhoA/ROCK pathway plays an important role in heparin inhibition on PASMC proliferation, and reveals that heparin inhibits PASMC proliferation through GEF-H1/RhoA/ROCK/p27 signaling pathway, by down-regulating GEF-H1, RhoA, and ROCK, and then up-regulating p27.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Heparin/pharmacology , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Cycle/drug effects , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanosine Triphosphate/metabolism , Humans , Mice , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , RNA, Small Interfering/metabolism , Rho Guanine Nucleotide Exchange Factors
10.
Respirology ; 15(7): 1131-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20874749

ABSTRACT

BACKGROUND AND OBJECTIVE: High MW hyaluronan (HMW HA) as opposed to low MW hyaluronan (LMW HA) has been shown to have anti-inflammatory and anti-apoptotic effects. We hypothesized that treatment with HMW HA would block smoke inhalation lung injury by inhibiting smoke-induced lung inflammation and airway epithelial cell apoptosis. METHODS: Anesthetized, intubated male rats were randomly allocated to either control or smoke inhalation injury groups. Rats were treated with 3-mL subcutaneous normal saline solution (sham) or LMW HA (35 kDa) or HMW HA (1600 kDa) 18 h before exposure to 15 min of cotton smoke (n = 5 each). Rats were also treated post smoke inhalation with 1600 kDa HA by intra-peritoneal injection (3 mL) or intra-tracheal nebulization (200 µL). Lung neutrophil infiltration, airway apoptosis, airway mucous plugging and lung injury were assessed 4 h after smoke inhalation injury. RESULTS: Rats pretreated with 1600 kDa HA had significantly less smoke-induced neutrophil infiltration, lung oedema, airway apoptosis and mucous plugging. Pretreatment with 35 kDa HA, in contrast, increased smoke-induced neutrophil infiltration and lung injury score. Intra-tracheal administration of a single dose 1600 kDa HA, but not intra-peritoneal injection, significantly improved survival post smoke inhalation. CONCLUSIONS: High MW hyaluronan (1600 kDa) may prove to be a beneficial therapy for smoke inhalation through inhibition of smoke-induced inflammation, lung oedema, airway epithelial cell apoptosis and airway mucous plugging.


Subject(s)
Hyaluronic Acid/therapeutic use , Smoke Inhalation Injury/drug therapy , Animals , Apoptosis/drug effects , Lung/drug effects , Lung/immunology , Lung Injury/drug therapy , Male , Mucus/drug effects , Mucus/immunology , Neutrophil Infiltration/drug effects , Pneumonia/drug therapy , Pulmonary Edema/drug therapy , Rats , Rats, Sprague-Dawley
11.
J Cardiothorac Surg ; 5: 32, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20441584

ABSTRACT

BACKGROUND AND OBJECTIVE: Chronic hypoxia induces pulmonary hypertension in mice. Smooth muscle cell hyperplasia and medial thickening characterize the vasculature of these animals. Thrombospondin-1 null (TSP-1(-/-)) mice spontaneously develop pulmonary smooth muscle cell hyperplasia and medial thickening. In addition, TSP-1 produced by the pulmonary endothelium inhibits pulmonary artery smooth muscle cell growth. Based on these observations we sought to describe the pulmonary vascular changes in TSP-1(-/-) mice exposed to chronic hypoxia. METHODS: We exposed TSP-1(-/-) and wild type (WT) mice to a fraction of inspired oxygen (FiO2) of 0.1 for up to six weeks. Pulmonary vascular remodeling was evaluated using tissue morphometrics. Additionally, right ventricle systolic pressures (RVSP) and right ventricular hypertrophy by right ventricle/left ventricle + septum ratios (RV/LV+S) were measured to evaluate pulmonary hypertensive changes. Finally, acute pulmonary vasoconstriction response in both TSP-1(-/-) and WT mice was evaluated by acute hypoxia and U-46619 (a prostaglandin F2 analog) response. RESULTS: In hypoxia, TSP-1(-/-) mice had significantly lower RVSP, RV/LV+S ratios and less pulmonary vascular remodeling when compared to WT mice. TSP-1(-/-) mice also had significantly lower RVSP in response to acute pulmonary vasoconstriction challenges than their WT counterparts. CONCLUSION: TSP-1(-/-) mice had diminished pulmonary vasoconstriction response and were less responsive to hypoxia-induced pulmonary hypertension than their wild type counterparts. This observation suggests that TSP-1 could play an active role in the pathogenesis of pulmonary hypertension associated with hypoxia.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Thrombospondin 1/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/physiopathology , Thrombospondin 1/genetics , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Ventricular Function, Right
12.
Carbohydr Res ; 345(9): 1084-7, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20399420

ABSTRACT

Heparin (HP) inhibits the proliferation of bovine pulmonary artery smooth muscle cells (BPASMC's), among other cell types in vitro. In order to develop a potential therapeutic agent to reverse vascular remodeling, we are involved in deciphering the relationship between the native HP structure and its antiproliferative potency. We have previously reported the influence of the molecular size and the effects of various O-sulfo and N-acetyl groups of HP on growth-inhibitory activity. In this study, to understand the influence of carboxyl groups in the HP structure required for endogenous activity, a chemically modified derivative of native HP was prepared by converting the carboxyl groups of hexuronic acid residues in HP to primary hydroxyl groups. This modification procedure involves the treatment of HP with N-(3-dimethylaminopropyl)-N-ethylcarbodiimide followed by reduction with NaBH(4) to yield carboxyl-reduced heparin (CR-HP). When compared to the antiproliferative potency of native HP on cultured BPASMC's at three dose levels (1, 10, and 100 microg/mL), the CR-HP showed significantly less potency at all the doses. These results suggest that hexuronic acid residues in both major and variable sequences in HP are essential for the antiproliferative properties of native HP.


Subject(s)
Heparin/chemistry , Heparin/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/cytology , Animals , Borohydrides/chemistry , Cattle , Cell Proliferation/drug effects , Hexuronic Acids/chemistry , Hydroxides/chemistry , Oxidation-Reduction
13.
Curr Cancer Drug Targets ; 10(2): 229-41, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20201787

ABSTRACT

Whole unfractionated heparin can modestly decrease tumor growth, but the dose of heparin is limited by its anticoagulant properties. To overcome this limitation, we modified the chemical structure of heparin and have prepared a heparin derivative by O-acylating low molecular weight heparin with butyric anhydride, producing a more potent antiproliferative compound, which is only weakly anticoagulant so that the dose may be escalated without threat of hemorrhage. In this study, we investigated the effect of this chemically modified heparin, butanoylated heparin, on the growth of lung cancer in vitro and in vivo. We found that butanoylated heparin a) significantly inhibited lung cancer cell proliferation in vitro and lung cancer growth in mice and rats; b) had very low anticoagulant effect; c) had no significant toxicity on heart, liver, kidney and lung; d) significantly although modestly induced apoptosis and decreased expression of the cell proliferation pathway consisting of mutant p53, phospho-Rb and E2F1 expression in the tumor tissues. We also found that butanoylated heparin significantly inhibited CXCL12 and CXCR4 expression, suggesting that CXCL12/CXCR4 axis may be involved in regulation of tumor growth inhibition by heparin. We concluded that chemically modified butanoylated heparin has potent antiproliferative activity against lung cancer and may represent a new chemical therapeutic agent for cancer patients.


Subject(s)
Anticoagulants/pharmacology , Antineoplastic Agents/pharmacology , Butanols/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Heparin/pharmacology , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Rats , Rats, Nude , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Cells, Cultured
14.
Zhonghua Bing Li Xue Za Zhi ; 37(5): 300-4, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18956646

ABSTRACT

OBJECTIVE: To study the methylation of CpG islands in the promoter region, expression of caveolin 1 (Cav-1) gene and their clinical significance in non-small cell lung cancers (NSCLC). METHODS: Immunohistochemistry and quanta Qd600 staining were used to detect the expression of Cav-1 in tissues from benign lung lesions (n = 17) and NSCLC (n = 123). DNA was treated with sodium bisulfite and the Cav-1 promoter region was screened using methylation-specific polymerase chain reaction for the possible methylation sites. RESULTS: Cav-1 protein was highly expressed in cytoplasm and cell membrane of normal bronchial epithelium, alveolar epithelium, endothelial cells, fibroblasts and smooth muscle cells. The expression rates of Cav-1 protein were 100% (17/17) in the control group and 43.1% (53/123) in the NSCLC group (P = 0.001). Amongst the NSCLC group, there was no statistically significant difference in Cav-1 protein expression in different histologic types (P = 0.552) and tumor grades (P = 0.160). On the other hand, Cav-1 protein immunoreactivity was remarkably higher in advanced tumor stage: 72.7% in stage III A + III B, compared with 9.4% in stage I A + I B and 38.3% in stage II A + II B (P = 0.001). The expression rate of Cav-1 protein in the NSCLC cases with lymph node metastasis was 53.6%, compared with 20.5% in those without nodal involvement (P = 0.001). DNA from 40 NSCLC cases with negative Cav-1 protein expression and 12 cases of peritumoral lung tissues were extracted. Methylation in the promoter region of Cav-1 gene was not detected in lung cancer or peritumoral tissues. CONCLUSIONS: High expression of Cav-1 protein is respected of the aggressive clinical behavior and advanced tumor stage. Loss of Cav-1 protein expression seems not correlated to the methylation status in the promoter region of Cav-1 gene.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Caveolin 1/metabolism , Gene Regulatory Networks/genetics , Lung Neoplasms/genetics , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Caveolin 1/genetics , DNA Methylation , Female , Humans , Immunohistochemistry , Lung Neoplasms/classification , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging/methods , Somatoform Disorders
15.
Carbohydr Res ; 343(14): 2406-10, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18656855

ABSTRACT

Heparin inhibits the growth of several cell types in vitro, including bovine pulmonary artery smooth muscle cells (BPASMCs). To understand more about the heparin structure required for endogenous activity, chemically modified derivatives of native heparin and glycol-split heparin, namely, 2-O-desulfonated iduronic/glucuronic acid residues in heparin, and 2-O-desulfonated iduronic residues in glycol-split heparin were prepared. These were assayed for their antiproliferative potency on cultured BPASMCs. All of the 2-O-desulfonated heparin derivatives had significantly decreased less antiproliferative activity on BPASMCs. These results suggest that the 2-O-sulfo group of iduronic acid residues in heparin's major sequence is essential for the antiproliferative properties of heparin. The size of heparin does not affect the growth-inhibitory properties of heparin on BPASMCs at the three dose levels examined.


Subject(s)
Heparin/chemistry , Heparin/pharmacology , Iduronic Acid/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Pulmonary Artery/cytology , Sulfur/chemistry , Animals , Carbohydrate Sequence , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Heparin/analogs & derivatives , Molecular Sequence Data
16.
Am J Respir Crit Care Med ; 177(11): 1276-84, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18310478

ABSTRACT

RATIONALE: Our previous studies found that Na(+)/H(+) exchanger (NHE) activity played an essential role in pulmonary artery smooth muscle cell (PASMC) proliferation and in the development of hypoxia-induced pulmonary hypertension and vascular remodeling. Other investigators recently observed increased expression of the NHE isoform 1 (NHE1) gene in rodents with pulmonary hypertension induced by hypoxia. However, a causal role for the NHE1 gene in pulmonary hypertension has not been determined. OBJECTIVES: To determine the causal role of the NHE1 gene in pulmonary hypertension and vascular remodeling. METHODS: We used NHE1-null mice to define the role of the NHE1 gene in the development of pulmonary hypertension and remodeling induced by hypoxia and to delineate the NHE1 regulatory pathway. MEASUREMENTS AND MAIN RESULTS: After 2 weeks of exposure to hypoxia, in contrast to wild-type hypoxic littermates, there was no significant increase in right ventricular systolic pressure, in the ratio of right ventricular to left ventricular plus septal weight [RV/(LV + S)], or in medial wall thickness of the pulmonary arterioles in homozygous mice (NHE1(-/-)). There was a significant decrease in Rho kinase (ROCK1 and ROCK2) expression, accompanied by an increase in p27 expression in NHE1(-/-) mice. CONCLUSIONS: Our study demonstrated that deficiency of the NHE1 gene prevented the development of hypoxia-induced pulmonary hypertension and vascular remodeling in mice and revealed a novel regulatory pathway associated with NHE1 signaling.


Subject(s)
Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Sodium-Hydrogen Exchangers/genetics , Animals , Disease Models, Animal , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Sodium-Hydrogen Exchanger 1 , Vascular Resistance/physiology , Ventricular Pressure/physiology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
17.
Chest ; 132(6): 1898-905, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18079223

ABSTRACT

RATIONALE: We have shown previously that antiproliferative unfractionated heparins block hypoxia-induced pulmonary arterial hypertension (PAH) and vascular remodeling, and hypothesized that low-molecular-weight heparins (LMWHs) would too. OBJECTIVES: To determine the potential role and mechanisms of dalteparin and enoxaparin (two LMWHs) in inhibiting hypoxic PAH and vascular remodeling. METHODS: Male Hartley guinea pigs were exposed for 10 days to normobaric 10% oxygen with dalteparin (5 mg/kg), enoxaparin (5 mg/kg), or with an equivalent volume of normal saline solution. Normoxic control animals (n = 5) received room air for 10 days. Bovine pulmonary artery smooth-muscle cells (PASMCs) were grown in 10% fetal bovine serum without heparin, with dalteparin (1 microg/mL) or with enoxaparin (1 microg/mL). MEASUREMENTS: Pulmonary arterial pressure (PAP), cardiac index, right ventricular heart weight divided by left ventricular plus septum weight (RV/LV+S), hematocrit, percentage of wall thickness of intraacinar vessels (%WT-IA), percentage of wall thickness of terminal bronchiole vessels (%WT-TA), and the percentage of thick-walled vessels (%Thick) were determined. In PASMCs, expression of p27 and cell growth were compared because in mice whole heparin depends on p27 for its antiproliferative action. MAIN RESULTS: In hypoxic animals, hematocrit, PAP, total pulmonary vascular resistance index, RV/LV+S, %WT-IA, %WT-TA, and %Thick all rose significantly vs normoxic control animals (p < 0.05); cardiac index was unchanged. Dalteparin but not enoxaparin significantly reduced PAP, total pulmonary vascular resistance index, and RV/LV + S (p < 0.05 vs hypoxia alone); inhibited PASMC growth; and upregulated p27 expression. Enoxaparin moderately reduced vascular remodeling, which did not translate into less pulmonary hypertension. CONCLUSIONS: Not all LMWHs are the same. Dalteparin was more effective than enoxaparin in inhibiting pulmonary hypertension and vascular remodeling in hypoxic guinea pigs.


Subject(s)
Heparin, Low-Molecular-Weight/pharmacology , Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Muscle, Smooth, Vascular/drug effects , Analysis of Variance , Animals , Cattle , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Guinea Pigs , Hemodynamics/drug effects , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Pulmonary Artery/drug effects
18.
Carbohydr Res ; 341(15): 2607-12, 2006 Nov 06.
Article in English | MEDLINE | ID: mdl-16920092

ABSTRACT

Proliferation of pulmonary artery smooth muscle cells (PASMCs) appears to play a significant role in chronic pulmonary hypertension. The proliferation of PASMCs is strongly inhibited by some commercial heparin preparations. Heparin fragments were prepared by periodate treatment, followed by sodium borohydride reduction, to enhance potency. The tributylammonium salt of this fragmented heparin was O-acylated with hexanoic anhydride. Gradient polyacrylamide gel electrophoresis showed that the major heparin fragment contained eight disaccharide units. NMR analysis showed that approximately one hexanoyl group per disaccharide residue was present. The O-hexanoyl heparin fragments were assayed for growth inhibitory effect on bovine PASMCs in culture. This derivative was found to be more effective in growth inhibition of bovine PASMCs in culture than the heparin from which it was derived. In the future, it is envisioned that this or similar derivatives may be an effective treatment for pulmonary hypertension.


Subject(s)
Heparin, Low-Molecular-Weight/chemistry , Heparin, Low-Molecular-Weight/pharmacology , Muscle, Smooth, Vascular/cytology , Pulmonary Artery/cytology , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Cattle , Cell Division/drug effects , Hypertension, Pulmonary , Models, Molecular , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Pulmonary Artery/drug effects
19.
Biochem Biophys Res Commun ; 345(4): 1565-72, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16729969

ABSTRACT

The balance between cell proliferation and cell quiescence is regulated delicately by a variety of mediators, in which cyclin-dependent kinases (CDK) and CDK inhibitors (CDKI) play a very important role. Heparin which inhibits pulmonary artery smooth muscle cell (PASMC) proliferation increases the levels of two CDKIs, p21 and p27, although only p27 is important in inhibition of PASMC growth in vitro and in vivo. In the present study we investigated the expression profile of all the cell cycle regulating genes, including all seven CDKIs (p21, p27, p57, p15, p16, p18, and p19), in the lungs of mice with hypoxia-induced pulmonary hypertension. A cell cycle pathway specific gene microarray was used to profile the 96 genes involved in cell cycle regulation. We also observed the effect of heparin on gene expression. We found that (a) hypoxic exposure for two weeks significantly inhibited p27 expression and stimulated p18 activity, showing a 98% decrease in p27 and 81% increase in p18; (b) other CDKIs, p21, p57, p15, p16, and p19 were not affected significantly in response to hypoxia; (c) heparin treatment restored p27 expression, but did not influence p18; (d) ERK1/2 and p38 were mediators in heparin upregulation of p27. This study provides an expression profile of cell cycle regulating genes under hypoxia in mice with hypoxia-induced pulmonary hypertension and strengthens the previous finding that p27 is the only CDKI involved in heparin regulation of PASMC proliferation and hypoxia-induced pulmonary hypertension.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Gene Expression Profiling , Gene Expression/drug effects , Heparin/pharmacology , Hypertension, Pulmonary/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cyclin D2 , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclins/genetics , E2F4 Transcription Factor/genetics , Flavonoids/pharmacology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Imidazoles/pharmacology , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Pyridines/pharmacology , Retinoblastoma-Like Protein p107/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
20.
Circ Res ; 97(9): 937-45, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16195480

ABSTRACT

Heparin has growth inhibitory effects on pulmonary artery smooth muscle cell (PASMC) in vitro and in vivo. However, the mechanism has not been fully defined. In this study, we investigated the role of cyclin-dependent kinase inhibitors, p21(WAF1/cip1) (p21) and p27Kip1 (p27), in the inhibitory effect of heparin on PASMC proliferation in vitro and on hypoxia-induced pulmonary hypertension in vivo using p21 and p27-null mice. In vitro, loss of the p27 gene negated the inhibitory effect of heparin on PASMC proliferation, but p21 was not critical for this inhibition. In vivo, heparin significantly inhibited the development of hypoxia-induced pulmonary hypertension and remodeling, as evidenced by decreased right ventricular systolic pressure, ratio of right ventricular weight to left ventricle plus septum weight, and percent wall thickness of pulmonary artery, in p21(+/+), p21(-/-), p27(+/+), and p27(+/-), but not in p27(-/-) mice. We also observed that hypoxia decreased p27 expression significantly in mouse lung, which was restored by heparin. Heparin inhibited Ki67 proliferative index in terminal bronchial vessel walls in p27(+/+) and p27(+/-), but not in p27(-/-) mice exposed to hypoxia. Therefore, we conclude that the cyclin-dependent kinase inhibitor p27, but not p21, is required for the inhibition of hypoxic pulmonary vascular remodeling by heparin.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Heparin/pharmacology , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Pulmonary Artery/drug effects , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Hyperplasia , Ki-67 Antigen/analysis , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...