Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Amyloid ; 20(1): 34-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23339420

ABSTRACT

The polyphenol, 1,2,3,4,6-penta-O-galloyl-ß-D-glucose (PGG) has been found to exhibit a host of positive pharmacologic activities, including anti-cancer and anti-diabetic. Little is known about the mode of action of PGG in yielding these positive activities. We show here that PGG is a potent inhibitor of IAPP (islet amyloid polypeptide, amylin) aggregation. Preventing the initial aggregation event of IAPP is one strategy for slowing, and possibly preventing, the toxic effects of IAPP oligomeric intermediates. Equal molar ratios of PGG to IAPP substantially reduced the ability of IAPP to bind thioflavin T. Atomic force microscopy revealed that PGG prevented amyloid-based fiber formation under rigorous conditions conducive to forming IAPP aggregates. PGG was also found to protect PC12 rat cells from toxic IAPP. PGG was compared to the known amyloid inhibitors (and structural relatives); tannic acid and gallic acid. In every test, PGG was far superior to tannic and gallic acids at inhibiting amyloid aggregation. These results indicate that PGG is a potent inhibitor of IAPP amyloid aggregation and a potential lead molecule for development of an amyloid inhibiting therapeutic.


Subject(s)
Hydrolyzable Tannins/pharmacology , Islet Amyloid Polypeptide/pharmacology , Animals , Benzothiazoles , Cell Survival/drug effects , Gallic Acid/pharmacology , Humans , Islet Amyloid Polypeptide/antagonists & inhibitors , Microscopy, Atomic Force , PC12 Cells , Rats , Spectrometry, Fluorescence , Tannins/pharmacology , Thiazoles
3.
Open Biotechnol J ; 5: 39-46, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-24653784

ABSTRACT

The misfolding and aggregation of proteins into amyloid has been linked to a variety of age-related diseases. Aggregation of proteins, such as Aß in Alzheimer's disease and Islet Amyloid Polypeptide (IAPP, amylin) in type 2 diabetes, appears to lead to the formation of toxic assemblies. These assemblies range in size from small oligomers (2-8 proteins) to large fibrils (thousands of proteins). It remains unclear how these amyloidogenic proteins misfold and form toxic species, but growing evidence suggests that inhibiting the aggregation of these proteins could slow, if not prevent altogether, the progression of these diseases. We describe the use of small peptides (<43 amino acids) as inhibitors of amyloid-based aggregation. These peptides, often short complementary segments of the amyloid proteins, can be useful (i) for identifying the aggregation-prone regions of the amyloid proteins (ii) as models for drug discovery and (iii) as potential therapeutic agents themselves.

4.
Cancer Res ; 67(13): 6417-24, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616702

ABSTRACT

Idiopathic myelofibrosis (IM) is likely the consequence of both the acquisition of genetic mutations and epigenetic changes that silence critical genes that control cell proliferation, differentiation, and apoptosis. We have explored the effects of the sequential treatment with the DNA methyltransferase inhibitor, decitabine [5-aza-2'-deoxycytidine (5azaD)], followed by the histone deacetylase inhibitor, trichostatin A (TSA), on the behavior of IM CD34(+) cells. Unlike normal CD34(+) cells where 5azaD/TSA treatment leads to the expansion of CD34(+) cells and marrow-repopulating cells, treatment of IM CD34(+) cells results in a reduction of the number of total cells, CD34(+) cells, and assayable hematopoietic progenitor cells (HPC). In IM, HPCs are either heterozygous or homozygous for the JAK2V617F mutation or possess wild-type JAK2 in varying proportions. Exposure of IM CD34(+) cells to 5azaD/TSA resulted in a reduction of the proportion of JAK2V617F-positive HPCs in 83% of the patients studied and the reduction in the proportion of homozygous HPCs in 50% of the patients. 5azaD/TSA treatment led to a dramatic reduction in the number of HPCs that contained chromosomal abnormalities in two JAK2V617F-negative IM patients. IM is characterized by constitutive mobilization of HPCs, which has been partly attributed to decreased expression of the chemokine receptor CXCR4. Treatment of IM CD34(+) cells with 5azaD/TSA resulted in the up-regulation of CXCR4 expression by CD34(+) cells and restoration of their migration in response to SDF-1. These data provide a rationale for sequential therapy with chromatin-modifying agents for patients with IM.


Subject(s)
Antigens, CD34/biosynthesis , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Chromatin/chemistry , Chromatin/metabolism , Hydroxamic Acids/pharmacology , Primary Myelofibrosis/blood , Primary Myelofibrosis/drug therapy , Protein Synthesis Inhibitors/pharmacology , Antigens, CD34/metabolism , Azacitidine/pharmacology , Cell Movement , DNA (Cytosine-5-)-Methyltransferases/metabolism , Decitabine , Hematopoietic Stem Cells/metabolism , Homozygote , Humans , Mutation , Receptors, CXCR4/metabolism , Stem Cells/metabolism
5.
Blood ; 110(3): 986-93, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17473062

ABSTRACT

In order to investigate the biologic processes underlying and resulting from the megakaryocytic hyperplasia that characterizes idiopathic myelofibrosis (IMF), peripheral blood CD34+ cells isolated from patients with IMF, polycythemia vera (PV), and G-CSF-mobilized healthy volunteers were cultured in the presence of stem cell factor and thrombopoietin. IMF CD34+ cells generated 24-fold greater numbers of megakaryocytes (MKs) than normal CD34+ cells. IMF MKs were also shown to have a delayed pattern of apoptosis and to overexpress the antiapoptotic protein bcl-xL. MK hyperplasia in IMF is, therefore, likely a consequence of both the increased ability of IMF progenitor cells to generate MKs and a decreased rate of MK apoptosis. Media conditioned (CM) by CD61+ cells generated in vitro from CD34+ cells were then assayed for the levels of growth factors and proteases. Higher levels of transforming growth factor-beta (TGF-beta) and active matrix metalloproteinase-9 (MMP9) were observed in media conditioned with IMF CD61+ cells than normal or PV CD61+ cells. Both normal and IMF CD61+ cells produced similar levels of VEGF. MK-derived TGF-B and MMP-9, therefore, likely contribute to the development of many pathological epiphenomena associated with IMF.


Subject(s)
Hematopoietic Stem Cells/metabolism , Matrix Metalloproteinase 9/biosynthesis , Megakaryocytes/metabolism , Primary Myelofibrosis/metabolism , Transforming Growth Factor beta/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Adult , Aged , Antigens, CD34 , Apoptosis/drug effects , Cells, Cultured , Culture Media, Conditioned , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/pathology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Integrin beta3/metabolism , Male , Megakaryocytes/pathology , Middle Aged , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Primary Myelofibrosis/pathology , Thrombopoietin/pharmacology , bcl-X Protein/metabolism
7.
Blood Cells Mol Dis ; 38(3): 280-6, 2007.
Article in English | MEDLINE | ID: mdl-17350297

ABSTRACT

PURPOSE: We studied the expression of the chemokine receptor CXCR4 on circulating CD34+ cells of patients with myelofibrosis with myeloid metaplasia (MMM), and examined its relationship to the severity of disease. PATIENTS AND METHODS: Surface and intracellular CXCR4 expression were measured flow cytometrically in 84 consecutive MMM patients, 16 patients with polycythemia vera (PV), and 20 healthy subjects. In 23 MMM patients, CXCR4 gene expression level was also quantitated by real time-RT-PCR in CD34+ cells. RESULTS: The expression of CXCR4 on circulating CD34+ cells was significantly reduced in patients with MMM (P<0.001) as compared to normal controls and patients with PV (P=0.01). The levels of CXCR4 mRNA in CD34+ cells were lower in patients with MMM as compared with normal subjects, and were directly correlated with the degree of CXCR4 surface expression, demonstrating that transcriptional defects were the major cause for receptor down-regulation. No statistical association was found between JAK2V617F mutational status and the extent of CXCR4 down-regulation. CXCR4 expression on CD34+ cells inversely correlated with the number of circulating CD34+ cells (R=-0.55; P<0.001), and was severely down-regulated in high risk patients and patients with a high "myelodepletion severity index". CXCR4 down-regulation was associated with advanced patient age, the presence of severe anemia, thrombocytopenia, and degree of bone marrow fibrosis. CONCLUSIONS: Reduced expression of CXCR4 by CD34+ cells is a characteristic of MMM which is associated with the constitutive mobilization of CD34+ cells and occurs in patients with advanced forms of the disease.


Subject(s)
Primary Myelofibrosis/blood , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD34 , Biomarkers , Blood Cells/metabolism , Down-Regulation , Female , Humans , Male , Middle Aged , Primary Myelofibrosis/physiopathology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Severity of Illness Index
8.
Blood ; 108(9): 3128-34, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16757685

ABSTRACT

The JAK2(V617F) mutation has been shown to occur in the overwhelming majority of patients with polycythemia vera (PV). To study the role of the mutation in the excessive production of differentiated hematopoietic cells in PV, CD19+, CD3+, CD34+, CD33+, and glycophorin A+ cells and granulocytes were isolated from the peripheral blood (PB) of 8 patients with PV and 3 healthy donors mobilized with G-CSF, and the percentage of JAK2(V617F) mutant allele was determined by quantitative real-time polymerase chain reaction (PCR). The JAK2(V617F) mutation was present in cells belonging to each of the myeloid lineages and was also present in B and T lymphocytes in a subpopulation of patients with PV. The proportion of hematopoietic cells expressing the JAK2(V617F) mutation decreased after differentiation of CD34+ cells in vitro in the presence of optimal concentrations of SCF, IL-3, IL-6, and Epo. These data suggest that the JAK2(V617F) mutation may not provide a proliferative and/or survival advantage for the abnormal PV clone. Although the JAK2(V617F) mutation plays an important role in the biologic origins of PV, it is likely not the sole event leading to PV.


Subject(s)
Hematopoietic Stem Cells/physiology , Janus Kinase 2/genetics , Mutation , Polycythemia Vera/genetics , Amino Acid Substitution , Antigens, CD/analysis , DNA Primers , Glycophorins/analysis , Granulocytes/physiology , Hematopoietic Stem Cells/enzymology , Humans , Janus Kinase 2/chemistry , Polymerase Chain Reaction , Reference Values
9.
Blood ; 105(11): 4508-15, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15705794

ABSTRACT

Idiopathic myelofibrosis (IM) is characterized by increased numbers of CD34(+) cells in the peripheral blood (PB). We explored the possible mechanisms underlying this abnormal trafficking of CD34(+) cells. Plasma levels of neutrophil elastase (NE), total and active matrix metalloproteinase 9 (MMP-9), and soluble vascular cell adhesion molecule-1 (sVCAM-1) were dramatically increased in IM. The absolute number of CD34(+) cells in the PB was correlated with the levels of sVCAM-1. Marked elevations of the levels of NE but not total and active MMP-9 as well as MMP-2 were detected in media conditioned by IM mononuclear cells (MNCs) as compared with that of healthy volunteers. IM MNC-conditioned media, however, was shown by zymographic analysis to contain increased gelatinolytic activity corresponding to the molecular weight of MMP-9. IM MNC-conditioned media also exhibited a greater ability to cleave VCAM-1 and c-kit in vitro, consistent with the biologic actions of NE. In addition, the increased ability of IM PB CD34(+) cells to migrate through a reconstituted basement membrane was diminished by several inhibitors of MMP-9 activity, indicating that these cells express increased levels of this MMP. These data indicate that a proteolytic environment exists in IM which might result in the sustained mobilization of CD34(+) cells.


Subject(s)
Cell Movement , Hematopoietic Stem Cells/pathology , Peptide Hydrolases/metabolism , Primary Myelofibrosis/enzymology , Antigens, CD34 , Blood Cells/pathology , Culture Media, Conditioned/chemistry , Humans , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Proteins/metabolism , Primary Myelofibrosis/blood , Primary Myelofibrosis/etiology , Primary Myelofibrosis/pathology , Vascular Cell Adhesion Molecule-1/metabolism
10.
Blood ; 105(4): 1699-705, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15471948

ABSTRACT

Idiopathic myelofibrosis (IM) is characterized by the constitutive mobilization of CD34(+) cells. IM peripheral blood (PB) CD34(+) cells had a reduced cloning efficiency and a lower frequency of cobblestone areas compared with normal granulocyte colony-stimulating factor (G-CSF)-mobilized PB CD34(+) cells. IM CD34(+) cells engrafted nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, demonstrating that they contain bone marrow (BM)-repopulating cells. G-CSF-mobilized CD34(+) cells produced multiple hematopoietic lineages within the NOD/SCID mice with a predominance of CD19(+) cells. By contrast, IM CD34(+) cells produced predominantly CD33(+) cells, increased numbers of CD41(+) cells, but fewer CD19(+) cells. Transcriptional clonality assays of the engrafted human IM cells demonstrated their clonal origin. CD34(+) cells from one patient isolated prior to leukemic transformation were capable of generating acute leukemia in NOD/SCID mice. The engrafted human cells exhibited the same abnormal karyotype as primary cells in a portion of the population. These findings demonstrate that BM-repopulating cells and more differentiated progenitor cells are constitutively mobilized into the PB in IM, and that their differentiation program is abnormal. In addition, the NOD/SCID model may be useful in gaining an understanding of the events occurring during the transition of IM to acute leukemia.


Subject(s)
Bone Marrow Cells/pathology , Cell Movement , Hematopoiesis , Primary Myelofibrosis/blood , Primary Myelofibrosis/pathology , Acute Disease , Animals , Antigens, CD34/biosynthesis , Bone Marrow Cells/immunology , Clone Cells , Female , Graft Survival/genetics , Graft Survival/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Leukemia/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Primary Myelofibrosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL