Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209117

ABSTRACT

Progressive loss of muscle and muscle function is associated with significant fibrosis in Duchenne muscular dystrophy (DMD) patients. Halofuginone, an analog of febrifugine, prevents fibrosis in various animal models, including those of muscular dystrophies. Effects of (+)/(-)-halofuginone enantiomers on motor coordination and diaphragm histopathology in mdx mice, the mouse model for DMD, were examined. Four-week-old male mice were treated with racemic halofuginone, or its separate enantiomers, for 10 weeks. Controls were treated with saline. Racemic halofuginone-treated mice demonstrated better motor coordination and balance than controls. However, (+)-halofuginone surpassed the racemic form's effect. No effect was observed for (-)-halofuginone, which behaved like the control. A significant reduction in collagen content and degenerative areas, and an increase in utrophin levels were observed in diaphragms of mice treated with racemic halofuginone. Again, (+)-halofuginone was more effective than the racemic form, whereas (-)-halofuginone had no effect. Both racemic and (+)-halofuginone increased diaphragm myofiber diameters, with no effect for (-)-halofuginone. No effects were observed for any of the compounds tested in an in-vitro cell viability assay. These results, demonstrating a differential effect of the halofuginone enantiomers and superiority of (+)-halofuginone, are of great importance for future use of (+)-halofuginone as a DMD antifibrotic therapy.


Subject(s)
Muscle, Skeletal , Muscular Dystrophy, Duchenne , Piperidines/pharmacology , Quinazolinones/pharmacology , Animals , Disease Models, Animal , Fibrosis , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology
2.
Neuromuscul Disord ; 30(6): 472-482, 2020 06.
Article in English | MEDLINE | ID: mdl-32451154

ABSTRACT

Dysferlinopathies are a non-lethal group of late-onset muscular dystrophies. Here, we evaluated the fusion ability of primary myoblasts from young dysf-/- mice and the muscle histopathology prior to, and during early stages of disease onset. The ability of primary myoblasts of 5-week-old dysf-/- mice to form large myotubes was delayed compared to their wild-type counterparts, as evaluated by scanning electron microscopy. However, their fusion activity, as reflected by the presence of actin filaments connecting several cells, was enhanced by the antifibrotic drug halofuginone. Early dystrophic signs were already apparent in 4-week-old dysf-/- mice; their collagen level was double that in wild-type mice and continued to rise until 5 months of age. Continuous treatment with halofuginone from 4 weeks to 5 months of age reduced muscle fibrosis in a phosphorylated-Smad3 inhibition-related manner. Halofuginone also enhanced myofiber hypertrophy, reduced the percentage of centrally nucleated myofibers, and increased muscle performance. Together, the data suggest an inhibitory effect of halofuginone on the muscle histopathology at very early stages of dysferlinopathy, and enhancement of muscle performance. These results offer new opportunities for early pharmaceutical treatment in dysferlinopathies with favorable outcomes at later stages of life.


Subject(s)
Dysferlin , Muscle, Skeletal/drug effects , Muscular Dystrophies, Limb-Girdle/drug therapy , Piperidines/pharmacology , Protein Synthesis Inhibitors/pharmacology , Quinazolinones/pharmacology , Animals , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Muscular Dystrophies, Limb-Girdle/physiopathology
3.
Histol Histopathol ; 34(7): 791-801, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30628720

ABSTRACT

In Duchenne muscular dystrophy (DMD), the progressive loss of muscle and its ability to function is associated with significant fibrosis, representing the major disease complication in patients. Halofuginone, a halogenated analog of the naturally occurring febrifugine, has been shown to prevent fibrosis in various animal models, including those of muscular dystrophies. Here, two optically active enantiomers of deoxyhalofuginone - a halofuginone analogue in which the hydroxy group in position 3 was removed from the piperidinyl entity - were evaluated with respect to their effect on muscle histopathology in mdx mice. Male mdx mice were treated with either deoxyhalofuginone (as single enantiomers or in racemic form), or halofuginone, for 10 weeks, starting at the age of 4 weeks. Halofuginone caused a significant reduction in total collagen content, degenerative areas, as well as in utrophin and phosphorylated-Smad3 levels in the mdx diaphragms. However, neither the deoxyhalofuginone enantiomers, nor its racemic form had any effect on these parameters. A positive effect of the deoxyhalofuginone (+)-enantiomer was observed on myofiber diameters; however, it was lesser than that of halofuginone. It is concluded that the hydroxy group plays a key role in halofuginone's effects related to fibrosis in DMD, and points towards the transforming growth factor ß/Smad3 signaling pathway being involved in this inhibition. Elucidation of the structure-function relationship of halofuginone, in relation to inhibiting fibrosis in muscular dystrophies, is of the utmost importance for creating the next generation of anti-fibrotic therapies that will be more efficacious and less toxic, hence improving life quality of patients.


Subject(s)
Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Piperidines/chemistry , Piperidines/therapeutic use , Quinazolinones/chemistry , Quinazolinones/therapeutic use , Animals , Disease Models, Animal , Fibrillar Collagens/metabolism , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Phosphorylation , Signal Transduction/drug effects , Smad3 Protein/metabolism , Utrophin/metabolism
4.
Hum Mol Genet ; 27(16): 2817-2829, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29771357

ABSTRACT

In the absence of dysferlin, skeletal muscle cells fail to reseal properly after injury, resulting in slow progress of the dysferlinopathy muscular dystrophy (MD). Halofuginone, a leading agent in preventing fibrosis in MDs, was tested for its effects on membrane resealing post-injury. A hypo-osmotic shock assay on myotubes derived from wild-type (Wt) and dysferlin-null (dysf-/-) mice revealed that pre-treatment with halofuginone reduces the percentage of membrane-ruptured myotubes only in dysf-/- myotubes. In laser-induced injury of isolated myofibers, halofuginone decreased the amount of FM1-43 at the injury site of dysf-/- myofibers while having no effect on Wt myofibers. These results implicate halofuginone in ameliorating muscle-cell membrane integrity in dysf-/- mice. Halofuginone increased lysosome scattering across the cytosol of dysf-/- primary myoblasts, in a protein kinase/extracellular signal-regulated protein kinase and phosphoinositide 3 kinase/Akt-dependent manner, in agreement with an elevation in lysosomal exocytotic activity in these cells. A spatial- and age-dependent synaptotagmin-7 (Syt-7) expression pattern was shown in dysf-/- versus Wt mice, suggesting that these pattern alterations are related to the disease progress and that sytnaptotagmin-7 may be compensating for the lack of dysferlin at least with regard to membrane resealing post-injury. While halofuginone did not affect patch-repair-complex key proteins, it further enhanced Syt-7 levels and its spread across the cytosol in dysf-/- myofibers and muscle tissue, and increased its co-localization with lysosomes. Together, the data imply a novel role for halofuginone in membrane-resealing events with Syt-7 possibly taking part in these events.


Subject(s)
Dysferlin/genetics , Muscular Dystrophies, Limb-Girdle/drug therapy , Piperidines/administration & dosage , Quinazolinones/administration & dosage , Synaptotagmins/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Myoblasts/metabolism , Phosphatidylinositol 3-Kinases/genetics
5.
Am J Pathol ; 187(3): 654-664, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28082118

ABSTRACT

Fibrosis is the main complication of muscular dystrophies. We identified moesin, a member of the ezrin-radixin-moesin family, in dystrophic muscles of mice representing Duchenne and congenital muscular dystrophies (DMD and CMD, respectively) and dysferlinopathy, but not in the wild type. High levels of moesin were also observed in muscle biopsy specimens from DMD, Ullrich CMD, and merosin-deficient CMD patients, all of which present high levels of fibrosis. The myofibroblasts, responsible for extracellular matrix protein synthesis, and the macrophages infiltrating the dystrophic muscles were the source of moesin. Moesin-positive cells were embedded within the fibrotic areas between the myofibers adjacent to the collagen type I fibers. Radixin was also synthesized by the myofibroblasts, whereas ezrin colocalized with the myofiber membranes. In animal models and patients' muscles, part of the moesin was in its active phosphorylated form. Inhibition of fibrosis by halofuginone, an antifibrotic agent, resulted in a major decrease in moesin levels in the muscles of DMD and CMD mice. In summary, the results of this study may pave the way for exploiting moesin as a novel target for intervention in MDs, and as part of a battery of biomarkers to evaluate treatment success in preclinical studies and clinical trials.


Subject(s)
Muscular Dystrophies/metabolism , Adult , Animals , Child , Child, Preschool , Collagen Type I/metabolism , Cytoskeletal Proteins/metabolism , Diaphragm/drug effects , Diaphragm/metabolism , Homozygote , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred mdx , Microfilament Proteins , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphorylation/drug effects , Piperidines/pharmacology , Quinazolinones/pharmacology
6.
Biochim Biophys Acta ; 1862(1): 1-11, 2016 01.
Article in English | MEDLINE | ID: mdl-26454207

ABSTRACT

Halofuginone is a leading agent in preventing fibrosis and inflammation in various muscular dystrophies. We hypothesized that in addition to these actions, halofuginone directly promotes the cell-cycle events of satellite cells in the mdx and dysf(-/-) mouse models of early-onset Duchenne muscular dystrophy and late-onset dysferlinopathy, respectively. In both models, addition of halofuginone to freshly prepared single gastrocnemius myofibers derived from 6-week-old mice increased BrdU incorporation at as early as 18h of incubation, as well as phospho-histone H3 (PHH3) and MyoD protein expression in the attached satellite cells, while having no apparent effect on myofibers derived from wild-type mice. BrdU incorporation was abolished by an inhibitor of mitogen-activated protein kinase/extracellular signal-regulated protein kinase, suggesting involvement of this pathway in mediating halofuginone's effects on cell-cycle events. In cultures of myofibers and myoblasts isolated from dysf(-/-) mice, halofuginone reduced Bax and induced Bcl2 expression levels and induced Akt phosphorylation in a time-dependent manner. Addition of an inhibitor of the phosphinositide-3-kinase/Akt pathway reversed the halofuginone-induced cell survival, suggesting this pathway's involvement in mediating halofuginone's effects on survival. Thus, in addition to its known role in inhibiting fibrosis and inflammation, halofuginone plays a direct role in satellite cell activity and survival in muscular dystrophies, regardless of the mutation. These actions are of the utmost importance for improving muscle pathology and function in muscular dystrophies.


Subject(s)
Cell Cycle/drug effects , Cell Survival/drug effects , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Piperidines/therapeutic use , Quinazolinones/therapeutic use , Satellite Cells, Skeletal Muscle/drug effects , Animals , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Phosphatidylinositol 3-Kinases/metabolism , Piperidines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolinones/pharmacology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction/drug effects
7.
Cancer Biol Ther ; 16(11): 1682-8, 2015.
Article in English | MEDLINE | ID: mdl-26192476

ABSTRACT

Strigolactones (SLs) are a novel class of plant hormones. Previously, we found that analogs of SLs induce growth arrest and apoptosis in breast cancer cell lines. These compounds also inhibited the growth of breast cancer stem cell enriched-mammospheres with increased potency. Furthermore, strigolactone analogs inhibited growth and survival of colon, lung, prostate, melanoma, osteosarcoma and leukemia cancer cell lines. To further examine the anti-cancer activity of SLs in vivo, we have examined their effects on growth and viability of MDA-MB-231 tumor xenografts model either alone or in combination with paclitaxel. We show that strigolactone act as new anti-cancer agents in inhibition of breast cancer in xenograft model. In addition we show that SLs affect the integrity of the microtubule network and therefore may inhibit the migratory phenotype of the highly invasive breast cancer cell lines that were examined.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Lactones/pharmacology , Paclitaxel/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Drug Synergism , Female , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Lactones/therapeutic use , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Paclitaxel/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Lab Invest ; 95(8): 914-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26097999

ABSTRACT

Accumulation of cholesterol in the liver is associated with the development of non-alcoholic steatohepatitis-related fibrosis. However, underlying mechanisms are not well understood. The present study investigated the role of inducible nitric oxide synthase (iNOS) in cholesterol-induced liver fibrosis by feeding wild-type (WT) and iNOS-deficient mice with control or high-cholesterol diet (HCD) for 6 weeks. WT mice fed with HCD developed greater liver fibrosis, compared with iNOS-deficient mice, as evident by Sirius red staining and higher expression levels of profibrotic genes. Enhanced liver fibrosis in the presence of iNOS was associated with hypoxia-inducible factor-1α stabilization, matrix metalloproteinase-9 expression, and enhanced hepatic DNA damage. The profibrotic role of iNOS was also demonstrated in vivo using a selective inhibitor of iNOS as well as in vitro in a rat liver stellate cell line (HSC-T6). In conclusion, these findings suggest that iNOS is an important mediator in HCD-induced liver fibrosis.


Subject(s)
Cholesterol/toxicity , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Nitric Oxide Synthase Type II/genetics , Animals , Cell Line , DNA Damage/drug effects , DNA Damage/genetics , Diet, High-Fat , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/drug effects , Liver/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Rats
9.
Oncotarget ; 6(27): 23249-60, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26015394

ABSTRACT

Duchenne Muscular Dystrophy is characterized by: near absence of dystrophin in skeletal muscles; low percentage of revertant myofibers; up-regulation of utrophin synthesis; and a high degree of muscle fibrosis. In patient quadriceps femoris biopsies (n = 6, ages between 3-9 years) an inverse correlation was observed between the levels of collagen type I - representing fibrosis - and the levels of utrophin. This correlation was independent of the patient's age and was observed in the entire muscle biopsy sections. In the mdx mice diaphragm (n = 6/group), inhibition of fibrosis by halofuginone resulted in increases in the levels of utrophin. The utrophin/fibrosis relationships were not limited to collagen type I, but also applied to other constituents of the fibrosis machinery. The inverse correlation was found also in old mdx mice with established fibrosis. In addition, inhibition of collagen type I levels was associated with increases in the numbers of revertant myofibers, both as single myofibers and in clusters in the diaphragm and the gastrocnemius. In summary, our results demonstrate an inverse correlation between the level of muscle fibrosis and the level of utrophin and that of the number of revertant myofibers. These findings may reveal common links between the fibrotic and utrophin-synthesis pathways and offer new insights into the regulation of utrophin synthesis.


Subject(s)
Gene Expression Regulation , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Utrophin/metabolism , Animals , Biopsy , Child , Child, Preschool , Collagen/metabolism , Collagen Type I/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Piperidines/chemistry , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Quinazolinones/chemistry
10.
Molecules ; 20(1): 573-94, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25569515

ABSTRACT

Halofuginone is an analog of febrifugine-an alkaloid originally isolated from the plant Dichroa febrifuga. During recent years, halofuginone has attracted much attention because of its wide range of beneficial biological activities, which encompass malaria, cancer, and fibrosis-related and autoimmune diseases. At present two modes of halofuginone actions have been described: (1) Inhibition of Smad3 phosphorylation downstream of the TGFß signaling pathway results in inhibition of fibroblasts-to-myofibroblasts transition and fibrosis. (2) Inhibition of prolyl-tRNA synthetase (ProRS) activity in the blood stage of malaria and inhibition of Th17 cell differentiation thereby inhibiting inflammation and the autoimmune reaction by activation of the amino acid starvation and integrated stress responses. This review deals with the history and origin of this natural product, its synthesis, its known modes of action, and it's various biological activities in pre-clinical animal models and in humans.


Subject(s)
Piperidines/therapeutic use , Quinazolinones/therapeutic use , Animals , Antimalarials/therapeutic use , Antineoplastic Agents/therapeutic use , Antiprotozoal Agents/therapeutic use , Apoptosis/drug effects , Clinical Trials as Topic , Humans , Piperidines/chemical synthesis , Quinazolinones/chemical synthesis
11.
World J Gastroenterol ; 20(40): 14778-86, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25356039

ABSTRACT

Organ fibrosis and architectural remodeling can severely disrupt tissue function, often with fatal consequences. Fibrosis is the end result of chronic inflammatory reactions induced by a variety of stimuli, and the key cellular mediator of fibrosis comprises the myofibroblasts which, when activated, serve as the primary collagen-producing cells. Complex links exist between fibrosis, regeneration and carcinogenesis, and the concept that all organs contain common tissue fibrosis pathways that could be potential therapeutic targets is an attractive one. Because of the major impact of fibrosis on human health there is an unmet need for safe and effective therapies that directly target fibrosis. Halofuginone inhibits tissue fibrosis and regeneration, and thereby affects the development of tumors in various tissues along the gastrointestinal tract. The high efficacy of halofuginone in reducing the fibrosis that affects tumor growth and tissue regeneration is probably due to its dual role in inhibiting the signaling pathway of transforming growth factor ß, on the one hand, and inhibiting the development of Th17 cells, on the other hand. At present halofuginone is being evaluated in a clinical trial for other fibrotic indication, and any clinical success in that trial would allow the use of halofuginone, also for all other fibrotic indications, including those of the gastrointestinal tract.


Subject(s)
Antineoplastic Agents/therapeutic use , Digestive System Diseases/drug therapy , Digestive System Neoplasms/drug therapy , Gastrointestinal Agents/therapeutic use , Gastrointestinal Tract/drug effects , Piperidines/therapeutic use , Quinazolinones/therapeutic use , Regeneration/drug effects , Animals , Digestive System Diseases/metabolism , Digestive System Diseases/pathology , Digestive System Diseases/physiopathology , Digestive System Neoplasms/metabolism , Digestive System Neoplasms/pathology , Digestive System Neoplasms/physiopathology , Fibrosis , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Humans , Signal Transduction/drug effects
12.
Biochim Biophys Acta ; 1843(7): 1339-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24703880

ABSTRACT

Halofuginone has been shown to prevent fibrosis via the transforming growth factor-ß/Smad3 pathway in muscular dystrophies. We hypothesized that halofuginone would reduce apoptosis--the presumed cause of satellite-cell depletion during muscle degradation-in the mdx mouse model of Duchenne muscular dystrophy. Six-week-old mdx mouse diaphragm exhibited fourfold higher numbers of apoptotic nuclei compared with wild-type mice as determined by a TUNEL assay. Apoptotic nuclei were found in macrophages and in Pax7-expressing cells; some were located in centrally-nucleated regenerating myofibers. Halofuginone treatment of mdx mice reduced the apoptotic nuclei number in the diaphragm, together with reduction in Bax and induction in Bcl2 levels in myofibers isolated from these mice. A similar effect was observed when halofuginone was added to cultured myofibers. No apparent effect of halofuginone was observed in wild-type mice. Inhibition of apoptosis or staurosporine-induced apoptosis by halofuginone in mdx primary myoblasts and C2 myogenic cell line, respectively, was reflected by less pyknotic/apoptotic cells and reduced Bax expression. This reduction was reversed by a phosphinositide-3-kinase and mitogen-activated protein kinase/extracellular signal-regulated protein kinase inhibitors, suggesting involvement of these pathways in mediating halofuginone's effects on apoptosis. Halofuginone increased apoptosis in α smooth muscle actin- and prolyl 4-hydroxylase ß-expressing cells in mdx diaphragm and in myofibroblasts, the major source of extracellular matrix. The data suggest an additional mechanism by which halofuginone improves muscle pathology and function in muscular dystrophies.


Subject(s)
Diaphragm/drug effects , Macrophages/drug effects , Myoblasts/drug effects , Myofibrils/drug effects , Piperidines/pharmacology , Quinazolinones/pharmacology , Actins/genetics , Actins/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Diaphragm/metabolism , Diaphragm/pathology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred mdx , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Myoblasts/pathology , Myofibrils/metabolism , Myofibrils/pathology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Bioorg Med Chem ; 22(7): 1993-2004, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24650700

ABSTRACT

The trans-2,3-disubstituted piperidine, quinazolinone-containing natural product febrifugine (also known as dichroine B) and its synthetic analogue, halofuginone, possess antimalarial activity. More recently studies have also shown that halofuginone acts as an agent capable of reducing fibrosis, an indication with clinical relevance for several disease states. This review summarizes historical isolation studies and the chemistry performed which culminated in the correct structural elucidation of naturally occurring febrifugine and its isomer isofebrifugine. It also includes the range of febrifugine analogues prepared for antimalarial evaluation, including halofuginone. Finally, a section detailing current opinion in the field of halofuginone's human biology is included.


Subject(s)
Antimalarials/pharmacology , Piperidines/pharmacology , Plasmodium/drug effects , Quinazolines/pharmacology , Quinazolinones/pharmacology , Antimalarials/chemistry , Antimalarials/isolation & purification , Humans , Molecular Structure , Parasitic Sensitivity Tests , Piperidines/chemistry , Piperidines/isolation & purification , Quinazolines/chemistry , Quinazolines/isolation & purification , Quinazolinones/chemistry , Quinazolinones/isolation & purification , Structure-Activity Relationship
14.
Am J Pathol ; 182(3): 905-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23274062

ABSTRACT

Fibrosis is the main complication of muscular dystrophies. We identified collagen triple helix repeat containing 1 (Cthrc1) in skeletal and cardiac muscles of mice, representing Duchenne and congenital muscle dystrophies (DMD and CMD, respectively), and dysferlinopathy. In all of the mice, Cthrc1 was associated with high collagen type I levels; no Cthrc1 or collagen was observed in muscles of control mice. High levels of Cthrc1 were also observed in biopsy specimens from patients with DMD, in whom they were reversibly correlated with that of ß-dystroglycan, whereas collagen type I levels were elevated in all patients with DMD. At the muscle sites where collagen and Cthrc1 were adjacent, collagen fibers appeared smaller, suggesting involvement of Cthrc1 in collagen turnover. Halofuginone, an inhibitor of Smad3 phosphorylation downstream of the transforming growth factor-ß signaling, reduced Cthrc1 levels in skeletal and cardiac muscles of mice, representing DMD, CMD, and dysferlinopathy. The myofibroblasts infiltrating the dystrophic muscles of the murine models of DMD, CMD, and dysferlinopathy were the source of Cthrc1. Transforming growth factor-ß did not affect Cthrc1 levels in the mdx fibroblasts but decreased them in the control fibroblasts, in association with increased migration of mdx fibroblasts and dystrophic muscle invasion by myofibroblasts. To our knowledge, this is the first demonstration of Cthrc1 as a marker of the severity of the disease progression in the dystrophic muscles, and as a possible target for therapy.


Subject(s)
Extracellular Matrix Proteins/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Blotting, Western , Cell Movement , Collagen Type I/metabolism , Diaphragm/metabolism , Diaphragm/pathology , Disease Progression , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology
15.
Histol Histopathol ; 28(2): 211-26, 2013 02.
Article in English | MEDLINE | ID: mdl-23275304

ABSTRACT

Absence of, or loss-of-function mutations in the dysferlin gene (dysf) result in dysferlinopathy, characterized by increased muscle inflammation, collagen deposition and deterioration in muscle function. We evaluated halofuginone efficacy in improving muscle histopathology in mice with deleted dysf transmembrane domain. Quadriceps sublumbar and longissimus muscles of 9-month-old dysf-/- mice treated with halofuginone for 4 months exhibited a reduction in centrally-nucleated myofibers, inflammatory infiltrates and collagen content. Late onset of dysferlinopathy makes it ideal for evaluating the efficacy of early treatments on late outcome. The dysf-/- mice were treated with halofuginone for 3 to 4 months starting at 1, 5 or 9 months of age, and quadricep muscle histopathology was evaluated at 12 months. Collagen content and number of centrally nucleated myofibers decreased after early halofuginone treatment, administered when myofibers with central nuclei and inflammatory infiltrates are evident, but there was almost no fibrosis. When administered at the beginning of fibrosis it resulted in a further decrease in the number of centrally-nucleated myofibers with no additional decrease in collagen levels. Cardiac fibrosis was almost completely abolished following early halofuginone treatment. Halofuginone inhibited Smad3 phosphorylation and its translocation to the nucleus and increased the activity of matrix metalloproteinases 9 and 2 responsible for resolution of pre-existing collagen. Macrophage and myofibroblast invasion into the dystrophic muscle at the site of myofibers with central nuclei was inhibited by halofuginone. These results suggest that early halofuginone treatment can prevent the late outcome of dysferlinopathy and can cause resolution of the established fibrosis when administered at later stages.


Subject(s)
Membrane Proteins/deficiency , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophies, Limb-Girdle/pathology , Piperidines/therapeutic use , Protein Synthesis Inhibitors/therapeutic use , Quinazolinones/therapeutic use , Animals , Collagen/metabolism , Disease Models, Animal , Dysferlin , Fibrosis , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscular Dystrophies, Limb-Girdle/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Protein Synthesis Inhibitors/pharmacology , Quinazolinones/pharmacology , Smad3 Protein/antagonists & inhibitors , Treatment Outcome
16.
PLoS One ; 7(7): e41833, 2012.
Article in English | MEDLINE | ID: mdl-22848627

ABSTRACT

INTRODUCTION: Stroma cells and extracellular matrix (ECM) components provide the pivotal microenvironment for tumor development. The study aimed to evaluate the importance of the pancreatic stroma for tumor development. METHODS: Pancreatic tumor cells were implanted subcutaneously into green fluorescent protein transgenic mice, and stroma cells invading the tumors were identified through immunohistochemistry. Inhibition of tumor invasion by stroma cells was achieved with halofuginone, an inhibitor of TGFß/Smad3 signaling, alone or in combination with chemotherapy. The origin of tumor ECM was evaluated with species-specific collagen I antibodies and in situ hybridization of collagen α1(I) gene. Pancreatic fibrosis was induced by cerulean injection and tumors by spleen injection of pancreatic tumor cells. RESULTS: Inhibition of stroma cell infiltration and reduction of tumor ECM levels by halofuginone inhibited development of tumors derived from mouse and human pancreatic cancer cells. Halofuginone reduced the number only of stroma myofibroblasts expressing both contractile and collagen biosynthesis markers. Both stroma myofibroblasts and tumor cells generated ECM that contributes to tumor growth. Combination of treatments that inhibit stroma cell infiltration, cause apoptosis of myofibroblasts and inhibit Smad3 phosphorylation, with chemotherapy that increases tumor-cell apoptosis without affecting Smad3 phosphorylation was more efficacious than either treatment alone. More tumors developed in fibrotic than in normal pancreas, and prevention of tissue fibrosis greatly reduced tumor development. CONCLUSIONS: The utmost importance of tissue fibrosis and of stroma cells for tumor development presents potential new therapy targets, suggesting combination therapy against stroma and neoplastic cells as a treatment of choice.


Subject(s)
Pancreatic Neoplasms/pathology , Stromal Cells/pathology , Animals , Anticarcinogenic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Cell Proliferation/drug effects , Ceruletide/adverse effects , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Fibrosis , Humans , Male , Mice , Mice, Transgenic , Myofibroblasts/drug effects , Myofibroblasts/pathology , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Piperidines/pharmacology , Quinazolinones/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism
17.
Br J Haematol ; 157(6): 718-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22533681

ABSTRACT

Multiple Myeloma (MM), a malignancy of plasma cells, remains incurable despite the use of conventional and novel therapies. Halofuginone (HF), a synthetic derivative of quinazolinone alkaloid, has recently been shown to have anti-cancer activity in various preclinical settings. This study demonstrated the anti-tumour activity of HF against a panel of human MM cell lines and primary patient-derived MM cells, regardless of their sensitivity to conventional therapy or novel agents. HF showed anti-MM activity in vivo using a myeloma xenograft mouse model. HF suppressed proliferation of myeloma cells alone and when co-cultured with bone marrow stromal cells. Similarly, HF induced apoptosis in MM cells even in the presence of insulin-like growth factor 1 or interleukin 6. Importantly, HF, even at high doses, did not induce cytotoxicity against CD40 activated peripheral blood mononuclear cells from normal donors. HF treatment induced accumulation of cells in the G(0) /G(1) cell cycle and induction of apoptotic cell death associated with depletion of mitochondrial membrane potential; cleavage of poly (ADP-ribose) polymerase and caspases-3, 8 and 9 as well as down-regulation of anti-apoptotic proteins including Mcl-1 and X-IAP. Multiplex analysis of phosphorylation of diverse components of signalling cascades revealed that HF induced changes in P38MAPK activation; increased phosphorylation of c-jun, c-jun NH(2)-terminal kinase (JNK), p53 and Hsp-27. Importantly, HF triggered synergistic cytotoxicity in combination with lenalidomide, melphalan, dexamethasone, and doxorubicin. Taken together, these preclinical studies provide the preclinical framework for future clinical studies of HF in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , G1 Phase/drug effects , Multiple Myeloma/drug therapy , Piperidines/pharmacology , Quinazolinones/pharmacology , Resting Phase, Cell Cycle/drug effects , Animals , Antineoplastic Agents/agonists , Antineoplastic Agents/therapeutic use , Caspases/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Piperidines/agonists , Piperidines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolinones/agonists , Quinazolinones/therapeutic use , Tumor Suppressor Protein p53/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Can J Gastroenterol ; 26(1): 33-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22288068

ABSTRACT

BACKGROUND: Intestinal fibrosis is a challenging clinical condition in several fibrostenosing enteropathies, particularly Crohn's disease. Currently, no effective preventive measures or medical therapies are available for intestinal fibrosis. Fibrosis, due to an abnormal accumulation of extracellular matrix proteins, is a chronic and progressive process mediated by cell/matrix/cytokine and growth factor interactions, but may be a reversible phenomenon. Of the several molecules regulating fibrogenesis, transforming growth factor-beta 1 (TGF-b1) appears to play a pivotal role; it is strongly induced by the local activation of angiotensin II. The levels of both TGF-b1 and angiotensin II are elevated in fibrostenosing Crohn's disease. AIMS: To evaluate the in vivo effect of losartan - an angiotensin II receptor antagonist - on the course of chronic colitis-associated fibrosis and on TGF-b1 expression. METHODS: Colitis was induced by intrarectal instillation of trinitrobenzene sulphonic acid (TNBS) (15 mg/mL) while losartan was administered orally daily by gavage (7 mg/kg/day) for 21 days. Three groups of rats were evaluated: control (n=10); TNBS treated (n=10); and TNBS + losartan treated (n=10). Inflammation and fibrosis of the colon were evaluated by macro- and microscopic score analysis. Colonic TGF-b1 levels was measured using ELISA. RESULTS: Twenty-one days after induction, losartan significantly improved the macro- and microscopic scores of fibrosis in the colonic wall and reduced TGF-b1 concentration. CONCLUSIONS: Prophylactic oral administration of losartan reduces the colorectal fibrosis complicating the TNBS-induced chronic colitis, an effect that appears to be mediated by a downregulation of TGF-b1 expression.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Colitis/drug therapy , Intestinal Mucosa/pathology , Losartan/therapeutic use , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Colitis/chemically induced , Colitis/pathology , Colitis/physiopathology , Disease Models, Animal , Disease Progression , Down-Regulation/drug effects , Fibrosis , Intestinal Mucosa/chemistry , Losartan/administration & dosage , Losartan/pharmacology , Male , Rats , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/physiology , Trinitrobenzenesulfonic Acid/adverse effects
19.
PLoS One ; 6(7): e22770, 2011.
Article in English | MEDLINE | ID: mdl-21818387

ABSTRACT

BACKGROUND: Th17 cells play a major role in coordinating the host defence in oropharyngeal candidiasis. In this study we investigated the involvement of the Th17 response in an animal model of vulvovaginal candidiasis (VVC). METHODS: To monitor the course of infection we exploited a new in vivo imaging technique. RESULTS: i) The progression of VVC leads to a strong influx of neutrophils in the vagina soon after the challenge which persisted despite the resolution of infection; ii) IL-17, produced by vaginal cells, particularly CD4 T cells, was detected in the vaginal wash during the infection, reaching a maximum 14 days after the challenge; iii) The amount and kinetics of IL-23 in vaginal fluids were comparable to those in vaginal cells; iv) The inhibition of Th17 differentiation led to significant inhibition of IL-17 production with consequent exacerbation of infection; v) An increased production of ßdefensin 2 was manifested in cells of infected mice. This production was strongly reduced when Th17 differentiation was inhibited and was increased by rIL-17 treatment. CONCLUSIONS: These results imply that IL-17 and Th17, along with innate antimicrobial factors, have a role in the immune response to vaginal candidiasis.


Subject(s)
Candidiasis, Vulvovaginal/immunology , Candidiasis, Vulvovaginal/prevention & control , Immunity/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Animals , Candida albicans/drug effects , Candida albicans/immunology , Candidiasis, Vulvovaginal/microbiology , Disease Models, Animal , Female , Immunity/drug effects , Interleukin-23/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Piperidines/pharmacology , Piperidines/therapeutic use , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Th17 Cells/drug effects , Vagina/drug effects , Vagina/immunology , Vagina/microbiology , Vagina/pathology , beta-Defensins/metabolism
20.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R140-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21562096

ABSTRACT

Thiram-induced tibial dyschondroplasia (TD) and vitamin-D deficiency rickets are avian bone disorders of different etiologies characterized by abnormal chondrocyte differentiation, enlarged and unvascularized growth plates, and lameness. Heat-shock protein 90 (Hsp90) is a proangiogenic factor in mammalian tissues and in tumors; therefore, Hsp90 inhibitors were developed as antiangiogenic factors. In this study, we evaluated the association between Hsp90, hypoxia, and angiogenesis in the chick growth plate. Administration of the Hsp90 inhibitor to TD- and rickets-afflicted chicks at the time of induction resulted in reduction in growth-plate size and, contrary to its antiangiogenic effect in tumors, a major invasion of blood vessels occurred in the growth plates. This was the result of upregulation of the VEGF receptor Flk-1, the major rate-limiting factor of vascularization in TD and rickets. In addition, the abnormal chondrocyte differentiation, as characterized by collagen type II expression and alkaline phosphatase activity, and the changes in hypoxia-inducible factor-1α (HIF-1α) in both disorders were restored. All these changes resulted in prevention of lameness. Inhibition of Hsp90 activity reduced growth-plate size, increased vascularization, and mitigated lameness also in TD chicks with established lesions. In summary, this is the first reported demonstration of involvement of Hsp90 in chondrocyte differentiation and growth-plate vascularization. In contrast to the antiangiogenic effect of Hsp90 inhibitors observed in mammals, inhibition of Hsp90 activity in the unvascularized TD- and rickets-afflicted chicks resulted in activation of the angiogenic switch and reinstated normal growth-plate morphology.


Subject(s)
Growth Plate/blood supply , Growth Plate/physiopathology , HSP90 Heat-Shock Proteins/physiology , Neovascularization, Physiologic/physiology , Osteochondrodysplasias/physiopathology , Rickets/physiopathology , Alkaline Phosphatase/metabolism , Animals , Benzoquinones/pharmacology , Cell Differentiation/physiology , Chickens , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/metabolism , Disease Models, Animal , Growth Plate/pathology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactams, Macrocyclic/pharmacology , Male , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Rickets/metabolism , Rickets/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...