Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
Curr Rheumatol Rep ; 10(3): 235-48, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18638433

ABSTRACT

Rheumatic syndromes are cause for morbidity in patients with end-stage renal disease. Recent advances in understanding the role of tissue remodeling have provided insight into the pathogenic mechanisms responsible for some of these manifestations. Here, we survey recent and clinically relevant advances in translational research that impact our understanding of rheumatic syndromes seen in patients with significant renal disease. The management of acute and chronic crystalline arthropathies in chronic kidney disease and hemodialysis patients is discussed.


Subject(s)
Chondrocalcinosis/etiology , Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Gout/etiology , Rheumatic Diseases/etiology , Uremia/complications , Bone Remodeling , Calcium Oxalate/metabolism , Chondrocalcinosis/metabolism , Chondrocalcinosis/physiopathology , Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/physiopathology , Crystallization , Gout/metabolism , Gout/physiopathology , Humans , Rheumatic Diseases/metabolism , Rheumatic Diseases/physiopathology , Uremia/metabolism , Uremia/physiopathology
3.
Curr Rheumatol Rep ; 8(4): 303-11, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16839510

ABSTRACT

Wegener's granulomatosis (WG) is a complex autoimmune disorder that has been transformed from a uniformly lethal process to a chronic disease with a relapsing-remitting course. In the setting of frequent relapses, the need to manage cumulative disease damage and drug toxicities has spurred the identification and development of new potent and directed therapies. Biologic agents, which offer the potential for remission-induction and drug-sparing approaches to treat WG, have been studied in several small, open-label clinical series and one large, randomized, placebo-controlled clinical trial. This article discusses the results of these trials and the potential of these biologic agents to treat WG.


Subject(s)
Biological Therapy , Granulomatosis with Polyangiitis/drug therapy , Immunologic Factors/therapeutic use , Animals , Controlled Clinical Trials as Topic , Disease Models, Animal , Granulomatosis with Polyangiitis/etiology , Humans
4.
Expert Rev Mol Med ; 7(8): 1-19, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15890083

ABSTRACT

Wegener's granulomatosis (WG) is a complex autoimmune syndrome that is characterised by upper/lower respiratory necrotising granulomatosis, glomerulonephritis and small-vessel vasculitis. Since Wegener's 1936 description, considerable advances in recognition and treatment have changed this disease from a rapidly and uniformly fatal illness to a chronic disease characterised by remissions and relapses. The serendipitous discovery of anti-neutrophil cytoplasmic antibodies (ANCAs) as a marker associated with WG focused attention on the potential pathogenic role of these antibodies and has recently led to the development of novel animal models that might facilitate our understanding of the disease pathogenesis. Future animal models of this disease will have to account for the role of both ANCA-mediated pathology and granulomatous inflammation to enable us to understand the chronic and persistent features of WG in humans.


Subject(s)
Autoimmune Diseases/etiology , Granulomatosis with Polyangiitis/etiology , Adolescent , Adult , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Autoantigens/genetics , Autoantigens/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Chemokines/physiology , Child , Cytokines/physiology , Female , Granulomatosis with Polyangiitis/drug therapy , Granulomatosis with Polyangiitis/immunology , Granulomatosis with Polyangiitis/pathology , Humans , Kidney/pathology , Lung/pathology , Male , Mice , Mice, Knockout , Myeloblastin/deficiency , Myeloblastin/genetics , Myeloblastin/immunology , Peroxidase/deficiency , Peroxidase/genetics , Peroxidase/immunology , Th1 Cells/immunology
5.
Mol Cell Biol ; 23(20): 7222-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517292

ABSTRACT

The nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPAR gamma) is an important regulator of lipid and glucose homeostasis and cellular differentiation. Studies of many cell types in vitro and in vivo have demonstrated that activation of PPAR gamma can reduce cellular proliferation. We show here that activation of PPAR gamma is sufficient to reduce the proliferation of cultured insulinoma cell lines. We created a model with mice in which the expression of the PPARG gene in beta cells was eliminated (beta gamma KO mice), and these mice were found to have significant islet hyperplasia on a chow diet. Interestingly, the normal expansion of beta-cell mass that occurs in control mice in response to high-fat feeding is markedly blunted in these animals. Despite this alteration in beta-cell mass, no effect on glucose homeostasis in beta gamma KO mice was noted. Additionally, while thiazolidinediones enhanced insulin secretion from cultured wild-type islets, administration of rosiglitazone to insulin-resistant control and beta gamma KO mice revealed that PPAR gamma in beta cells is not required for the antidiabetic actions of these compounds. These data demonstrate a critical physiological role for PPAR gamma function in beta-cell proliferation and also indicate that the mechanisms controlling beta-cell hyperplasia in obesity are different from those that regulate baseline cell mass in the islet.


Subject(s)
Glucose/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Blood Glucose/metabolism , Cell Differentiation , Cell Division , Cell Separation , Chromans/pharmacology , Crosses, Genetic , Dose-Response Relationship, Drug , Exons , Flow Cytometry , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Immunohistochemistry , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Cytoplasmic and Nuclear/genetics , Thiazolidinediones/pharmacology , Time Factors , Transcription Factors/genetics , Troglitazone
6.
Breast Cancer Res Treat ; 79(3): 391-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12846423

ABSTRACT

PURPOSE: To evaluate the therapeutic effects of the peroxisome proliferator-activated receptor (PPAR) gamma activating ligand, troglitazone, in patients with refractory metastatic breast cancer. EXPERIMENTAL DESIGN: Patients with advanced breast cancer refractory to at least one chemotherapy regimen (ER negative tumors) or two hormonal regimens (ER positive tumors) were treated with troglitazone at 800 mg p.o. QD until disease progression, to determine the percentage of patients free of progression at 6 months. Tumor response, toxicity, and changes in serum tumor markers (CEA, CA27.29) that might reflect alteration in tumor differentiation, were also examined. RESULTS: Twenty-two patients were enrolled before suspension of protocol accrual and treatment when troglitazone was withdrawn from commercial availability following FDA warnings on hepatic toxicity. No objective responses (CR or PR) were observed; only three patients had SD at 8 weeks. Patients came off study for PD (16), DLT (1), FDA withdrawal (2), or other (3) reasons. No patients took troglitazone for more than 20 weeks; all had experienced disease progression or began other systemic therapy within 6 months. All patients with elevated serum tumor markers (CEA and CA27.29) at baseline had rising tumor markers within 8 weeks. CONCLUSIONS: While clinical trials among different patient populations might uncover subtle effects on tumor differentiation, PPARgamma activation by troglitazone has little apparent clinical value among patients with treatment-refractory metastatic breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chromans/pharmacology , Thiazoles/pharmacology , Thiazolidinediones , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Breast Neoplasms/pathology , Chromans/administration & dosage , Chromans/adverse effects , Disease Progression , Female , Humans , Liver/drug effects , Liver/pathology , Middle Aged , Neoplasm Invasiveness , Thiazoles/administration & dosage , Thiazoles/adverse effects , Treatment Outcome , Troglitazone
7.
J Biol Chem ; 278(25): 22669-77, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12591919

ABSTRACT

Activation of the nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARgamma) inhibits cell growth and induces differentiation in both adipocyte and epithelial cell lineages, although it is unclear whether this occurs through common or cell-type specific mechanisms. We have identified four human colon cancer cell lines that do no undergo growth inhibition or induce markers of differentiation after exposure to PPARgamma agonists. Sequence analysis of the PPARgamma gene revealed that all four cell lines contain a previously unidentified point mutation in the ninth alpha-helix of the ligand binding domain at codon 422 (K422Q). The mutant receptor did not exhibit any defects in DNA binding or retinoid X receptor heterodimerization and was transcriptionally active in an artificial reporter assay. However, only retroviral transduction of the wild-type (WT), but not mutant, receptor could restore PPARgamma ligand-induced growth inhibition and differentiation in resistant colon cancer cell lines. In contrast, there was no difference in the ability of fibroblast cells expressing WT or K422Q mutant receptor to undergo growth inhibition, express adipocyte differentiation markers, or uptake lipid after treatment with a PPARgamma agonist. Finally, analysis of direct PPARgamma target genes in colon cancer cells expressing the WT or K422Q mutant allele suggests that the mutation may disrupt the ability of PPARgamma to repress the basal expression of a subset of genes in the absence of exogenous ligand. Collectively, these data argue that codon 422 may be a part of a co-factor(s) interaction domain necessary for PPARgamma to induce terminal differentiation in epithelial, but not adipocyte, cell lineages and argues that the receptor induces growth inhibition and differentiation via cell lineage-specific mechanisms.


Subject(s)
Colonic Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Differentiation , Colonic Neoplasms/pathology , Genetic Variation , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Organ Specificity , Protein Conformation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/chemistry , Transcription Factors/physiology , Tumor Cells, Cultured
8.
J Biol Chem ; 278(9): 7431-8, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12468551

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) and transforming growth factor-beta (TGF-beta) are key regulators of epithelial cell biology. However, the molecular mechanisms by which either pathway induces growth inhibition and differentiation are incompletely understood. We have identified transforming growth factor-simulated clone-22 (TSC-22) as a target gene of both pathways in intestinal epithelial cells. TSC-22 is member of a family of leucine zipper containing transcription factors with repressor activity. Although little is known regarding its function in mammals, the Drosophila homolog of TSC-22, bunched, plays an essential role in fly development. The ability of PPARgamma to induce TSC-22 was not dependent on an intact TGF-beta1 signaling pathway and was specific for the gamma isoform. Localization studies revealed that TSC-22 mRNA is enriched in the postmitotic epithelial compartment of the normal human colon. Cells transfected with wild-type TSC-22 exhibited reduced growth rates and increased levels of p21 compared with vector-transfected cells. Furthermore, transfection with a dominant negative TSC-22 in which both repressor domains were deleted was able to reverse the p21 induction and growth inhibition caused by activation of either the PPARgamma or TGF-beta pathways. These results place TSC-22 as an important downstream component of PPARgamma and TGF-beta signaling during intestinal epithelial cell differentiation.


Subject(s)
Epithelial Cells/cytology , Intestines/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/biosynthesis , Thiazolidinediones , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Northern , Blotting, Western , COS Cells , Cell Differentiation , Cell Division , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Genes, Dominant , Genetic Vectors , Humans , In Situ Hybridization , Intermediate Filament Proteins/metabolism , Keratin-20 , Ligands , Mutation , Oligonucleotide Array Sequence Analysis , Precipitin Tests , Protein Structure, Tertiary , Rosiglitazone , Signal Transduction , Thiazoles/pharmacology , Time Factors , Transcription, Genetic , Transfection
9.
Proc Natl Acad Sci U S A ; 99(21): 13771-6, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370429

ABSTRACT

Activation of PPARgamma by synthetic ligands, such as thiazolidinediones, stimulates adipogenesis and improves insulin sensitivity. Although thiazolidinediones represent a major therapy for type 2 diabetes, conflicting studies showing that these agents can increase or decrease colonic tumors in mice have raised concerns about the role of PPARgamma in colon cancer. To analyze critically the role of this receptor, we have used mice heterozygous for Ppargamma with both chemical and genetic models of this malignancy. Heterozygous loss of PPARgamma causes an increase in beta-catenin levels and a greater incidence of colon cancer when animals are treated with azoxymethane. However, mice with preexisting damage to Apc, a regulator of beta-catenin, develop tumors in a manner insensitive to the status of PPARgamma. These data show that PPARgamma can suppress beta-catenin levels and colon carcinogenesis but only before damage to the APC/beta-catenin pathway. This finding suggests a potentially important use for PPARgamma ligands as chemopreventative agents in colon cancer.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Genes, APC , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Gene Silencing , Humans , Hypoglycemic Agents/adverse effects , Male , Mice , Mice, Knockout , Mutation , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/adverse effects , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , beta Catenin
10.
J Biol Chem ; 277(44): 41925-30, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12200443

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) gamma is a nuclear receptor that is a key regulator of adipogenesis and is present in two isoforms generated by alternative splicing, PPARgamma1 and PPARgamma2. Studies of the ability of each isoform to stimulate fat differentiation have yielded ambiguous results, in part because PPARgamma stimulates its own expression. We have thus undertaken a formal genetic analysis using PPARgamma-null fibroblast cell lines to assess the specific role of each individual isoform in adipogenesis. We show here that both PPARgamma1 and PPARgamma2 have the intrinsic ability to stimulate robust adipogenesis. Adipose cells stimulated by either PPARgamma1 or PPARgamma2 express a similar gene profile and show similar responses to insulin. However, in response to low ligand concentrations, PPARgamma2 shows a quantitatively greater ability to induce adipogenesis. Analyses involving coactivator binding and transcriptional assays indicate that PPARgamma2 has an enhanced ability to bind components of the DRIP/TRAP complex, coactivators required for fat differentiation.


Subject(s)
Adipocytes/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Mediator Complex Subunit 1 , Mice , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Thyroid Hormone/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...