1
|
Cure of Alzheimer's Dementia Requires Addressing All of the Affected Brain Cell Types. J Clin Med 2023; 12:jcm12052049. [PMID: 36902833 PMCID: PMC10004473 DOI: 10.3390/jcm12052049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Multiple genetic, metabolic, and environmental abnormalities are known to contribute to the pathogenesis of Alzheimer's dementia (AD). If all of those abnormalities were addressed it should be possible to reverse the dementia; however, that would require a suffocating volume of drugs. Nevertheless, the problem may be simplified by using available data to address, instead, the brain cells whose functions become changed as a result of the abnormalities, because at least eleven drugs are available from which to formulate a rational therapy to correct those changes. The affected brain cell types are astrocytes, oligodendrocytes, neurons, endothelial cells/pericytes, and microglia. The available drugs include clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole. This article describes the ways by which the individual cell types contribute to AD's pathogenesis and how each of the drugs corrects the changes in the cell types. All five of the cell types may be involved in the pathogenesis of AD; of the 11 drugs, fingolimod, fluoxetine, lithium, memantine, and pioglitazone, each address all five of the cell types. Fingolimod only slightly addresses endothelial cells, and memantine is the weakest of the remaining four. Low doses of either two or three drugs are suggested in order to minimize the likelihood of toxicity and drug-drug interactions (including drugs used for co-morbidities). Suggested two-drug combinations are pioglitazone plus lithium and pioglitazone plus fluoxetine; a three-drug combination could add either clemastine or memantine. Clinical trials are required to validate that the suggest combinations may reverse AD.
Collapse
|
2
|
Fessel J. Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD). J Clin Med 2023; 12:1680. [PMID: 36836215 PMCID: PMC9967886 DOI: 10.3390/jcm12041680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Used as a supplement to psychotherapy, pharmacotherapy that addresses all of the known metabolic and genetic contributions to the pathogenesis of psychiatric conditions caused by stressors would require an inordinate number of drugs. Far simpler is to address the abnormalities caused by those metabolic and genetic changes in the cell types of the brain that mediate the behavioral abnormality. Relevant data regarding the changed brain cell types are described in this article and are derived from subjects with the paradigmatic behavioral abnormality of PTSD and from subjects with traumatic brain injury or chronic traumatic encephalopathy. If this analysis is correct, then therapy is required that benefits all of the affected brain cell types; those are astrocytes, oligodendrocytes, synapses and neurons, endothelial cells, and microglia (the pro-inflammatory (M1) subtype requires switching to the anti-inflammatory (M2) subtype). Combinations are advocated using several drugs, erythropoietin, fluoxetine, lithium, and pioglitazone, that benefit all of the five cell types, and that should be used to form a two-drug combination, suggested as pioglitazone with either fluoxetine or lithium. Clemastine, fingolimod, and memantine benefit four of the cell types, and one chosen from those could be added to the two-drug combination to form a three-drug combination. Using low doses of chosen drugs will limit both toxicity and drug-drug interactions. A clinical trial is required to validate both the advocated concept and the choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA 94123, USA
| |
Collapse
|
3
|
Fessel J. Formulating treatment of major psychiatric disorders: algorithm targets the dominantly affected brain cell-types. DISCOVER MENTAL HEALTH 2023; 3:3. [PMID: 37861813 PMCID: PMC10501034 DOI: 10.1007/s44192-022-00029-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 10/21/2023]
Abstract
BACKGROUND Pharmacotherapy for most psychiatric conditions was developed from serendipitous observations of benefit from drugs prescribed for different reasons. An algorithmic approach to formulating pharmacotherapy is proposed, based upon which combination of changed activities by brain cell-types is dominant for any particular condition, because those cell-types contain and surrogate for genetic, metabolic and environmental information, that has affected their function. The algorithm performs because functions of some or all the affected cell-types benefit from several available drugs: clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole PROCEDURES/FINDINGS: Bipolar disorder, major depressive disorder, schizophrenia, Alzheimer's disease, and post-traumatic stress disorder, illustrate the algorithm; for them, literature reviews show that no single combination of altered cell-types accounts for all cases; but they identify, for each condition, which combination occurs most frequently, i.e., dominates, as compared with other possible combinations. Knowing the dominant combination of altered cell-types in a particular condition, permits formulation of therapy with combinations of drugs taken from the above list. The percentage of patients who might benefit from that therapy, depends upon the frequency with which the dominant combination occurs in patients with that particular condition. CONCLUSIONS Knowing the dominant combination of changed cell types in psychiatric conditions, permits an algorithmically formulated, rationally-based treatment. Different studies of the same condition often produce discrepant results; all might be correct, because identical clinical phenotypes result from different combinations of impaired cell-types, thus producing different results. Clinical trials would validate both the proposed concept and choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA, 94123, USA.
| |
Collapse
|
4
|
Saha PS, Mayhan WG. Prenatal exposure to alcohol: mechanisms of cerebral vascular damage and lifelong consequences. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10818. [PMID: 38390614 PMCID: PMC10880760 DOI: 10.3389/adar.2022.10818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/01/2022] [Indexed: 02/24/2024]
Abstract
Alcohol is a well-known teratogen, and prenatal alcohol exposure (PAE) leads to a greater incidence of many cardiovascular-related pathologies. Alcohol negatively impacts vasculogenesis and angiogenesis in the developing fetal brain, resulting in fetal alcohol spectrum disorders (FASD). Ample preclinical evidence indicates that the normal reactivity of cerebral resistance arterioles, which regulate blood flow distribution in response to metabolic demand (neurovascular coupling), is impaired by PAE. This impairment of dilation of cerebral arteries may carry implications for the susceptibility of the brain to cerebral ischemic damage well into adulthood. The focus of this review is to consolidate findings from studies examining the influence of PAE on vascular development, give insights into relevant pathological mechanisms at the vascular level, evaluate the risks of ethanol-driven alterations of cerebrovascular reactivity, and revisit different preventive interventions that may have promise in reversing vascular changes in preclinical FASD models.
Collapse
Affiliation(s)
- Partha S Saha
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
5
|
Saha PS, Kim Sawtelle KR, Bamberg BN, Arrick DM, Watt MJ, Scholl JL, Zheng H, Mayhan WG. Rosiglitazone restores nitric oxide synthase-dependent reactivity of cerebral arterioles in rats exposed to prenatal alcohol. Alcohol Clin Exp Res 2021; 45:1359-1369. [PMID: 34120346 DOI: 10.1111/acer.14634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Prenatal exposure to alcohol leads to a greater incidence of many cardiovascular-related diseases, presumably via a mechanism that may involve increased oxidative stress. An agonist of peroxisome proliferator-activated receptor gamma (PPARγ; rosiglitazone) has been shown to suppress alcohol-induced neuroinflammation and oxidative stress. The goal of this study was to determine whether acute and chronic treatment with rosiglitazone could restore or prevent impaired nitric oxide synthase (NOS)-dependent responses of cerebral arterioles in male and female adult (14-16 weeks old) rats exposed to alcohol in utero. METHODS We fed Sprague-Dawley dams a liquid diet with or without 3% ethanol for the duration of their pregnancy (21-23 days). In the first series of studies, we examined the reactivity of cerebral arterioles to eNOS- (ADP), nNOS-dependent (NMDA), and NOS-independent agonists in male and female adult rats before and during acute (1 hour) topical application of rosiglitazone (1 µM). In a second series of studies, we examined the influence of chronic treatment with rosiglitazone (3 mg/kg/day in drinking water for 2-3 weeks) on the responses of cerebral arterioles in male and female adult rats exposed to alcohol in utero. RESULTS We found that in utero exposure to alcohol similarly reduced responses of cerebral arterioles to ADP and NMDA, but not to nitroglycerin in male and female adult rats. In addition, acute treatment of the male and female adult rats with rosiglitazone similarly restored this impairment in cerebral vascular function to that observed in controls. We also found that chronic treatment with rosiglitazone prevented impaired vascular function in male and female adult rats that were exposed to alcohol in utero. CONCLUSIONS PPARγ activation may be an effective and relevant treatment to reverse or prevent cerebral vascular abnormalities associated with prenatal exposure to alcohol.
Collapse
Affiliation(s)
- Partha S Saha
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kirsten R Kim Sawtelle
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Brittany N Bamberg
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Denise M Arrick
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Michael J Watt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jamie L Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
6
|
Ciavarella C, Motta I, Valente S, Pasquinelli G. Pharmacological (or Synthetic) and Nutritional Agonists of PPAR-γ as Candidates for Cytokine Storm Modulation in COVID-19 Disease. Molecules 2020; 25:molecules25092076. [PMID: 32365556 PMCID: PMC7248959 DOI: 10.3390/molecules25092076] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/25/2022] Open
Abstract
The cytokine storm is an abnormal production of inflammatory cytokines, due to the over-activation of the innate immune response. This mechanism has been recognized as a critical mediator of influenza-induced lung disease, and it could be pivotal for COVID-19 infections. Thus, an immunomodulatory approach targeting the over-production of cytokines could be proposed for viral aggressive pulmonary disease treatment. In this regard, the peroxisome proliferator-activated receptor (PPAR)-γ, a member of the PPAR transcription factor family, could represent a potential target. Beside the well-known regulatory role on lipid and glucose metabolism, PPAR-γ also represses the inflammatory process. Similarly, the PPAR-γ agonist thiazolidinediones (TZDs), like pioglitazone, are anti-inflammatory drugs with ameliorating effects on severe viral pneumonia. In addition to the pharmacological agonists, also nutritional ligands of PPAR-γ, like curcuma, lemongrass, and pomegranate, possess anti-inflammatory properties through PPAR-γ activation. Here, we review the main synthetic and nutritional PPAR-γ ligands, proposing a dual approach based on the strengthening of the immune system using pharmacological and dietary strategies as an attempt to prevent/treat cytokine storm in the case of coronavirus infection.
Collapse
|
7
|
Yang BR, Yuen SC, Fan GY, Cong WH, Leung SW, Lee SMY. Identification of certain Panax species to be potential substitutes for Panax notoginseng in hemostatic treatments. Pharmacol Res 2018; 134:1-15. [DOI: 10.1016/j.phrs.2018.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 04/19/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
|
8
|
Liu C, Xing J, Gao Y. UNBS5162 inhibits the proliferation of human A549 non-small-cell lung cancer cells by promoting apoptosis. Thorac Cancer 2018; 9:105-111. [PMID: 29130641 PMCID: PMC5754305 DOI: 10.1111/1759-7714.12546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Lung cancer is one of the most frequently diagnosed malignancies in the world, thus developing novel anticancer reagents for lung cancer treatment is critical. METHODS We performed cell counting kit-8 and cell colony formation assays to investigate the role of UNBS5162 in the proliferation of A549 cells. Invasion and migration assays were applied to study the inhibitory effect of UNBS5162 on non-small cell lung cancer cells. To detect the effect of UNBS5162 on A549 cell apoptosis, Annexin-V fluorescein isothiocyanate and propidium iodide staining methods were used. Protein expression was analyzed using Western blot assay. RESULTS UNBS5162 not only inhibited proliferation but also decreased invasion and migration in A549 cells. Most cells were intact (96.93%) under control conditions, but the number of intact cells decreased (84.8%) after 24 hours of treatment with UNBS5162, and the number of early and late apoptotic cells significantly increased (P < 0.05). Anti-apoptotic protein Bcl-2 expression in the UNBS5162 group was significantly decreased (P < 0.05), and expression of proapoptotic proteins Bim, Bax, and active caspase-3 were significantly increased (P < 0.05) compared to the control. In the PI3K signaling pathway, phospo-AKT and phospo-mTOR levels were significantly decreased (P < 0.05), while S6K and Cyclin D1 protein levels were significantly decreased in UNBS5162 treated A549 cells (P < 0.05). CONCLUSION These findings suggest that UNBS5162 could inhibit A549 cell proliferation and metastasis by inhibiting PI3K pathway mediated apoptosis.
Collapse
Affiliation(s)
- Cuicui Liu
- Department of OncologyLinyi City People's HospitalLinyiChina
| | - Jiaqiang Xing
- Department of Thoracic SurgeryLinyi Cancer HospitalLinyiChina
| | - Yujun Gao
- Department of Thoracic SurgeryAffiliated Hospital of Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
9
|
Finiuk N, Boiko N, Klyuchivska O, Коbylinska L, Kril I, Zimenkovsky B, Lesyk R, Stoika R. 4-Thiazolidinone derivative Les-3833 effectively inhibits viability of human melanoma cells through activating apoptotic mechanisms. Croat Med J 2017; 58:129-139. [PMID: 28409496 PMCID: PMC5410740 DOI: 10.3325/cmj.2017.58.129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aim To evaluate cytotoxic action of 4-thiazolidinone derivative Les-3833 and study the mechanisms of its pro-apoptotic action toward human melanoma cells and human tumor cell lines of other tissue origin. Methods The effect of Les-3833 or doxorubicin on the viability of 9 cell lines was studied using MTT assay, while human melanoma cells of WM793 line were additionally examined using light and fluorescent microscopies for evaluating cytomorphological changes. The Western-blot and flow cytometric analyses were carried out to study signaling pathways of melanoma cell cycling and death. Results Les-3833 was the most efficient against melanoma cells. Its half maximal inhibitory concentration (IC50) was 0.22 μg/mL for WM793 cells and 0.3 μg/mL for SK-Mel-28 melanoma cells. For human lung A549, breast MCF-7, colon HCT116, and ovarian SKOV3 carcinoma cell lines IC50 was in between 2.5 to >5.0 μg/mL. Les-3833 was relatively not toxic (IC50 ˃ 5 μg/mL) for human embryonic kidney HEK293 cells. Results of Annexin V/PI staining of melanoma cells and activation of caspase 3, PARP, MAPK, and EndoG protein suggest apoptosis in Les-3833-treated cells. Les-3833 also induced ROS production in melanoma cells and their arrest in G0/G1 phase of cell cycle. Conclusion Novel 4-thiazolidinone derivative Les-3833 is effective against human melanoma cells in vitro, and such effect is tumor specific since it is much less pronounced in human carcinoma and leukemia cells. In melanoma cells Les-3833 induces apoptosis (morphological changes and increased pro-apoptotic proteins), ROS production, and arrest in G0/G1 phase of cell cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rostyslav Stoika
- Rostyslav Stoika, Institute of Cell Biology, NAS of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine,
| |
Collapse
|
10
|
Kotlinowski J, Jozkowicz A. PPAR Gamma and Angiogenesis: Endothelial Cells Perspective. J Diabetes Res 2016; 2016:8492353. [PMID: 28053991 PMCID: PMC5174176 DOI: 10.1155/2016/8492353] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/22/2016] [Accepted: 11/01/2016] [Indexed: 12/24/2022] Open
Abstract
We summarize the current knowledge concerning PPARγ function in angiogenesis. We discuss the mechanisms of action for PPARγ and its role in vasculature development and homeostasis, focusing on endothelial cells, endothelial progenitor cells, and bone marrow-derived proangiogenic cells.
Collapse
Affiliation(s)
- Jerzy Kotlinowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- *Alicja Jozkowicz:
| |
Collapse
|
11
|
Peroxisome proliferator-activated receptor (PPAR) gamma in cardiovascular disorders and cardiovascular surgery. J Cardiol 2015; 66:271-8. [DOI: 10.1016/j.jjcc.2015.05.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/08/2015] [Accepted: 05/14/2015] [Indexed: 12/28/2022]
|
12
|
Thiazolidine-2,4-diones as multi-targeted scaffold in medicinal chemistry: Potential anticancer agents. Eur J Med Chem 2014; 87:814-33. [DOI: 10.1016/j.ejmech.2014.10.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/22/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022]
|
13
|
Popolo A, Adesso S, Pinto A, Autore G, Marzocco S. L-Arginine and its metabolites in kidney and cardiovascular disease. Amino Acids 2014; 46:2271-86. [PMID: 25161088 DOI: 10.1007/s00726-014-1825-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 08/13/2014] [Indexed: 12/19/2022]
Abstract
L-Arginine is a semi essential amino acid synthesised from glutamine, glutamate and proline via the intestinal-renal axis in humans and most mammals. L-Arginine degradation occurs via multiple pathways initiated by arginase, nitric-oxide synthase, Arg: glycine amidinotransferase, and Arg decarboxylase. These pathways produce nitric oxide, polyamines, proline, glutamate, creatine and agmatine with each having enormous biological importance. Several disease are associated to an L-arginine impaired levels and/or to its metabolites: in particular various L-arginine metabolites may participate in pathogenesis of kidney and cardiovascular disease. L-Arginine and its metabolites may constitute both a marker of pathology progression both the rationale for manipulating L-arginine metabolism as a strategy to ameliorate these disease. A large number of studies have been performed in experimental models of kidney disease with sometimes conflicting results, which underlie the complexity of Arg metabolism and our incomplete knowledge of all the mechanisms involved. Moreover several lines of evidence demonstrate the role of L-arg metabolites in cardiovascular disease and that L-arg administration role in reversing endothelial dysfunction, which is the leading cause of cardiovascular diseases, such as hypertension and atherosclerosis. This review will discuss the implication of the mains L-arginine metabolites and L-arginine-derived guanidine compounds in kidney and cardiovascular disease considering the more recent literature in the field.
Collapse
Affiliation(s)
- Ada Popolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, SA, Italy
| | | | | | | | | |
Collapse
|
14
|
Campia U, Matuskey LA, Tesauro M, Cardillo C, Panza JA. PPARγ activation does not affect endothelin activity in non-diabetic patients with hypertension or hypercholesterolemia. Atherosclerosis 2014; 234:436-40. [PMID: 24769306 DOI: 10.1016/j.atherosclerosis.2014.03.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/12/2014] [Accepted: 03/27/2014] [Indexed: 01/29/2023]
Abstract
OBJECTIVES This study tested the hypothesis that pioglitazone reduces endothelin-1 activity in the forearm vasculature in non-diabetic patients with hypertension or hypercholesterolemia and variable degrees of insulin resistance. METHODS We conducted a single center, randomized, double-blind, placebo controlled, cross-over trial in 80 patients with either hypertension or hypercholesterolemia and further classified as insulin-sensitive or insulin-resistant based on a published insulin sensitivity index. Participants received pioglitazone 45 mg daily or matching placebo for eight weeks. The main endpoint was the change in forearm vascular endothelin-1 activity, as assessed by intra-arterial infusion of the endothelin type A receptor blocker BQ-123, measured at the end of each 8-week treatment period. RESULTS Pioglitazone lowered plasma insulin (P < 0.001), improved insulin sensitivity (P < 0.001), increased HDL (P < 0.001), and reduced triglycerides (P = 0.003), free fatty acids (P = 0.005), and C-reactive protein (P = 0.001). However, pioglitazone did not affect the vasodilator response to BQ-123 in the whole group (P = 0.618) and in the diagnosis or insulin sensitivity subgroups. Hence, in non-diabetic patients with hypertension or hypercholesterolemia, PPARγ activation with pioglitazone does not affect endothelin-1 activity, despite enhancing insulin sensitivity and reducing plasma insulin and C-reactive protein levels. CONCLUSIONS In non-diabetic patients with hypertension or hypercholesterolemia, pioglitazone improves insulin sensitivity, lipid profile, and inflammation but does not affect endothelin activity. Our data suggest that the determinants of endothelin-1 vascular activity in vivo may differ and/or be more complex than those suggested by the results of previous in vitro studies.
Collapse
Affiliation(s)
- Umberto Campia
- MedStar Cardiovascular Research Network, MedStar Heart Institute, 110 Irving Street NW, Room NA-1041, Washington, DC 20010, USA.
| | - Linda A Matuskey
- MedStar Cardiovascular Research Network, MedStar Heart Institute, 110 Irving Street NW, Room NA-1041, Washington, DC 20010, USA
| | - Manfredi Tesauro
- System Medicine, University of Tor Vergata, Viale Oxford, 81, 00133 Rome, Italy
| | - Carmine Cardillo
- Internal Medicine, Catholic University, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Julio A Panza
- Westchester Medical Center, 100 Woods Road, Macy Pavilion, Room 102, Valhalla, NY, USA
| |
Collapse
|
15
|
Ochodnicky P, Mesarosova L, Cernecka H, Klimas J, Krenek P, Goris M, van Dokkum RPE, Henning RH, Kyselovic J. Pioglitazone, a PPARγ agonist, provides comparable protection to angiotensin converting enzyme inhibitor ramipril against adriamycin nephropathy in rat. Eur J Pharmacol 2014; 730:51-60. [PMID: 24582928 DOI: 10.1016/j.ejphar.2014.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 11/26/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) agonists have been shown to ameliorate diabetic nephropathy, but much less are known about their effects in non-diabetic nephropathies. In the present study, metabolic parameters, blood pressure, aortic endothelial function along with molecular and structural markers of glomerular and tubulointerstitial renal damage, were studied in a rat model of normotensive nephropathy induced by adriamycin and treated with PPARγ agonist pioglitazone (12mg/kg, po), angiotensin converting enzyme (ACE) inhibitor ramipril (1mg/kg, po) or their combination. Pioglitazone had no effect on systolic blood pressure, marginally reduced glycemia and improved aortic endothelium-dependent relaxation. In the kidney, pioglitazone prevented the development of proteinuria and focal glomerulosclerosis to the similar extent as blood-pressure lowering ramipril. Renoprotection provided by either treatment was associated with a reduction in the cortical expression of profibrotic plasminogen activator inhibitor-1 and microvascular damage-inducing endothelin-1, and a limitation of interstitial macrophage influx. Treatment with PPARγ agonist, as well as ACE inhibitor comparably affected renal expression of the renin-angiotensin system (RAS) components, normalizing increased renal expression of ACE and enhancing the expression of Mas receptor. Interestingly, combined pioglitazone and ramipril treatment did not provide any additional renoprotection. These results demonstrate that in a nondiabetic renal disease, such as adriamycin-induced nephropathy, PPARγ agonist pioglitazone provides renoprotection to a similar extent as an ACE inhibitor by interfering with the expression of local RAS components and attenuating related profibrotic and inflammatory mechanisms. The combination of the both agents, however, does not lead to any additional renal benefit.
Collapse
Affiliation(s)
- Peter Ochodnicky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic.
| | - Lucia Mesarosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic
| | - Hana Cernecka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic
| | - Maaike Goris
- Department of Clinical Pharmacology, University Medical Center Groningen (UMCG) and Groningen Institute for Drug Exploration (GUIDE), University of Groningen, The Netherlands
| | - Richard P E van Dokkum
- Department of Clinical Pharmacology, University Medical Center Groningen (UMCG) and Groningen Institute for Drug Exploration (GUIDE), University of Groningen, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacology, University Medical Center Groningen (UMCG) and Groningen Institute for Drug Exploration (GUIDE), University of Groningen, The Netherlands
| | - Jan Kyselovic
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Slovak Republic
| |
Collapse
|
16
|
Bionaz M, Chen S, Khan MJ, Loor JJ. Functional Role of PPARs in Ruminants: Potential Targets for Fine-Tuning Metabolism during Growth and Lactation. PPAR Res 2013; 2013:684159. [PMID: 23737762 PMCID: PMC3657398 DOI: 10.1155/2013/684159] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 12/31/2022] Open
Abstract
Characterization and biological roles of the peroxisome proliferator-activated receptor (PPAR) isotypes are well known in monogastrics, but not in ruminants. However, a wealth of information has accumulated in little more than a decade on ruminant PPARs including isotype tissue distribution, response to synthetic and natural agonists, gene targets, and factors affecting their expression. Functional characterization demonstrated that, as in monogastrics, the PPAR isotypes control expression of genes involved in lipid metabolism, anti-inflammatory response, development, and growth. Contrary to mouse, however, the PPARγ gene network appears to controls milk fat synthesis in lactating ruminants. As in monogastrics, PPAR isotypes in ruminants are activated by long-chain fatty acids, therefore, making them ideal candidates for fine-tuning metabolism in this species via nutrients. In this regard, using information accumulated in ruminants and monogastrics, we propose a model of PPAR isotype-driven biological functions encompassing key tissues during the peripartal period in dairy cattle.
Collapse
Affiliation(s)
- Massimo Bionaz
- Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97330, USA
| | - Shuowen Chen
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Muhammad J. Khan
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Juan J. Loor
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
17
|
Sheng CC, Hong CC. Mixing of the Old With the New. Arterioscler Thromb Vasc Biol 2012; 32:2337-8. [DOI: 10.1161/atvbaha.112.300217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Calvin C. Sheng
- From the Division of Cardiovascular Medicine (C.C.S., C.C.H.) and Departments of Medicine, Pharmacology, and Cell and Developmental Biology (C.C.H.), Vanderbilt University School of Medicine, Nashville, TN; and Research Medicine, Veterans Affairs TVHS, Nashville, TN (C.C.H.)
| | - Charles C. Hong
- From the Division of Cardiovascular Medicine (C.C.S., C.C.H.) and Departments of Medicine, Pharmacology, and Cell and Developmental Biology (C.C.H.), Vanderbilt University School of Medicine, Nashville, TN; and Research Medicine, Veterans Affairs TVHS, Nashville, TN (C.C.H.)
| |
Collapse
|
18
|
Nagahama R, Matoba T, Nakano K, Kim-Mitsuyama S, Sunagawa K, Egashira K. Nanoparticle-Mediated Delivery of Pioglitazone Enhances Therapeutic Neovascularization in a Murine Model of Hindlimb Ischemia. Arterioscler Thromb Vasc Biol 2012; 32:2427-34. [DOI: 10.1161/atvbaha.112.253823] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Critical limb ischemia is a severe form of peripheral artery disease (PAD) for which neither surgical revascularization nor endovascular therapy nor current medicinal therapy has sufficient therapeutic effects. Peroxisome proliferator activated receptor-γ agonists present angiogenic activity in vitro; however, systemic administration of peroxisome proliferator−activated receptor-γ agonists is hampered by its side effects, including heart failure. Here, we demonstrate that the nanoparticle (NP)-mediated delivery of the peroxisome proliferator activated receptor-γ agonist pioglitazone enhances its therapeutic efficacy on ischemia-induced neovascularization in a murine model.
Methods and Results—
In a nondiabetic murine model of hindlimb ischemia, a single intramuscular injection of pioglitazone-incorporated NP (1 µg/kg) into ischemic muscles significantly improved the blood flow recovery in the ischemic limbs, significantly increasing the number of CD31-positive capillaries and α-smooth muscle actin–positive arterioles. The therapeutic effects of pioglitazone-incorporated NP were diminished by the peroxisome proliferator activated receptor-γ antagonist GW9662 and were not observed in endothelial NO synthase–deficient mice. Pioglitazone-incorporated NP induced endothelial NO synthase phosphorylation, as demonstrated by Western blot analysis, as well as expression of multiple angiogenic growth factors in vivo, including vascular endothelial growth factor-A, vascular endothelial growth factor-B, and fibroblast growth factor-1, as demonstrated by real-time polymerase chain reaction. Intramuscular injection of pioglitazone (1 µg/kg) was ineffective, and oral administration necessitated a >500 μg/kg per day dose to produce therapeutic effects equivalent to those of pioglitazone-incorporated NP.
Conclusion—
NP-mediated drug delivery is a novel modality that may enhance the effectiveness of therapeutic neovascularization, surpassing the effectiveness of current treatments for peripheral artery disease with critical limb ischemia.
Collapse
Affiliation(s)
- Ryoji Nagahama
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| | - Tetsuya Matoba
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| | - Kaku Nakano
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| | - Shokei Kim-Mitsuyama
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| | - Kenji Sunagawa
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| | - Kensuke Egashira
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (R.N., T.M., K.S.); Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (K.N., K.E.); and Department of Pharmacology and Molecular Therapeutics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.K.-M.)
| |
Collapse
|
19
|
Green DE, Murphy TC, Kang BY, Kleinhenz JM, Szyndralewiez C, Page P, Sutliff RL, Hart CM. The Nox4 inhibitor GKT137831 attenuates hypoxia-induced pulmonary vascular cell proliferation. Am J Respir Cell Mol Biol 2012; 47:718-26. [PMID: 22904198 DOI: 10.1165/rcmb.2011-0418oc] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Increased NADP reduced (NADPH) oxidase 4 (Nox4) and reduced expression of the nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) contribute to hypoxia-induced pulmonary hypertension (PH). To examine the role of Nox4 activity in pulmonary vascular cell proliferation and PH, the current study used a novel Nox4 inhibitor, GKT137831, in hypoxia-exposed human pulmonary artery endothelial or smooth muscle cells (HPAECs or HPASMCs) in vitro and in hypoxia-treated mice in vivo. HPAECs or HPASMCs were exposed to normoxia or hypoxia (1% O(2)) for 72 hours with or without GKT137831. Cell proliferation and Nox4, PPARγ, and transforming growth factor (TGF)β1 expression were measured. C57Bl/6 mice were exposed to normoxia or hypoxia (10% O(2)) for 3 weeks with or without GKT137831 treatment during the final 10 days of exposure. Lung PPARγ and TGF-β1 expression, right ventricular hypertrophy (RVH), right ventricular systolic pressure (RVSP), and pulmonary vascular remodeling were measured. GKT137831 attenuated hypoxia-induced H(2)O(2) release, proliferation, and TGF-β1 expression and blunted reductions in PPARγ in HPAECs and HPASMCs in vitro. In vivo GKT137831 inhibited hypoxia-induced increases in TGF-β1 and reductions in PPARγ expression and attenuated RVH and pulmonary artery wall thickness but not increases in RVSP or muscularization of small arterioles. This study shows that Nox4 plays a critical role in modulating proliferative responses of pulmonary vascular wall cells. Targeting Nox4 with GKT137831 provides a novel strategy to attenuate hypoxia-induced alterations in pulmonary vascular wall cells that contribute to vascular remodeling and RVH, key features involved in PH pathogenesis.
Collapse
Affiliation(s)
- David E Green
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
INTRODUCTION Thiazolidinediones (TZDs) initially showed great promise as unique receptor-mediated oral therapy for type 2 diabetes, but a host of serious side effects, primarily cardiovascular, have limited their utility. It is crucial at this point to perform a risk-benefit analysis to determine what role TZDs should play in our current treatment of type 2 diabetes and where the future of this class of drugs is headed. AREAS COVERED This review provides a comprehensive overview of the literature from 2000 onward reporting the known side effects of rosiglitazone and pioglitazone, with commentary on the quality of the data available, putative mechanism of each side effect and clinical significance. Finally, a perspective on the future of the TZDs as a class is provided. EXPERT OPINION The current TZDs are first-generation, non-specific activators of peroxisome proliferator-activated receptor (PPAR) gamma, resulting in a wide array of deleterious side effects that currently limit their use. However, the development of highly targeted selective PPAR gamma modulators (SPPARγMs) and dual PPAR gamma/alpha agonists is on the horizon.
Collapse
Affiliation(s)
- Jacqueline Kung
- Tufts University, Division of Endocrinology, Diabetes and Metabolism, 800 Washington St #268, Boston, MA 02111, USA.
| | | |
Collapse
|
21
|
Fujii M, Inoki I, Saga M, Morikawa N, Arakawa KI, Inaba S, Yoshioka K, Konoshita T, Miyamori I. Aldosterone inhibits endothelial morphogenesis and angiogenesis through the downregulation of vascular endothelial growth factor receptor-2 expression subsequent to peroxisome proliferator-activated receptor gamma. J Steroid Biochem Mol Biol 2012; 129:145-52. [PMID: 22212769 DOI: 10.1016/j.jsbmb.2011.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/16/2011] [Accepted: 12/18/2011] [Indexed: 12/23/2022]
Abstract
Angiogenesis plays a pivotal role in cardiovascular diseases such as ischemic heart disease, limb ischemia and heart failure, and has recently been shown to mediate various biological activities related to the pathogenesis of these diseases. In the present study, we evaluated the role of aldosterone in angiogenesis. Tube formation assay on Matrigel using human umbilical vein endothelial cells (HUVEC) revealed that aldosterone inhibited endothelial morphogenesis in a manner sensitive to eplerenone, a selective mineralocorticoid receptor antagonist. The anti-angiogenic effect of aldosterone was further confirmed by an in vivo angiogenesis assay using a Matrigel plug model in mice. Reverse transcription-mediated polymerase chain reaction and immunoblotting demonstrated that aldosterone downregulated the expression levels of vascular endothelial growth factor receptor-2 (VEGFR-2) and peroxisome proliferators-activated receptor gamma (PPAR gamma). VEGFR-2 expression was found to be enhanced in response to PPAR gamma activation by troglitazone, and attenuated by GW9662, a specific antagonist of PPAR gamma. In the tube formation assay, endothelial morphogenesis was stimulated by troglitazone, and inhibited by GW9662, indicating that PPAR gamma activation mediates positive regulation of angiogenesis through enhancement of VEGFR-2 expression. These data suggest that aldosterone inhibits angiogenesis through VEGFR-2 downregulation, subsequent to, at least in part, attenuation of PPAR gamma expression. The present findings provide a new insight into the possible therapeutic application of mineralocorticoid receptor blockade to various cardiovascular diseases.
Collapse
Affiliation(s)
- Miki Fujii
- Third Department of Internal Medicine, Faculty of Medical Sciences, Fukui University, 23-3 Matsuokashimoaizuki, Yoshida-gun, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Magri CJ, Gatt N, Xuereb RG, Fava S. Peroxisome proliferator-activated receptor-γ and the endothelium: implications in cardiovascular disease. Expert Rev Cardiovasc Ther 2012; 9:1279-94. [PMID: 21985541 DOI: 10.1586/erc.11.140] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Peroxisome proliferator-activated receptors-γ (PPARγs) are ligand-activated transcription factors that play a crucial regulatory role in the transcription of a large number of genes involved in lipid metabolism and inflammation. In addition to physiological ligands, synthetic ligands (the thiazoledinediones) have been developed. In spite of the much publicized adverse cardiovascular effects of one such thiazoledinedione (rosiglitazone), PPARγ activation may have beneficial cardiovascular effects. In this article we review the effects of PPARγ activation on the endothelium with special emphasis on the possible implications in cardiovascular disease. We discuss its possible role in inflammation, vasomotor function, thrombosis, angiogenesis, vascular aging and vascular rhythm. We also briefly review the clinical implications of these lines of research.
Collapse
Affiliation(s)
- Caroline Jane Magri
- Department of Cardiac Services, Mater Dei Hospital, Tal-Qroqq, Msida MSD 2090, Malta
| | | | | | | |
Collapse
|
23
|
Vecchiet J, Ucciferri C, Falasca K, Mancino P, Di Iorio A, De Caterina R. Antihypertensive and metabolic effects of telmisartan in hypertensive HIV-positive patients. Antivir Ther 2012; 16:639-45. [PMID: 21817185 DOI: 10.3851/imp1809] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Hypertension is more prevalent among HIV-infected individuals than in the general population and contributes to increased cardiovascular risk. The angiotensin II receptor blocker telmisartan is also a partial peroxisome proliferator activated receptor-γ agonist with documented effects on glucose and lipid homeostasis. The aim of this study was to evaluate the antihypertensive and metabolic effects of telmisartan in hypertensive HIV-positive patients. METHODS A total of 18 HIV-positive men treated with antiretroviral therapy and recently diagnosed with hypertension were administered 80 mg telmisartan daily. Systolic blood pressure (SBP) and diastolic blood pressure (DBP), viroimmunological and metabolic parameters, insulin resistance, C-reactive protein, microalbuminuria, cystatin C and plasma levels of interleukin-18 and endothelin-1 were measured at baseline (T0), 1 month (T1), 3 months (T3) and 6 months (T6). RESULTS Treatment with telmisartan not only decreased SBP and DBP levels, but also improved insulin resistance and microalbuminuria by T1. Levels of triglycerides significantly decreased and high-density lipoprotein cholesterol increased at T1, whereas total and low-density lipoprotein cholesterol levels were statistically reduced at T3 and T6. Cystatin C and endothelin-1 showed a significant reduction at T1, whereas interleukin-18 decreased at both T3 and T6. CONCLUSIONS Telmisartan was effective in reducing blood pressure and improving lipid metabolism and renal function. Reduction of endothelin-1 might be related to an endothelial protective effect. On the basis of these findings, and because of properties unrelated to blood pressure lowering, telmisartan might be the first choice antihypertensive drug for the treatment of HIV-positive patients.
Collapse
Affiliation(s)
- Jacopo Vecchiet
- Division of Infectious Diseases, Department of Medicine and Sciences of Aging, G d'Annunzio University School of Medicine, Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
24
|
Green DE, Sutliff RL, Hart CM. Is peroxisome proliferator-activated receptor gamma (PPARγ) a therapeutic target for the treatment of pulmonary hypertension? Pulm Circ 2011; 1:33-47. [PMID: 21547012 PMCID: PMC3085428 DOI: 10.4103/2045-8932.78101] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH), a progressive disorder associated with significant morbidity and mortality, is caused by complex pathways that culminate in structural and functional alterations of the pulmonary circulation and increases in pulmonary vascular resistance and pressure. Diverse genetic, pathological, or environmental triggers stimulate PH pathogenesis culminating in vasoconstriction, cell proliferation, vascular remodeling, and thrombosis. We conducted a thorough literature review by performing MEDLINE searches via PubMed to identify articles pertaining to PPARγ as a therapeutic target for the treatment of PH. This review examines basic and preclinical studies that explore PPARγ and its ability to regulate PH pathogenesis. Despite the current therapies that target specific pathways in PH pathogenesis, including prostacyclin derivatives, endothelin-receptor antagonists, and phosphodiesterase type 5 inhibitors, morbidity and mortality related to PH remain unacceptably high, indicating the need for novel therapeutic approaches. Consequently, therapeutic targets that simultaneously regulate multiple pathways involved in PH pathogenesis have gained attention. This review focuses on peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors. While the PPARγ receptor is best known as a master regulator of lipid and glucose metabolism, a growing body of literature demonstrates that activation of PPARγ exerts antiproliferative, antithrombotic, and vasodilatory effects on the vasculature, suggesting its potential efficacy as a PH therapeutic target.
Collapse
Affiliation(s)
- David E. Green
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Roy L. Sutliff
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - C. Micheal Hart
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
25
|
Longo-Mbenza B, Kasiam Lasi On'kin JB, Nge Okwe A, Kangola Kabangu N. The metabolic syndrome in a Congolese population and its implications for metabolic syndrome definitions. Diabetes Metab Syndr 2011; 5:17-24. [PMID: 22814836 DOI: 10.1016/j.dsx.2010.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIM Metabolic syndrome defined by International cut-off values are limited to detect people at high cardiometabolic risk in Central Africans in comparison with metabolic syndrome defined by ethnic-specific definition. We examined the relationship between metabolic syndromes, diabetes control, abdominal obesity, HDL-cholesterol groups and atherosclerotic complications. MATERIALS AND METHODS A representative sample of type-2 diabetic central Africans from Kinshasa were studied. Outcome measures included control of diabetes, atherosclerosis, abdominal obesity, insulin resistance, total cholesterol, triglycerides, HDL-cholesterol, metabolic syndromes and atherosclerosis. RESULTS Of 1266 type-2 diabetic patients (48.8%), (61.8%), (27.1%) and (81%) had uncontrolled diabetes, atherosclerotics, metabolic syndrome (IDF/Europe), and metabolic syndrome (IDF/local) respectively. There was a significant U-shaped relationship between atherosclerotics complications, insulin resistance, delta postprandial glycaemia and HDL-cholesterol stratification. There was also a significant U-shaped relationship between cardiometabolic risk (P<0.01) and atherosclerotic complications. CONCLUSION Type-2 diabetic Central Africans exhibit very high rates of uncontrolled diabetes, atherosclerotic complications and metabolic syndrome. Both, abdominal obesity, insulin resistance, low and very high HDL-cholesterol levels are cardiometabolic risk factors.
Collapse
Affiliation(s)
- B Longo-Mbenza
- Faculty of Health Sciences, Walter Sisulu University, Mthatha, South Africa.
| | | | | | | |
Collapse
|
26
|
Mendonça MC, Koles N, Doi SQ, Sellitti DF. Transforming growth factor-β1 regulation of C-type natriuretic peptide expression in human vascular smooth muscle cells: dependence on TSC22D1. Am J Physiol Heart Circ Physiol 2010; 299:H2018-27. [DOI: 10.1152/ajpheart.00656.2010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
C-type natriuretic peptide (CNP) possesses nitric oxide-like signaling mechanisms and actions in the vasculature, including the inhibition of fibrosis and vascular remodeling through counterregulation of transforming growth factor-β (TGF-β) signaling. The leucine zipper protein transforming growth factor stimulated clone 22 domain 1 (TSC22D1), cloned via its presumed binding to a GC-rich element in the CNP promoter, was the first protein to be described as a CNP transcription factor, but the lack of supporting evidence since its discovery and its lack of a classical DNA-binding site have left in question its role in the regulation of CNP by TGF-β and other factors. To define a specific role for TSC22D1 in CNP transcription, we have examined the effects of the profibrotic growth factors TGF-β1 and PDGF-BB on CNP mRNA expression in cultured human vascular smooth muscle cells (SMC) in which TSC22D1 expression was suppressed with small interfering RNA. Results showed that TGF-β and PDGF-BB significantly increased CNP expression in all three SMC types. Twenty-four-hour TGF-β-induced elevations in CNP were strongly correlated with changes in TSC22D1 mRNA levels, and both genes exhibited their greatest response to TGF-β1 in coronary artery SMC. Furthermore, siRNA suppression of TSC22D1 expression in coronary artery and aortic SMC by ∼90% resulted in 45–65% reductions of both PDGF- and TGF-β-stimulated CNP expression, respectively. These results support a postulated role of TSC22D1 as an enhancer of CNP transcription and suggest that TGF-β-induced upregulation of CNP expression in SMC may be mediated in part by increased transcription of TSC22D1.
Collapse
Affiliation(s)
- Maria C. Mendonça
- Department of Medicine, Divisions of Endocrinology and Nephrology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Nancy Koles
- Department of Medicine, Divisions of Endocrinology and Nephrology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sonia Q. Doi
- Department of Medicine, Divisions of Endocrinology and Nephrology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Donald F. Sellitti
- Department of Medicine, Divisions of Endocrinology and Nephrology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
27
|
Kim EK, Lee JH, Oh YM, Lee YS, Lee SD. Rosiglitazone attenuates hypoxia-induced pulmonary arterial hypertension in rats. Respirology 2010; 15:659-68. [PMID: 20546541 DOI: 10.1111/j.1440-1843.2010.01756.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Expression of peroxisome proliferator-activated receptor gamma (PPARgamma) is decreased in the lungs of patients with pulmonary hypertension, and PPARgamma ligands have been associated with the release of vasoactive substances from vascular endothelial cells and prevention of vascular remodelling. We hypothesized that PPARgamma may play a critical role in the development of pulmonary hypertension induced by chronic hypoxia. METHODS Male adult Sprague-Dawley rats were exposed to normoxia, normoxia and rosiglitazone (8 mg/kg orally, 5 days/week), hypoxia (12% inspired O(2) fraction), or hypoxia and rosiglitazone for 4 weeks. On the last day of the fourth week, pulmonary arterial pressure was measured and morphological changes in pulmonary vessels were assessed. The expression of PPARgamma, endothelin (ET)-1 and vascular endothelial growth factor (VEGF) was also analysed. RESULTS Rosiglitazone inhibited the development of pulmonary hypertension, and pulmonary vascular remodelling induced by chronic hypoxia. PPARgamma expression was decreased and expression of ET-1 and VEGF was increased in lung tissues of the hypoxia group. Rosiglitazone treatment prevented the hypoxia-induced reduction in PPARgamma expression, and restored ET-1 and VEGF expression almost to the levels of the normoxia group. CONCLUSIONS Rosiglitazone inhibited the development of pulmonary hypertension induced by chronic hypoxia, perhaps by reversing the changes in PPARgamma, ET-1 and VEGF expression induced by hypoxia. These findings indicate that rosiglitazone may be beneficial in the treatment of chronic hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Eun Kyung Kim
- Department of Pulmonary and Critical Care Medicine, Bundang CHA Hospital, College of Medicine, CHA University, Seongnam, Korea
| | | | | | | | | |
Collapse
|
28
|
Sutliff RL, Kang BY, Hart CM. PPARgamma as a potential therapeutic target in pulmonary hypertension. Ther Adv Respir Dis 2010; 4:143-60. [PMID: 20530063 DOI: 10.1177/1753465809369619] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder of the pulmonary circulation associated with significant morbidity and mortality. The pathobiology of PH involves a complex series of derangements causing endothelial dysfunction, vasoconstriction and abnormal proliferation of pulmonary vascular wall cells that lead to increases in pulmonary vascular resistance and pressure. Recent evidence indicates that the ligand-activated transcription factor, peroxisome proliferator-activated receptor gamma (PPARgamma) can have a favorable impact on a variety of pathways involved in the pathogenesis of PH. This review summarizes PPARgamma biology and the emerging evidence that therapies designed to activate this receptor may provide novel approaches to the treatment of PH. Mediators of PH that are regulated by PPARgamma are reviewed to provide insights into potential mechanisms underlying therapeutic effects of PPARgamma ligands in PH.
Collapse
Affiliation(s)
- Roy L Sutliff
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta VA Medical Center, Decatur, GA 30033, USA.
| | | | | |
Collapse
|
29
|
Yamanouchi T. Concomitant therapy with pioglitazone and insulin for the treatment of type 2 diabetes. Vasc Health Risk Manag 2010; 6:189-97. [PMID: 20407626 PMCID: PMC2856574 DOI: 10.2147/vhrm.s5838] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Indexed: 01/21/2023] Open
Abstract
To prevent hyperinsulinemia, which may cause atherosclerosis, thiazolidinediones (TZDs), also known as insulin sensitizers, are often added to the therapeutic regimen of patients with type 2 diabetes who are receiving insulin. The combination of insulin with pioglitazone, a TZD, reduces glycoated hemoglobin (HbA(1c)) by 0.6%-2.1%. The higher the HbA(1c) baseline the larger the therapeutic reduction of HbA(1c). This combination therapy has been shown to be beneficial even in lean Japanese patients with diabetes. It should be noted that such combination therapy is much more useful when the main clinical aim is lowering not postprandial, but fasting and nocturnal glycemia. The glycemic-lowering effects of pioglitazone alone occur slowly, whereas the addition of insulin to pioglitazone often shows a dramatic glucose-lowering effect. Thus, such combination therapy increases the possibility of frequent hypoglycemia within 1 to 2 months of combining the drugs. Severe hypoglycemia in patients using this therapy is rare. Patients treated with combination therapy who show a predominant reduction of glycemia often have severe edema; in 10%-20% of patients, combination therapy leads to drug-related congestive heart failure (CHF). However, this phenomenon is usually weakened if low doses of pioglitazone which are added to insulin therapy (ie, 15 mg/day or even 7.5 mg/day for women). It is well known that pioglitazone has an anti-atherosclerotic effect, although it is unclear if hyperinsulinemia induces atherogenic changes, either directly or indirectly, by the promotion of obesity. Until now, we have not confirmed whether the anti-atherosclerotic effects of pioglitazone exceed the supposed disadvantageous action of insulin when used in combination therapy. The addition of pioglitazone tends to reduce daily insulin dosages, but study findings have not been consistent. Improvement of lipid profiles has also been weak with this combination therapy. Long-term trials are needed before any conclusions can be reached concerning atherogenic effects of treatment for type 2 diabetes. Combination therapy of even small doses of pioglitazone with insulin should be primarily used for patients who achieve insufficient reduction in glycemia with insulin monotherapy.
Collapse
Affiliation(s)
- Toshikazu Yamanouchi
- Kita-Tokyo-Jueien/Department of Internal Medicine, Teikyo University, Tokyo, Japan.
| |
Collapse
|
30
|
Xu M, Li Y, Suo H, Yan Y, Liu L, Wang Q, Ge Y, Xu Y. Fabricating a pearl/PLGA composite scaffold by the low-temperature deposition manufacturing technique for bone tissue engineering. Biofabrication 2010; 2:025002. [PMID: 20811130 DOI: 10.1088/1758-5082/2/2/025002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Here we developed a composite scaffold of pearl/poly(lactic-co-glycolic acid) (pearl/PLGA) utilizing the low-temperature deposition manufacturing (LDM). LDM makes it possible to fabricate scaffolds with designed microstructure and macrostructure, while keeping the bioactivity of biomaterials by working at a low temperature. Process optimization was carried out to fabricate a mixture of pearl powder, PLGA and 1,4-dioxane with the designed hierarchical structures, and freeze-dried at a temperature of -40 degrees C. Scaffolds with square and designated bone shape were fabricated by following the 3D model. Marrow stem cells (MSCs) were seeded on the pearl/PLGA scaffold and then cultured in a rotating cell culture system. The adhesion, proliferation and differentiation of MSCs into osteoblasts were determined using scanning electronic microscopy, WST-1 assay, alkaline phosphatase activity assay, immunofluorescence staining and real-time reverse transcription polymerase chain reaction. The results showed that the composite scaffold had high porosity (81.98 +/- 3.75%), proper pore size (micropores: <10 microm; macropore: 495 +/- 54 microm) and mechanical property (compressive strength: 0.81 +/- 0.04 MPa; elastic modulus: 23.14 +/- 0.75 MPa). The pearl/PLGA scaffolds exhibited better biocompatibility and osteoconductivity compared with the tricalcium phosphate/PLGA scaffold. All these results indicate that the pearl/PLGA scaffolds fulfill the basic requirements of bone tissue engineering scaffold.
Collapse
Affiliation(s)
- Mingen Xu
- Center Laboratory of Biomanufacture and Tissue Engineering, Hang Zhou Dianzi University, Hangzhou 310018, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Dorafshar AH, Moodley K, Khoe M, Lyon C, Bryer-Ash M. Pioglitazone improves superoxide dismutase mediated vascular reactivity in the obese Zucker rat. Diab Vasc Dis Res 2010; 7:20-7. [PMID: 20368229 DOI: 10.1177/1479164109341688] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that the thiazolidinedione agent, pioglitazone, mediates its chronic BP lowering action via improving vascular reactivity. METHODS AND RESULTS Lean (Fa/fa) and obese (fa/fa) Zucker rats were treated with or without pioglitazone (20 mg/ kg/day) for 4 weeks (n=8 animals per group). Pioglitazone treatment was associated with a significant improvement in oral glucose tolerance in the obese animals (p<0.05 compared with untreated obese). Pioglitazone prevented the development of hypertension seen in obese untreated rats (SBP 126+/-1 versus 138+/-1 mmHg; p<0.0001). Aortic ring preparations from pioglitazone-treated obese rats showed improved relaxation responsiveness (ED(50) 0.28 versus 1.15 U/ ml, p<0.001) to SOD, a NO potentiator, compared with untreated obese animals. CONCLUSIONS SOD-mediated vasorelaxation may contribute to the chronic antihypertensive effect and/or the improvement in insulin sensitivity following pioglitazone treatment.
Collapse
Affiliation(s)
- Amir H Dorafshar
- Department of Medicine, University of California Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
32
|
Garcia-Bates TM, Baglole CJ, Bernard MP, Murant TI, Simpson-Haidaris PJ, Phipps RP. Peroxisome proliferator-activated receptor gamma ligands enhance human B cell antibody production and differentiation. THE JOURNAL OF IMMUNOLOGY 2009; 183:6903-12. [PMID: 19915048 DOI: 10.4049/jimmunol.0900324] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Protective humoral immune responses critically depend on the optimal differentiation of B cells into Ab-secreting cells. Because of the important role of Abs in fighting infections and in successful vaccination, it is imperative to identify mediators that control B cell differentiation. Activation of B cells through TLR9 by CpG-DNA induces plasma cell differentiation and Ab production. Herein, we examined the role of the peroxisome proliferator-activated receptor (PPAR)gamma/RXRalpha pathway on human B cell differentiation. We demonstrated that activated B cells up-regulate their expression of PPARgamma. We also show that nanomolar levels of natural (15-deoxy-Delta(12,14)-prostaglandin J(2)) or synthetic (rosiglitazone) PPARgamma ligands enhanced B cell proliferation and significantly stimulated plasma cell differentiation and Ab production. Moreover, the addition of GW9662, a specific PPARgamma antagonist, abolished these effects. Retinoid X receptor (RXR) is the binding partner for PPARgamma and is required to produce an active transcriptional complex. The simultaneous addition of nanomolar concentrations of the RXRalpha ligand (9-cis-retinoic acid) and PPARgamma ligands to CpG-activated B cells resulted in additive effects on B cell proliferation, plasma cell differentiation, and Ab production. Furthermore, PPARgamma ligands alone or combined with 9-cis-retinoic acid enhanced CpG-induced expression of Cox-2 and the plasma cell transcription factor BLIMP-1. Induction of these important regulators of B cell differentiation provides a possible mechanism for the B cell-enhancing effects of PPARgamma ligands. These new findings indicate that low doses of PPARgamma/RXRalpha ligands could be used as a new type of adjuvant to stimulate Ab production.
Collapse
Affiliation(s)
- Tatiana M Garcia-Bates
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone-receptor superfamily. Originally cloned in 1990, PPARs were found to be mediators of pharmacologic agents that induce hepatocyte peroxisome proliferation. PPARs also are expressed in cells of the cardiovascular system. PPAR gamma appears to be highly expressed during atherosclerotic lesion formation, suggesting that increased PPAR gamma expression may be a vascular compensatory response. Also, ligand-activated PPAR gamma decreases the inflammatory response in cardiovascular cells, particularly in endothelial cells. PPAR alpha, similar to PPAR gamma, also has pleiotropic effects in the cardiovascular system, including antiinflammatory and antiatherosclerotic properties. PPAR alpha activation inhibits vascular smooth muscle proinflammatory responses, attenuating the development of atherosclerosis. However, PPAR delta overexpression may lead to elevated macrophage inflammation and atherosclerosis. Conversely, PPAR delta ligands are shown to attenuate the pathogenesis of atherosclerosis by improving endothelial cell proliferation and survival while decreasing endothelial cell inflammation and vascular smooth muscle cell proliferation. Furthermore, the administration of PPAR ligands in the form of TZDs and fibrates has been disappointing in terms of markedly reducing cardiovascular events in the clinical setting. Therefore, a better understanding of PPAR-dependent and -independent signaling will provide the foundation for future research on the role of PPARs in human cardiovascular biology.
Collapse
Affiliation(s)
- Milton Hamblin
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
34
|
Wang N, Symons JD, Zhang H, Jia Z, Gonzalez FJ, Yang T. Distinct functions of vascular endothelial and smooth muscle PPARgamma in regulation of blood pressure and vascular tone. Toxicol Pathol 2009; 37:21-7. [PMID: 19075043 PMCID: PMC2791342 DOI: 10.1177/0192623308328545] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Thiazolidinediones (TZDs) are peroxisome proliferators-activated receptor gamma (PPARgamma) activators that exhibit antihypertensive and vasculo-protective effects. Here we describe the use of Tie2Cre/flox and SM22Cre/flox mice, which respectively lacked PPARgamma in the endothelium and the smooth muscle, to study vascular function of PPARgamma. Rosiglitazone (RGZ) induced a similar blood pressure (BP)-lowering effect in deoxycorticosterone acetate (DOCA) salt-treated PPARgamma(f/f) and SM22Cre/flox mice, whereas Tie2Cre/flox mice were completely resistant to this effect. The femoral arteries lacking endothelial PPARgamma exhibited increased reactivity to various vasoconstrictors without a significant alteration in acetylcholine-induced relaxation. In sharp contrast, the vasculature lacking smooth muscle PPARgamma had blunted sensitivity to alpha1-adrenergic agents but enhanced sensitivity to acetylcholine. Our results demonstrated endothelium but not smooth muscle as the site for TZD-induced BP-lowering effect and also uncovered distinct functions of endothelial and smooth muscle PPARgamma in regulation of vascular tone.
Collapse
MESH Headings
- Animals
- Blood Pressure/drug effects
- Blood Pressure/physiology
- Desoxycorticosterone/pharmacology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Femoral Artery/drug effects
- Femoral Artery/physiopathology
- Hypertension/chemically induced
- Hypertension/physiopathology
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- PPAR gamma/deficiency
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Rosiglitazone
- Thiazolidinediones/pharmacology
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Ningning Wang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City
| | - J. David Symons
- College of Health, University of Utah, Salt Lake City
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City
| | - Hui Zhang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City
| | - Zhanjun Jia
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City
| | - Frank J. Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
35
|
Zhou M, Xu H, Pan L, Wen J, Liao W, Chen K. Rosiglitazone promotes atherosclerotic plaque stability in fat-fed ApoE-knockout mice. Eur J Pharmacol 2008; 590:297-302. [DOI: 10.1016/j.ejphar.2008.05.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2007] [Revised: 05/09/2008] [Accepted: 05/22/2008] [Indexed: 11/25/2022]
|
36
|
Beyer AM, Baumbach GL, Halabi CM, Modrick ML, Lynch CM, Gerhold TD, Ghoneim SM, de Lange WJ, Keen HL, Tsai YS, Maeda N, Sigmund CD, Faraci FM. Interference with PPARgamma signaling causes cerebral vascular dysfunction, hypertrophy, and remodeling. Hypertension 2008; 51:867-71. [PMID: 18285614 PMCID: PMC2408877 DOI: 10.1161/hypertensionaha.107.103648] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 01/23/2008] [Indexed: 11/16/2022]
Abstract
The transcription factor PPARgamma is expressed in endothelium and vascular muscle where it may exert antiinflammatory and antioxidant effects. We tested the hypothesis that PPARgamma plays a protective role in the vasculature by examining vascular structure and function in heterozygous knockin mice expressing the P465L dominant negative mutation in PPARgamma (L/+). In L/+ aorta, responses to the endothelium-dependent agonist acetylcholine (ACh) were not affected, but there was an increase in contraction to serotonin, PGF(2alpha), and endothelin-1. In cerebral blood vessels both in vitro and in vivo, ACh produced dilation that was markedly impaired in L/+ mice. Superoxide levels were elevated in cerebral arterioles from L/+ mice and responses to ACh were restored to normal with a scavenger of superoxide. Diameter of maximally dilated cerebral arterioles was less, whereas wall thickness and cross-sectional area was greater in L/+ mice, indicating cerebral arterioles underwent hypertrophy and remodeling. Thus, interference with PPARgamma signaling produces endothelial dysfunction via a mechanism involving oxidative stress and causes vascular hypertrophy and inward remodeling. These findings indicate that PPARgamma has vascular effects which are particularly profound in the cerebral circulation and provide genetic evidence that PPARgamma plays a critical role in protecting blood vessels.
Collapse
Affiliation(s)
- Andreas M Beyer
- Genetics Graduate Program, Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hausman GJ, Poulos SP, Pringle TD, Azain MJ. The influence of thiazolidinediones on adipogenesis in vitro and in vivo: Potential modifiers of intramuscular adipose tissue deposition in meat animals1,2. J Anim Sci 2008; 86:E236-43. [PMID: 17686902 DOI: 10.2527/jas.2007-0219] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Thiazolidinediones (TZD) are insulin sensitizing agents currently used for the treatment of type 2 diabetes and are widely used as adipogenic agents because they are ligands of peroxisome proliferator-activated receptor gamma (PPARgamma), a key adipogenic transcription factor. In vivo and in vitro studies of TZD as potential modifiers of intramuscular or marbling adipogenesis are reviewed. Thiazolidinedione-induced adipogenesis has been reported in numerous cell culture systems, including rodent, human, bovine, and porcine adipose tissue stromal-vascular (S-V) cell cultures. Studies of porcine S-V cell cultures derived from semitendinosus muscle show that TZD can potentially modify intramuscular or marbling adipogenesis. Preadipocyte recruitment was TZD-dependent in muscle S-V cultures but TZD-independent in adipose S-V cultures. There appear to be differences between adipocytes in muscle and subcutaneous adipose tissue, reminiscent of differences observed in adipocytes from different adipose tissue depots. Troglitazone, a TZD, induces marbling adipogenesis without inhibiting myogenesis when cells are grown on laminin precoated culture dishes. Additionally, troglitazone treatment does not increase lipid content in porcine adipose tissue or muscle S-V cell cultures. Thiazolidinedione treatment increases lipid content of muscle in rodents and humans; however, rosiglitazone treatment for 49 d in pigs did not influence muscle lipid content and meat quality, but several significant changes in muscle fatty acid composition were observed. Although timing of treatment with TZD needs to be optimized, evidence suggests these compounds may enhance marbling deposition in swine.
Collapse
|
38
|
Rohatgi A, McGuire DK. Effects of the thiazolidinedione medications on micro- and macrovascular complications in patients with diabetes--update 2008. Cardiovasc Drugs Ther 2008; 22:233-40. [PMID: 18373186 DOI: 10.1007/s10557-008-6093-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Accepted: 01/24/2008] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The thiazolidinedione (TZD) drugs, including pioglitazone (Actos) and rosiglitazone (Avandia), are commonly prescribed in patients with type 2 diabetes mellitus (T2DM), largely due to their favorable effects on hyperglycemia, insulin sensitivity, and cardiometabolic profile. However, the data are sparse assessing the effects of TZDs on micro- and macrovascular disease risk. DISCUSSION Although no studies have been published on microvascular clinical outcomes, both TZDs significantly reduce the urine albumin-to-creatinine ratio. TZDs have consistently been associated with favorable effects on atherosclerosis and cardiovascular disease (CVD) risk. Only one study has been published to date specifically designed to assess the effects of a TZD (pioglitazone) on macrovascular outcomes, the PROactive trial. In this trial, pioglitazone versus placebo was associated with a non-significant 10% reduction in the combined primary endpoint of mortality, coronary and peripheral vascular events, and revascularizations. No individual trial has been published specifically assessing the CVD effects of rosiglitazone, but several meta-analyses and a published interim report from an ongoing trial (RECORD) point to safety concerns regarding rosiglitazone use and the risk of myocardial infarctions (MI), leading to amplified warnings in the product labeling for rosiglitazone to reflect these concerns. CONCLUSION All published trials and meta-analyses of TZDs have consistently shown increased risk of heart failure (HF) with both TZDs, though the actual placebo-subtracted incidence of HF is low (<0.5% per year). The initiation of either TZD is contraindicated in patients with NHYA class III or IV HF, and cautions exist for their use in any patient with heart failure. Much uncertainty remains regarding the aggregate CVD effects of the TZDs, and several trials are presently underway to further address these issues.
Collapse
Affiliation(s)
- Anand Rohatgi
- Cardiovascular Division, University of Texas Southwestern Medical Center, Dallas, TX 75235-9047, USA
| | | |
Collapse
|
39
|
Affiliation(s)
- Darren K McGuire
- Cardiovascular Division and Donald W. Reynolds Cardiovascular Clinical Research Center, University of Texas Southwestern Medical Center, 5909 Harry Hines Blvd, Dallas, TX 75235-9047, USA.
| | | |
Collapse
|
40
|
Westerweel PE, Verhaar MC. Protective Actions of PPAR-gamma Activation in Renal Endothelium. PPAR Res 2008; 2008:635680. [PMID: 19266048 PMCID: PMC2650079 DOI: 10.1155/2008/635680] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 12/08/2008] [Indexed: 01/30/2023] Open
Abstract
Renal endothelial damage is pivotal in the initiation and progression of renal disease. Damaged renal endothelium may be regenerated through proliferation of local endothelium and circulation-derived endothelial progenitor cells. Activation of the PPAR-gamma-receptors present on endothelial cells affects their cellular behavior. Proliferation, apoptosis, migration, and angiogenesis by endothelial cells are modulated, but may involve both stimulation and inhibition depending on the specific circumstances. PPAR-gamma-receptor activation stimulates the production of nitric oxide, C-type natriuretic peptide, and superoxide dismutase, while endothelin-1 production is inhibited. Together, they augment endothelial function, resulting in blood pressure lowering and direct renoprotective effects. The presentation of adhesion molecules and release of cytokines recruiting inflammatory cells are inhibited by PPAR-gamma-agonism. Finally, PPAR-gamma-receptors are also found on endothelial progenitor cells and PPAR-gamma-agonists stimulate progenitor-mediated endothelial repair. Together, the stimulatory effects of PPAR-gamma-agonism on endothelium make an important contribution to the beneficial actions of PPAR-gamma-agonists on renal disease.
Collapse
Affiliation(s)
- Peter E. Westerweel
- 1Department of Vascular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- 2Department of Internal Medicine, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Marianne C. Verhaar
- 1Department of Vascular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- *Marianne C. Verhaar:
| |
Collapse
|
41
|
Fang CC, Ng Jao YTF, Yi-Chen, Yu CL, Chen CL, Wang SP. Angiographic and clinical outcomes of rosiglitazone in patients with type 2 diabetes mellitus after percutaneous coronary interventions: a single center experience. Angiology 2007; 58:523-34. [PMID: 18024934 DOI: 10.1177/0003319707303587] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A beneficial effect of thiazolidinediones includes the reduction of intermediate markers, suggesting a potential for reducing atherosclerosis and restenosis. The objective of this study was to determine if rosiglitazone (RSG) reduced the odds of restenosis and if RSG improved the odds of clinical outcomes after percutaneous coronary intervention (PCI) in type 2 diabetes mellitus (DM) patients. A total of 609 patients with 734 lesions were selected from the period between January 1, 2001 and January 31, 2004. These patients were divided into 2 groups: a "control" group representing patients seen between January 1, 2001 and September 2002 when RSG was not available in our hospital and a "RSG treatment" group representing patients seen between September 2002 and January 31, 2004 when RSG was available in our hospital. Thus, 213 patients with 253 lesions (1.19 L/P) were placed in the RSG group and 396 patients with 481 lesions (1.21 L/P) were placed in the control group. Subgroup analysis based on the PCI received had 88 patients in the RSG arm receiving balloon angioplasty and 125 patients receiving coronary stenting; the control group had 187 and 209 patients, respectively, in the subgroups. Primary endpoint was angiographic restenosis at 6 months, and secondary endpoints were death, myocardial infarction, and target lesion revascularization. More patients in the control group were insulin-requiring, had poorer left ventricular function, but had a larger preprocedural minimal lumen diameter (pre-MLD). At 6 months, restenosis and reocclusion rates were lower in the RSG group (P = .014 and P = .006, respectively). Twenty-nine patients died in the control group versus 1 in the RSG group (P <or= .001). RSG (P = .019), stenting (P = .005), preprocedural reference vessel diameter (P = .017), metformin (P = .022), pre-MLD (P < .001), hyperlipidemia (P = .016), and combined RSG and metformin (P = .020) were predictors of restenosis, while RSG (P = .016) and metformin (P = .029) were predictors of survival. In conclusion, RSG was found safe and well tolerated and was associated with reduced odds of restenosis, reocclusion, and mortality rates in type 2 DM patients independent of glycemic control and PCI performed.
Collapse
|
42
|
Anan F, Masaki T, Fukunaga N, Teshima Y, Iwao T, Kaneda K, Umeno Y, Okada K, Wakasugi K, Yonemochi H, Eshima N, Saikawa T, Yoshimatsu H. Pioglitazone shift circadian rhythm of blood pressure from non-dipper to dipper type in type 2 diabetes mellitus. Eur J Clin Invest 2007; 37:709-14. [PMID: 17696960 DOI: 10.1111/j.1365-2362.2007.01854.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Insulin resistance significantly correlated with a non-dipper type of essential hypertension. Thiazolidinediones (TZD), oral hypoglycaemic agents that act as insulin sensitizers, have been demonstrated in multiple in vivo and in vitro studies to possess antihypertensive properties. This study examined the efficacy of TZD therapy with pioglitazone at transforming the circadian rhythms of blood pressure from a non-dipper to a dipper type. MATERIALS We examined 31 patients with type 2 diabetes mellitus during both a baseline period and a period of treatment with pioglitazone. Patients received 15 mg day(-1) pioglitazone for four weeks and 30 mg day(-1) for 12 weeks. Twenty-four hour ambulatory blood pressure monitoring (ABPM) and laboratory data (blood tests for cardiovascular risk factors) were obtained at the beginning and end of the study. RESULTS In non-dippers (n = 16), but not dippers (n = 15), we observed a significant interaction between pioglitazone therapy and nocturnal falls in systolic and diastolic blood pressure. This examination indicated that the magnitude of the nocturnal blood pressure fall was affected by pioglitazone therapy. In non-dippers, but not dippers, a significant correlation was observed between the percent decrease in nocturnal BP and the homeostasis model assessment (HOMA) index (r = 0.774, P = 0.0007). CONCLUSIONS The present study demonstrated that pioglitazone can restore the nocturnal BP declines in parallel to reductions in the HOMA index, suggesting that insulin resistance may play an important role in the genesis of circadian BP rhythms. TZD-based treatment may thus have the additional therapeutic advantage of reducing the risk of cardiovascular complications by transforming the circadian rhythm of BP.
Collapse
Affiliation(s)
- F Anan
- Faculty of Medicine, Oita University, Oita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Aguilar D, Bozkurt B, Pritchett A, Petersen NJ, Deswal A. The Impact of Thiazolidinedione Use on Outcomes in Ambulatory Patients With Diabetes Mellitus and Heart Failure. J Am Coll Cardiol 2007; 50:32-6. [PMID: 17601542 DOI: 10.1016/j.jacc.2007.01.096] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 01/11/2007] [Accepted: 01/16/2007] [Indexed: 11/16/2022]
Abstract
OBJECTIVES This study sought to examine the relationship between thiazolidinedione (TZD) use and outcomes in ambulatory patients with diabetes and heart failure (HF). BACKGROUND Thiazolidinediones have been relatively contraindicated in diabetic patients with HF. METHODS We conducted a retrospective study of a national cohort of veterans with HF and diabetes treated in ambulatory clinics at Veterans Affairs medical centers. Patients were classified into those using TZDs and those not using insulin-sensitizing medication based on prescriptions filled 90 days before or 30 days after the index outpatient visit. The outcomes were time to hospitalization for HF and time to death. RESULTS Of 7,147 ambulatory HF patients receiving diabetic therapy, 818 (11.4%) were receiving a TZD and 4,700 (65.8%) were not receiving insulin sensitizers. Over 2 years of follow-up, 134 (16.4%) patients receiving TZDs and 741 (15.8%) patients not receiving insulin-sensitizing medications required HF hospitalization (adjusted hazard ratio 1.00, 95% confidence interval 0.81 to 1.24, p = 0.97). A total of 168 (20.5%) patients receiving TZDs and 1,192 (25.4%) patients not receiving insulin-sensitizing medications died (adjusted hazard ratio 0.98, 95% confidence interval 0.81 to 1.17, p = 0.80). CONCLUSIONS In ambulatory patients with established HF and diabetes, the use of TZDs was not associated with an increased risk of HF hospitalization or total mortality when compared with those not receiving insulin-sensitizing medications.
Collapse
Affiliation(s)
- David Aguilar
- Winters Center for Heart Failure Research and Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
44
|
Crossno JT, Garat CV, Reusch JEB, Morris KG, Dempsey EC, McMurtry IF, Stenmark KR, Klemm DJ. Rosiglitazone attenuates hypoxia-induced pulmonary arterial remodeling. Am J Physiol Lung Cell Mol Physiol 2007; 292:L885-97. [PMID: 17189321 DOI: 10.1152/ajplung.00258.2006] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Thiazolidinediones (TZDs) are insulin-sensitizing agents that also decrease systemic blood pressure, attenuate the formation of atherosclerotic lesions, and block remodeling of injured arterial walls. Recently, TZDs were shown to prevent pulmonary arterial (PA) remodeling in rats treated with monocrotaline. Presently we report studies testing the ability of the TZD rosiglitazone (ROSI) to attenuate pathological arterial remodeling in the lung and prevent the development of pulmonary hypertension (PH) in rats subjected to chronic hypoxia. PA remodeling was reduced in ROSI-treated animals exposed to hypoxia compared with animals exposed to hypoxia alone. ROSI treatment blocked muscularization of distal pulmonary arterioles and reversed remodeling and neomuscularization in lungs of animals previously exposed to chronic hypoxia. Decreased PA remodeling in ROSI-treated animals was associated with decreased smooth muscle cell proliferation, decreased collagen and elastin deposition, and increased matrix metalloproteinase-2 activity in the PA wall. Cells expressing the c-Kit cell surface marker were observed in the PA adventitia of untreated animals exposed to hypoxia but not in ROSI-treated hypoxic rats. Right ventricular hypertrophy and cardiomyocyte hypertrophy were also blunted in ROSI-treated hypoxic animals. Interestingly, mean PA pressures were elevated equally in the untreated and ROSI-treated groups, indicating that ROSI had no effect on the development of PH. However, mean PA pressure was normalized acutely in both groups of hypoxia-exposed animals by Fasudil, an agent that inhibits RhoA/Rho kinase-mediated vasoconstriction. We conclude that ROSI can attenuate and reverse PA remodeling and neomuscularization associated with hypoxic PH. However, this agent fails to block the development of PH, apparently because of its inability to repress sustained Rho kinase-mediated arterial vasoconstriction.
Collapse
Affiliation(s)
- Joseph T Crossno
- Pulmonary and Critical Care Services, Veterans Affairs Medical Center, Denver, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Desouza CV, Gerety M, Hamel FG. Long-term effects of a PPAR-gamma agonist, pioglitazone, on neointimal hyperplasia and endothelial regrowth in insulin resistant rats. Vascul Pharmacol 2007; 46:188-94. [PMID: 17141574 DOI: 10.1016/j.vph.2006.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Accepted: 10/02/2006] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Insulin resistance is an independent risk factor for cardiovascular disease. PPAR-gamma agonists like pioglitazone decrease insulin resistance and have been shown to reduce neointimal hyperplasia in the short-term. However long-term studies on endothelial regrowth and neointimal hyperplasia have not been done. METHODS AND RESULTS We used hyperinsulinemic, normoglycemic Zucker fatty rats. Rats were treated with either 10 mg/kg body wt. pioglitazone or placebo till the end of the experiment. Rats underwent carotid angioplasty at age 12-14 weeks, 1 week after treatment was begun. In one set of experiments rats were sacrificed at 6 months and neointimal hyperplasia and VEGF expression was assessed. In another set of experiments rats were sacrificed at 3 and 6 months. Endothelial regrowth was determined. The rats were all normoglycemic and hyperinsulinemic. Pioglitazone treated rats had a significantly lesser degree of neointimal hyperplasia than control rats. Treated rats also had decreased VEGF expression. Endothelial regrowth was decreased in the treated rats at 6 months. CONCLUSION We conclude that although pioglitazone decreases neointimal hyperplasia even at 6 months, it retards endothelial regrowth, which could predispose the denuded vessel to thrombotic events. This may be modulated by a suppression of VEGF expression.
Collapse
Affiliation(s)
- Cyrus V Desouza
- Omaha Veterans Affairs Medical Center, Omaha, NE 68105, USA.
| | | | | |
Collapse
|
46
|
Sarafidis PA, Bakris GL. Review: Insulin and endothelin: an interplay contributing to hypertension development? J Clin Endocrinol Metab 2007; 92:379-85. [PMID: 17118997 DOI: 10.1210/jc.2006-1819] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
CONTEXT The aim of this article was to review the existing data on the interactions among insulin, insulin resistance, and endothelin and how those contribute to the development of hypertension in insulin-resistant states. EVIDENCE ACQUISITION A literature search of MEDLINE database was performed to identify English-language articles published during the last 20 yr. Search terms used were endothelin, insulin, insulin resistance, and hyperinsulinemia in combination with blood pressure and hypertension. Reference lists of retrieved articles were also evaluated for relevant information. EVIDENCE SYNTHESIS Several mechanisms connect insulin resistance and compensatory hyperinsulinemia with blood pressure elevation in the context of the metabolic syndrome, i.e. sodium retention, sympathetic activation, and impairment of endothelial nitric oxide production. Accumulating evidence suggests that activation of the endothelin system seems to be another important, yet less discussed, mechanism. In vitro studies have shown that insulin stimulates both endothelin-1 production and action on the vascular wall. In vivo, high levels of insulin result in increase in circulating endothelin-1 in healthy individuals, and this effect is also seen in insulin-resistant subjects, a relationship not observed with nitric oxide production. Moreover, endothelin receptor antagonism effectively reduces blood pressure in animal models of insulin resistance and hypertension. On the other hand, elevation of endothelin-1 levels can further increase insulin resistance, forming possibly a deleterious circle. CONCLUSIONS Endothelin-1 may play a crucial role in the pathogenesis of hypertension in insulin-resistant states. Future research should examine the potential of endothelin receptor antagonism to help blood pressure control in patients with insulin resistance.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- Hypertension/Clinical Research Center, Department of Preventive Medicine, Rush University Medical Center, 1700 West Van Buren, Suite 470, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
47
|
Galli A, Mello T, Ceni E, Surrenti E, Surrenti C. The potential of antidiabetic thiazolidinediones for anticancer therapy. Expert Opin Investig Drugs 2007; 15:1039-49. [PMID: 16916271 DOI: 10.1517/13543784.15.9.1039] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The thiazolidinediones (TZDs) are a class of synthetic compounds for treatment of insulin-resistant Type 2 diabetes mellitus. TZDs are known activators of the peroxisome proliferator-activated receptor-gamma (PPAR-gamma), and exert their antidiabetic action largely through this nuclear receptor family. Moreover, increasing experimental evidences of PPAR-gamma-independent effects are accumulating. Apart from the established metabolic actions, TZD treatment exerts additional biological effect such as control of cell growth, differentiation, motility and programmed cell death. In this context, considerable interest has focused on TZDs as potential chemopreventive agents in oncology; however, despite encouraging observation on the potential anticancer effect of these drugs in several in vitro experimental models, controversial results have been obtained with animal models and in pilot clinical trials. This review summarises the molecular mechanisms of the antineoplastic actions of TZDs and the relevance of these findings in human pathology and therapy.
Collapse
Affiliation(s)
- Andrea Galli
- University of Florence, Gastroenterology Unit, Department of Clinical Pathophysiology, Viale Morgani 85,50134 - Firenze, Florence, Italy.
| | | | | | | | | |
Collapse
|
48
|
Sander TL, Noll L, Klinkner DB, Weihrauch D, He BJ, Kaul S, Zangwill SD, Tweddell JS, Pritchard KA, Oldham KT. Rosiglitazone antagonizes vascular endothelial growth factor signaling and nuclear factor of activated T cells activation in cardiac valve endothelium. ACTA ACUST UNITED AC 2006; 13:181-90. [PMID: 16840174 DOI: 10.1080/10623320600760308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear factor of activated T cells, Cytoplasmic 1 (NFATc1) is required for heart valve formation. Vascular endothelial growth factor (VEGF) signaling, mediated by NFATc1 activation, positively regulates growth of valvular endothelial cells. However, regulators of VEGF/NFATc1 signaling in valve endothelium are poorly understood. Peroxisome proliferator-activated receptor gamma (PPARgamma) inhibits NFATc1 activity in T cells and cardiomyocytes, but it is not known if PPARgamma controls NFATc1 function in endothelial cells. The authors hypothesize PPARgamma antagonizes VEGF signaling in valve endothelium by inhibiting NFATc1. Endothelial cells isolated from human valve leaflet tissue were shown by immunocytochemistry to express the endothelial-specific markers von Willebrand factor (vWF) and platelet endothelial cell adhesion molecule (PECAM)-1. VEGF-induced proliferation and migration of human pulmonary valve endothelial cells (HPVECs) were inhibited by rosiglitazone (ROSI), a specific ligand of PPARgamma activation, suggesting that PPARgamma disrupts VEGF signaling in the valve endothelium. ROSI also antagonized VEGF-mediated NFATc1 nuclear translocation in HPVECs, suggesting that PPARgamma inhibits VEGF signaling of NFATc1 activation in the valve. The effect of ROSI on nonvalve human umbilical vein endothelial cells (HUVECs) was tested in parallel and a similar inhibition of NFATc1 activation was observed. These data provide the first demonstration that ROSI negatively regulates VEGF signaling in the valve endothelium by a mechanism involving NFATc1 activation and nuclear translocation.
Collapse
Affiliation(s)
- Tara L Sander
- Department of Surgery, Medical College of Wisconsin and Children's Research Institute, Milwaukee, Wisconsin 53226, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Patel CB, De Lemos JA, Wyne KL, McGuire DK. Thiazolidinediones and risk for atherosclerosis: pleiotropic effects of PPar gamma agonism. Diab Vasc Dis Res 2006; 3:65-71. [PMID: 17058625 DOI: 10.3132/dvdr.2006.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Despite advances in the development of anti-hyperglycaemic drugs and a greater focus on cardiovascular risk modification for patients with diabetes, cardiovascular disease remains the most common complication of type 2 diabetes. Since their initial availability in 1997, the thiazolidinediones have become one of the most commonly prescribed classes of medications for type 2 diabetes. In addition to glucose control, the thiazolidinediones have a number of pleiotropic effects on myriad traditional and non-traditional risk factors for cardiovascular disease, and hold promise with regard to modification of cardiovascular risk. In a recently reported large-scale clinical trial, pioglitazone was associated with improved cardiovascular outcomes in patients with type 2 diabetes and prevalent atherosclerotic disease. In this review, we summarise the experimental, preclinical and clinical data regarding the effects of the thiazolidinediones on cardiovascular risk factors and clinical outcomes.
Collapse
Affiliation(s)
- Chetan B Patel
- Department of Internal Medicine, Duke University Medical Center, Durham, NC, USA
| | | | | | | |
Collapse
|
50
|
Magee MF, Isley WL. Rationale, design, and methods for glycemic control in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) Trial. Am J Cardiol 2006; 97:20G-30G. [PMID: 16813735 DOI: 10.1016/j.amjcard.2006.02.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A major therapeutic question in considering accelerated atherogenesis in patients with type 2 diabetes mellitus is whether reducing insulin resistance, as a proximal defect of a host of proatherogenic abnormalities including hyperglycemia, will be superior for decreasing mortality and coronary artery disease (CAD) risk compared with treating hyperglycemia to overcome insulin resistance with insulin-providing agents. This question is highly relevant, since earlier targeted glycemic control trials utilizing conventional glucose-lowering strategies that increase insulin levels have generally failed to reduce CAD risk despite markedly reducing microvascular risk. The Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial seeks to determine whether primarily using an insulin-sensitizing strategy for treatment of type 2 diabetes is superior when compared with primarily using an insulin-providing strategy with regard to cardiovascular outcomes. This article presents the rationale, design, and methods being used to test the glycemic control hypothesis in BARI 2D.
Collapse
Affiliation(s)
- Michelle F Magee
- MedStar Diabetes Institute, MedStar Research Institute at Washington Hospital Center and Georgetown University Hospital, Washington, DC 20010, USA.
| | | |
Collapse
|