1
|
Katana Z, Sianidou K, Kaiopoulos G, Deligianni F, Tsetsakos S, Kouvatsi A, Sakellari I, Kritis A, Touraki M, Sotiropoulos D, Xagorari A. Molecular and biochemical evaluation of oxidative effects of cord blood CD34+ MPs on hematopoietic cells. Blood Cells Mol Dis 2024; 108:102871. [PMID: 39013336 DOI: 10.1016/j.bcmd.2024.102871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
A graft source for allogeneic hematopoietic stem cell transplantation is umbilical cord blood, which contains umbilical cord blood mononuclear cells (MNCs and mesenchymal stem cells, both an excellent source of extracellular microparticles (MPs). MPs act as cell communication mediators, which are implicated in reactive oxygen species formation or detoxification depending on their origin. Oxidative stress plays a crucial role in both the development of cancer and its treatment by triggering apoptotic mechanisms, in which CD34+ cells are implicated. The aim of this work is to investigate the oxidative stress status and the apoptosis of HL-60 and mononuclear cells isolated from umbilical cord blood (UCB) following a 24- and 48-hour exposure to CD34 + microparticles (CD34 + MPs). The activity of superoxide dismutase, glutathione reductase, and glutathione S-transferase, as well as lipid peroxidation in the cells, were employed as oxidative stress markers. A 24- and 48-hour exposure of leukemic and mononuclear cells to CD34 + -MPs resulted in a statistically significant increase in the antioxidant activity and lipid peroxidation in both cells types. Moreover, CD34 + MPs affect the expression of BCL2 and FAS and related proteins and downregulate the hematopoietic differentiation program in both HL-60 and mononuclear cells. Our results indicate that MPs through activation of antioxidant enzymes in both homozygous and nonhomozygous cells might serve as a means for graft optimization and enhancement.
Collapse
Affiliation(s)
- Zoi Katana
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki Sianidou
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gregory Kaiopoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Deligianni
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sarantis Tsetsakos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Anastasia Kouvatsi
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Aristeidis Kritis
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Touraki
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Damianos Sotiropoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Angeliki Xagorari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece.
| |
Collapse
|
2
|
Cizkova K, Koubova K, Tauber Z. Lipid Messenger Phosphatidylinositol-4,5-Bisphosphate Is Increased by Both PPARα Activators and Inhibitors: Relevance for Intestinal Cell Differentiation. BIOLOGY 2022; 11:biology11070997. [PMID: 36101378 PMCID: PMC9312331 DOI: 10.3390/biology11070997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Fibrates, such as fenofibrate, are widely used drugs for dyslipidaemia treatment. It is known that they activate peroxisome proliferator-activated receptor α (PPARα) which serves as a lipid sensor in the organism. This article addresses how activators and inhibitor of the PPARα could affect differentiation of intestinal cells. Carcinogenesis is a disruption of normal differentiation process and colorectal carcinoma is the third most common cancer in terms of incidence, but the secondp in terms of mortality. One of the important signalling pathways in intestinal cell differentiation as well as carcinogenesis is PI3K/Akt/PTEN. We showed that PPARα activators as well as inhibitor affected the levels of one member of this pathway called phosphatidylinositol-4,5-bisphosphate. This molecule is important for formation of microvilli, the essential structures of fully differentiated intestinal cells. Abstract We investigated the effects of PPARα activators fenofibrate and WY-14643 as well as the PPARα inhibitor GW6471 on the PI3K/Akt/PTEN pathway of intestinal cell differentiation. Our previous study showed that all these compounds increased the expression of villin, a specific marker of intestinal cell differentiation in HT-29 and Caco2 cells. Our current results confirmed the central role of lipid messenger phosphatidylinositol-4,5-bisphosphate (PIP2), a known player in brush border formation, in mediating the effects of tested PPARα ligands. Although all tested compounds increased its levels, surprisingly, each of them affected different PIP2-metabolizing enzymes, especially the levels of PIP5K1C and PTEN. Moreover, we found a positive relationship between the expression of PPARα itself and PIP2 as well as PIP5K1C. By contrast, PPARα was negatively correlated with PTEN. However, the expression of antigens of interest was independent of PPARα subcellular localization, suggesting that it is not directly involved in their regulation. In colorectal carcinoma tissues we found a decrease in PTEN expression, which was accompanied by a change in its subcellular localization. This change was also observed for the regulatory subunit of PI3K. Taken together, our data revealed that fenofibrate, WY-14643, and GW6471 affected different members of the PI3K/Akt/PTEN pathway. However, these effects were PPARα-independent.
Collapse
|
3
|
Cizkova K, Foltynkova T, Hanyk J, Kamencak Z, Tauber Z. When Activator and Inhibitor of PPARα Do the Same: Consequence for Differentiation of Human Intestinal Cells. Biomedicines 2021; 9:biomedicines9091255. [PMID: 34572440 PMCID: PMC8472525 DOI: 10.3390/biomedicines9091255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-dependent transcription factor that plays a role in various processes including differentiation of several cell types. We investigated the role of PPARα in the differentiation of intestinal cells using HT-29 and Caco2 cell lines as a model as well as human normal colon and colorectal carcinoma tissues. We detected a significant increase in PPARα expression in differentiated HT-29 cells as well as in normal surface colon epithelium where differentiated cells are localised. Thus, it seems that PPARα may play a role in differentiation of intestinal cells. Interestingly, we found that both PPARα activators (fenofibrate and WY-14643) as well as its inhibitor (GW6471) regulated proliferation and differentiation of HT-29 cells in vitro in the same way. Both compounds led to a decrease in proliferation accompanied by a significant increase in expression of villin, intestinal alkaline phosphatase (differentiation markers). Moreover, the same trend in villin expression was observed in Caco2 cells. Furthermore, villin expression was independent of subcellular localisation of PPARα. In addition, we found similar levels of PPARα expression in colorectal carcinomas in comparison to adjacent normal epithelium. All these findings support the hypothesis that differentiation of intestinal epithelium is PPARα-independent.
Collapse
Affiliation(s)
| | | | | | | | - Zdenek Tauber
- Correspondence: ; Tel.: +420-585-632-283; Fax: +420-585-632-966
| |
Collapse
|
4
|
Karpman D, Tontanahal A. Extracellular vesicles in renal inflammatory and infectious diseases. Free Radic Biol Med 2021; 171:42-54. [PMID: 33933600 DOI: 10.1016/j.freeradbiomed.2021.04.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles can mediate cell-to-cell communication, or relieve the parent cell of harmful substances, in order to maintain cellular integrity. The content of extracellular vesicles includes miRNAs, mRNAs, growth factors, complement factors, cytokines, chemokines and receptors. These may contribute to inflammatory and infectious diseases by the exposure or transfer of potent effectors that induce vascular inflammation by leukocyte recruitment and thrombosis. Furthermore, vesicles release cytokines and induce their release from cells. Extracellular vesicles possess immune modulatory and anti-microbial properties, and induce receptor signaling in the recipient cell, not least by the transfer of pro-inflammatory receptors. Additionally, the vesicles may carry virulence factors systemically. Extracellular vesicles in blood and urine can contribute to the development of kidney diseases or exhibit protective effects. In this review we will describe the role of EVs in inflammation, thrombosis, immune modulation, angiogenesis, oxidative stress, renal tubular regeneration and infection. Furthermore, we will delineate their contribution to renal ischemia/reperfusion, vasculitis, glomerulonephritis, lupus nephritis, thrombotic microangiopathies, IgA nephropathy, acute kidney injury, urinary tract infections and renal transplantation. Due to their content of miRNAs and growth factors, or when loaded with nephroprotective modulators, extracellular vesicles have the potential to be used as therapeutics for renal regeneration.
Collapse
Affiliation(s)
- Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, 22185, Lund, Sweden.
| | - Ashmita Tontanahal
- Department of Pediatrics, Clinical Sciences Lund, Lund University, 22185, Lund, Sweden
| |
Collapse
|
5
|
Fenofibrate Reverses Dysfunction of EPCs Caused by Chronic Heart Failure. J Cardiovasc Transl Res 2019; 13:158-170. [PMID: 31701352 DOI: 10.1007/s12265-019-09889-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022]
Abstract
The enhanced activity of endothelial progenitor cells (EPCs) by AMP-activated protein kinase (AMPK) agonists might explain the reversal of chronic heart failure (CHF)-mediated endothelial dysfunction. We studied baseline circulating EPC numbers in patients with heart failure and clarified the effect of fenofibrate on both circulating angiogenic cell (CAC) and late EPC activity. The numbers of circulating EPCs in CHF patients were quantified by flow cytometry. Blood-derived mononuclear cells were cultured, and CAC and late EPC functions, including fibronectin adhesion, tube formation, and migration, were evaluated. We focused on the effect of fenofibrate, an AMPK agonist, on EPC function and Akt/eNOS cascade activation in vitro. The number of circulating EPCs (CD34+/KDR+) was significantly lower in CHF patients (ischemic cardiomyopathy (ICMP): 0.07%, dilated cardiomyopathy (DCMP): 0.068%; p < 0.05) than in healthy subjects (0.102% of the gating region). In CACs, fibronectin adhesion function was reversed by fenofibrate treatment (p < 0.05). Similar results were also found for tube formation and migration in late EPCs, which were significantly improved by fenofibrate in an AMPK-dependent manner (p < 0.05), suggesting that fenofibrate reversed CACs and late EPC dysfunction in CHF patients. The present findings reveal the potential application of the AMPK agonist fenofibrate to reverse endothelial dysfunction in CHF patients.
Collapse
|
6
|
Molecular Mechanisms Underpinning Microparticle-Mediated Cellular Injury in Cardiovascular Complications Associated with Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6475187. [PMID: 30915196 PMCID: PMC6399542 DOI: 10.1155/2019/6475187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/13/2019] [Indexed: 12/15/2022]
Abstract
Microparticles (MPs) are small vesicles shed from the cytoplasmic membrane of healthy, activated, or apoptotic cells. MPs are very heterogeneous in size (100–1,000 nm), and they harbor proteins and surface antigens specific to cells they originate from. Virtually, all cells can shed MPs, and therefore, they can be found in all body fluids, but also entrapped in tissues. Of interest and because of their easy detection using a variety of techniques, circulating MPs were recognized as biomarkers for cell activation. MPs were also found to mediate critical actions in intercellular communication and transmitting biological messages by acting as paracrine vehicles. High plasma numbers of MPs were reported in many cardiovascular and metabolic disturbances that are closely associated with insulin resistance and low-grade inflammation and have been linked to adverse actions on cardiovascular function. This review highlights the involvement of MPs in cardiovascular complications associated with diabetes and discusses the molecular mechanisms that underpin the pathophysiological role of MPs in the onset and progression of cellular injury in diabetes.
Collapse
|
7
|
Joo MS, Koo JH, Kim TH, Kim YS, Kim SG. LRH1-driven transcription factor circuitry for hepatocyte identity: Super-enhancer cistromic analysis. EBioMedicine 2019; 40:488-503. [PMID: 30638865 PMCID: PMC6413675 DOI: 10.1016/j.ebiom.2018.12.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/19/2018] [Accepted: 12/26/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The injured liver loses normal function, with concomitant decrease of key identity genes. Super-enhancers contribute to mammalian cell identity. Here, we identified core transcription factors (TFs) that are active in hepatocytes, using genome-wide analysis and hierarchical ordering of super-enhancer distribution. METHODS Expression of core TFs was assessed in a cohort of patients with hepatitis or cirrhosis and animal models. Quantitative PCR, chromatin immunoprecipitation assays, and hydrodynamic gene delivery methods were used to assess gene regulation and hepatocyte viability. RNA-sequencing data were generated to investigate the role of LRH1 in hepatocyte protection from injury. RESULTS Network analysis of super-enhancer-associated gene interactions and expression arrays for cohorts of patients with hepatitis and cirrhosis enabled us to identify a super-enhancer-associated network, and LRH1, HNF4α, PPARα, and RXRα as core TFs. In mouse models, expression of core TFs was robustly inhibited by single and multiple challenge(s) with liver toxicant. RNA-seq analysis revealed changes in expression in the super-enhancer-associated genes sensitively biased toward repression by intoxication. LRH1 gene delivery prevented the loss of hepatic super-enhancer-associated signaling circuitry in toxicant-challenged mice, and protected the liver from injury, indicating the role of LRH1 in hepatocyte identity and viability. In hepatocytes, overexpression of each core TF promoted induction of other TFs. CONCLUSION Overall, this study identified LRH1-driven pathway as a circuitry responsible for hepatocyte identity by using cistromic analysis, improving our understanding of liver pathophysiology and identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Min Sung Joo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ja Hyun Koo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Tae Hyun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yun Seok Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Lo Gullo A, Aragona CO, Scuruchi M, Versace AG, Saitta A, Imbalzano E, Loddo S, Campo GM, Mandraffino G. Endothelial progenitor cells and rheumatic disease modifying therapy. Vascul Pharmacol 2018; 108:8-14. [PMID: 29842927 DOI: 10.1016/j.vph.2018.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/12/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023]
Abstract
Rheumatic diseases are associated with accelerated atherosclerosis and with increased risk of cardiovascular morbidity and mortality. The mechanisms underlying the higher prevalence of cardiovascular disease are not completely clarified, but it is likely that a pivotal role is played by vascular inflammation and consequently to altered vascular endothelium homeostasis. Also, high prevalence of traditional risk factors, proatherogenic activation and endothelial dysfunction further contribute to vascular damage. Circulating endothelial progenitor cells (EPCs) can restore dysfunctional endothelium and protect against atherosclerotic vascular disease. However, abnormalities in number and function of these cells in patients with rheumatic condition have been extensively reported. During the last years, growing interest in the mechanisms of endothelial renewal and its potential as a therapy for CVD has been shown; in addition, pioneering studies show that EPC dysfunction might be improved with pharmacological strategies. However, how to restore EPC function, and whether achieving this aim may be effective in preventing cardiovascular complications in rheumatic disease, remain to be established. In this review we report an overview on the current stand of knowledge on the effect of pharmaceutical and lifestyle intervention in improving EPCs number and function in rheumatic disease.
Collapse
Affiliation(s)
- Alberto Lo Gullo
- Department of Clinical and Experimental Medicine, University of Messina, Italy.
| | | | - Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | | | - Antonino Saitta
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Saverio Loddo
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | | | | |
Collapse
|
9
|
Exosomes of Endothelial Progenitor Cells Inhibit Neointima Formation After Carotid Artery Injury. J Surg Res 2018; 232:398-407. [PMID: 30463748 DOI: 10.1016/j.jss.2018.06.066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/04/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exosomes released from endothelial progenitor cells (EPCs) play a protective role in various disease models. Both endothelial cell (EC) damage and smooth muscle cell (SMC) proliferation are involved in the pathological process of restenosis after angioplasty and stenting. Few studies have focused on the therapeutic role of exosomes in EC damage and SMC proliferation. In this study, we sought to investigate the effect of exosomes released by human fetal aorta-derived EPCs on the rat carotid artery balloon injury model in vivo. We also sought to determine the effect of exosomes on both ECs and SMCs in vitro. METHODS Exosomes (Exo group) or saline (Con group) were injected in rat carotid balloon injury model animals. The rats were sacrificed after 2, 4, 14, and 28 d, and injured carotid specimens were collected for Evans blue staining, hematoxylin-eosin staining, and immunohistochemistry. RESULTS When the Con group and the Exo group were compared, the reendothelialized areas were not significantly different after 2 or 4 d, as shown by Evans blue staining. The hematoxylin-eosin results showed that the intimal to medial area ratio was slightly but not significantly higher in the Exo group after 2 and 4 d. The immunohistochemistry results showed that the proliferation of SMCs was slightly higher in the Exo group after 2 and 4 d, but the difference was not significant. The reendothelialization area of the Con group was significantly smaller than that of the Exo group at day 14. Both the intimal to medial area ratio and SMC proliferation in the Exo group were significantly smaller than those of the Con group at 14 or 28 d. In the in vitro study, exosome treatment significantly enhanced the proliferation and migration of both ECs and SMCs. CONCLUSIONS Exosomes derived from EPCs could inhibit neointimal hyperplasia after carotid artery injury in rats. The protective effect of exosomes may manifest through the promotion of EC repair rather than direct suppression of proliferation and migration of smooth muscles cells.
Collapse
|
10
|
Vergori L, Lauret E, Soleti R, Andriantsitohaina R, Carmen Martinez M. Microparticles Carrying Peroxisome Proliferator-Activated Receptor Alpha Restore the Reduced Differentiation and Functionality of Bone Marrow-Derived Cells Induced by High-Fat Diet. Stem Cells Transl Med 2017; 7:135-145. [PMID: 29080294 PMCID: PMC5746153 DOI: 10.1002/sctm.17-0098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/21/2017] [Indexed: 01/17/2023] Open
Abstract
Metabolic pathologies such as diabetes and obesity are associated with decreased level of circulating and bone marrow (BM)-derived endothelial progenitor cells (EPCs). It is known that activation of peroxisome proliferator-activated receptor alpha (PPARα) may stimulate cell differentiation. In addition, microparticles (MPs), small membrane vesicles produced by activated and apoptotic cells, are able to reprogram EPCs. Here, we evaluated the role of MPs carrying PPARα on both phenotype and function of progenitor cells from mice fed with a high-fat diet (HFD). HFD reduced circulating EPCs and, after 7 days of culture, BM-derived EPCs and monocytic progenitor cells from HFD-fed mice displayed impaired differentiation. At the same time, we show that MPs bearing PPARα, MPsPPARα+/+ , increased the differentiation of EPCs and monocytic progenitors from HFD-fed mice, whereas MPs taken from PPARα knockout mice (MPsPPARα-/- ) had no effect on the differentiation of all types of progenitor cells. Furthermore, MPsPPARα+/+ increased the ability of progenitor cells to promote in vivo angiogenesis in mice fed with HFD. The in vitro and in vivo effects of MPsPPARα+/+ were abolished in presence of MK886, a specific inhibitor of PPARα. Collectively, these data highlight the ability of MPs carrying PPARα to restore the failed differentiation and functionality of BM-derived cells induced by HFD. Stem Cells Translational Medicine 2018;7:135-145.
Collapse
Affiliation(s)
- Luisa Vergori
- INSERM U1063, Stress oxydant et pathologies métaboliques, Faculté de Médecine, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Emilie Lauret
- INSERM U1063, Stress oxydant et pathologies métaboliques, Faculté de Médecine, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Raffaella Soleti
- INSERM U1063, Stress oxydant et pathologies métaboliques, Faculté de Médecine, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Ramaroson Andriantsitohaina
- INSERM U1063, Stress oxydant et pathologies métaboliques, Faculté de Médecine, UNIV Angers, Université Bretagne Loire, Angers, France.,Centre Hospitalo-Universitaire d'Angers, Angers, France
| | - M Carmen Martinez
- INSERM U1063, Stress oxydant et pathologies métaboliques, Faculté de Médecine, UNIV Angers, Université Bretagne Loire, Angers, France.,Centre Hospitalo-Universitaire d'Angers, Angers, France
| |
Collapse
|
11
|
French KC, Antonyak MA, Cerione RA. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Semin Cell Dev Biol 2017; 67:48-55. [PMID: 28104520 DOI: 10.1016/j.semcdb.2017.01.002] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/22/2016] [Accepted: 01/06/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs), lipid bilayer-enclosed structures that contain a variety of biological molecules shed by cells, are increasingly becoming appreciated as a major form of cell-to-cell communication. Indeed, EVs have been shown to play important roles in several physiological processes, as well as diseases such as cancer. EVs dock on to the surfaces of recipient cells where they transmit signals from the cell surface and/or transfer their contents into cells to elicit functional responses. EV docking and uptake by cells represent critical, but poorly understood processes. Here, we focus on the mechanisms by which EVs dock and transfer their contents to cells. Moreover, we highlight how these findings may provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Kinsley C French
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14850, United States
| | - Marc A Antonyak
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14850, United States
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14850, United States.
| |
Collapse
|
12
|
Maamoun H, Zachariah M, McVey JH, Green FR, Agouni A. Heme oxygenase (HO)-1 induction prevents Endoplasmic Reticulum stress-mediated endothelial cell death and impaired angiogenic capacity. Biochem Pharmacol 2016; 127:46-59. [PMID: 28012960 DOI: 10.1016/j.bcp.2016.12.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022]
Abstract
Most of diabetic cardiovascular complications are attributed to endothelial dysfunction and impaired angiogenesis. Endoplasmic Reticulum (ER) and oxidative stresses were shown to play a pivotal role in the development of endothelial dysfunction in diabetes. Hemeoxygenase-1 (HO-1) was shown to protect against oxidative stress in diabetes; however, its role in alleviating ER stress-induced endothelial dysfunction remains not fully elucidated. We aim here to test the protective role of HO-1 against high glucose-mediated ER stress and endothelial dysfunction and understand the underlying mechanisms with special emphasis on oxidative stress, inflammation and cell death. Human Umbilical Vein Endothelial Cells (HUVECs) were grown in either physiological or intermittent high concentrations of glucose for 5days in the presence or absence of Cobalt (III) Protoporphyrin IX chloride (CoPP, HO-1 inducer) or 4-Phenyl Butyric Acid (PBA, ER stress inhibitor). Using an integrated cellular and molecular approach, we then assessed ER stress and inflammatory responses, in addition to apoptosis and angiogenic capacity in these cells. Our results show that HO-1 induction prevented high glucose-mediated increase of mRNA and protein expression of key ER stress markers. Cells incubated with high glucose exhibited high levels of oxidative stress, activation of major inflammatory and apoptotic responses [nuclear factor (NF)-κB and c-Jun N-terminal kinase (JNK)] and increased rate of apoptosis; however, cells pre-treated with CoPP or PBA were fully protected. In addition, high glucose enhanced caspases 3 and 7 cleavage and activity and augmented cleaved poly ADP ribose polymerase (PARP) expression whereas HO-1 induction prevented these effects. Finally, HO-1 induction and ER stress inhibition prevented high glucose-induced reduction in NO release and impaired the angiogenic capacity of HUVECs, and enhanced vascular endothelial growth factor (VEGF)-A expression. Altogether, we show here the critical role of ER stress-mediated cell death in diabetes-induced endothelial dysfunction and impaired angiogenesis and underscore the role of HO-1 induction as a key therapeutic modulator for ER stress response in ischemic disorders and diabetes. Our results also highlight the complex interplay between ER stress response and oxidative stress.
Collapse
Affiliation(s)
- Hatem Maamoun
- University of Surrey, Faculty of Health and Medical Sciences, School of Biosciences & Medicine, Guildford GU2 7XH, United Kingdom
| | - Matshediso Zachariah
- University of Surrey, Faculty of Health and Medical Sciences, School of Biosciences & Medicine, Guildford GU2 7XH, United Kingdom
| | - John H McVey
- University of Surrey, Faculty of Health and Medical Sciences, School of Biosciences & Medicine, Guildford GU2 7XH, United Kingdom
| | - Fiona R Green
- University of Surrey, Faculty of Health and Medical Sciences, School of Biosciences & Medicine, Guildford GU2 7XH, United Kingdom
| | - Abdelali Agouni
- Qatar University, College of Pharmacy, Pharmaceutical Sciences Section, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
13
|
Wei H, Zhao X, Yuan R, Dai X, Li Y, Liu L. Effects of PB-EPCs on Homing Ability of Rabbit BMSCs via Endogenous SDF-1 and MCP-1. PLoS One 2015; 10:e0145044. [PMID: 26660527 PMCID: PMC4682485 DOI: 10.1371/journal.pone.0145044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023] Open
Abstract
Traumas, infections, tumors, and some congenital malformations can lead to bone defects or even bone loss. The goal of the present study was to investigate whether inclusion of endothelial progenitor cells derived from peripheral blood (PB–EPCs) in cell-seeded partially deproteinized bone (PDPB) implants would stimulate recruitment of systemically injected bone marrow stromal cells (BMSCs) to the implant. Methods: BMSCs were injected intravenously with lentiviral expression vector expressing enhanced green fluorescent protein (eGFP) for tracing. Recruitment of eGFP-positive BMSCs was tested for the following implant configurations: 1) seeded with both BMSC and PB-EPC, 2) BMSC alone, 3) PB-EPC alone, and 4) unseeded PDPB. Protein and mRNA levels of endogenous stromal-derived factor-1 (SDF-1) and its receptor CXCR4, as well as monocyte chemotactic protein-1 (MCP-1) and its receptor CCR2, were evaluated on the 8th week. Immunohistochemical staining was performed to determine eGFP-positive areas at the defective sites. Masson’s trichrome staining was conducted to observe the distribution of collagen deposition and evaluate the extent of osteogenesis. Results: The mRNA and protein levels of SDF-1 and CXCR4 in the co-culture group were higher than those in other groups (p < 0.05) 8 weeks after the surgery. MCP-1 mRNA level in the co-culture group was also higher than that in the other groups (p < 0.05). Immunohistochemical assays revealed that the area covered by eGFP-positive cells was larger in the co-culture group than in the other groups (p < 0.05) after 4 weeks. Masson’s trichrome staining revealed better osteogenic potential of the co-culture group compared to the other groups (p < 0.05). Conclusion: These experiments demonstrate an association between PB-EPC and BMSC recruitment mediated by the SDF-1/CXCR4 axis that can enhance repair of bone defects.
Collapse
Affiliation(s)
- Hanxiao Wei
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Xian Zhao
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Ruihong Yuan
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Xiaoming Dai
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Yisong Li
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Liu Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
- * E-mail:
| |
Collapse
|
14
|
Saleh HA, Kabeer BS. Microparticles: Biomarkers and effectors in the cardiovascular system. Glob Cardiol Sci Pract 2015. [DOI: 10.5339/gcsp.2015.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
15
|
Vergori L, Lauret E, Gaceb A, Beauvillain C, Andriantsitohaina R, Martinez MC. PPARα Regulates Endothelial Progenitor Cell Maturation and Myeloid Lineage Differentiation Through a NADPH Oxidase-Dependent Mechanism in Mice. Stem Cells 2015; 33:1292-303. [DOI: 10.1002/stem.1924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 10/31/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Luisa Vergori
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques; Institut de Biologie en Santé Université d'Angers; Angers France
- Department of Biosciences, Biotechnologies and Biofarmaceutic; University of Bari; Bari Italy
- Centre Hospitalo-Universitaire d'Angers; Angers France
| | - Emilie Lauret
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques; Institut de Biologie en Santé Université d'Angers; Angers France
| | - Abderahim Gaceb
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques; Institut de Biologie en Santé Université d'Angers; Angers France
| | - Céline Beauvillain
- Centre Hospitalo-Universitaire d'Angers; Angers France
- INSERM U892, CNRS UMR6299; Université d'Angers; Angers France
| | - Ramaroson Andriantsitohaina
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques; Institut de Biologie en Santé Université d'Angers; Angers France
- Centre Hospitalo-Universitaire d'Angers; Angers France
| | - M. Carmen Martinez
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques; Institut de Biologie en Santé Université d'Angers; Angers France
- Centre Hospitalo-Universitaire d'Angers; Angers France
| |
Collapse
|
16
|
Sheu JJ, Lee FY, Wallace CG, Tsai TH, Leu S, Chen YL, Chai HT, Lu HI, Sun CK, Yip HK. Administered circulating microparticles derived from lung cancer patients markedly improved angiogenesis, blood flow and ischemic recovery in rat critical limb ischemia. J Transl Med 2015; 13:59. [PMID: 25889721 PMCID: PMC4369091 DOI: 10.1186/s12967-015-0381-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/05/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We hypothesized that lung cancer patient's circulating microparticles (Lc-MPs) could promote angiogenesis, blood flow in ischemic zone and ischemic recovery in rat critical limb ischemia (CLI). METHODS To investigate the impact of MP therapy on reversing the setting of CLI, adult-male Sprague-Dawley rats (n=50) equally randomized into sham control (SC) (group 1), SC-Lc-MPs (1.0 x 10(7) particles) (group 2), CLI (group 3), CLI-Hs-MPs (MPs from healthy-subject) (group 4), and CLI-Lc-MPs (group 5) were sacrificed by post-CLI day-14. RESULTS In vitro study showed that Lc-MPs enhanced VEGFR2 expression, angiogenesis, nitric-oxide production, and endothelial cell proliferation (all p<0.005). By days 7 and 14, Laser Doppler showed significantly higher ischemic/normal blood-flow ratio in groups 1 and 2 compared with group 3, and was significantly higher in group 4 and further elevated in group 5 (p<0.0001). Numbers of small vessels and endothelial markers (CD31(+) and vWF(+) cells) and protein expressions (eNOS, CD31) exhibited a pattern identical to Lasre Doppler among the five groups (all p<0.001). Pro-angiogenic factors (VEGF, CXCR4, SDF-1α, HGF) at cellular and protein levels showed a significant step-wise increase from groups 1 and 2 to groups 3, 4, and 5 (all p<0.001). Protein expressions of fibrotic (Smad3, TGF-β) and apoptotic (mitochondrial Bax, cleaved caspase 3, and PARP) biomarkers displayed an opposite pattern compared to that of Laser Doppler, whereas the protein expressions of anti-fibrotic (Smad1/5, BMP-2) and anti-apoptotic (Bcl-2) biomarkers showed an identical pattern compared with that of Laser Doppler among groups 1 to 3, and 5 (all p<0.001). CONCLUSION Administration of Lc-MPs augmented angiogenesis and restored blood flow in a rat of CLI.
Collapse
Affiliation(s)
- Jiunn-Jye Sheu
- Division of Thoracic and Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Fan-Yen Lee
- Division of Thoracic and Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | | | - Tzu-Hsien Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Steve Leu
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Yung-Lung Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Hung-I Lu
- Division of Thoracic and Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan.
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan. .,Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan. .,Institute of Shock Wave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
17
|
Werner CM, Schirmer SH, Gensch C, Pavlickova V, Pöss J, Wright MB, Böhm M, Laufs U. The dual PPARα/γ agonist aleglitazar increases the number and function of endothelial progenitor cells: implications for vascular function and atherogenesis. Br J Pharmacol 2014; 171:2685-703. [PMID: 24467636 DOI: 10.1111/bph.12608] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/30/2013] [Accepted: 01/16/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Aleglitazar is a dual PPARα/γ agonist but little is known about its effects on vascular function and atherogenesis. Hence, we characterized its effects on circulating angiogenic cells (CAC), neoangiogenesis, endothelial function, arteriogenesis and atherosclerosis in mice. EXPERIMENTAL APPROACH C57Bl/6 wild-type (WT, normal chow), endothelial NOS (eNOS)(-/-) (normal chow) and ApoE(-/-) (Western-type diet) mice were treated with aleglitazar (10 mg·kg(-1) ·day(-1) , i.p.) or vehicle. KEY RESULTS Aleglitazar enhanced expression of PPARα and PPARγ target genes, normalized glucose tolerance and potently reduced hepatic fat in ApoE(-/-) mice. In WT mice, but not in eNOS(-/-) , aleglitazar up-regulated Sca-1/VEGFR2-positive CAC in the blood and bone marrow and up-regulated diLDL/lectin-positive CAC. Aleglitazar augmented CAC migration and enhanced neoangiogenesis. In ApoE(-/-) mice, aleglitazar up-regulated CAC number and function, reduced markers of vascular inflammation and potently improved perfusion restoration after hindlimb ischaemia and aortic endothelium-dependent vasodilatation. This was associated with markedly reduced formation of atherosclerotic plaques. In human cultured CAC from healthy donors and patients with coronary artery disease with or without diabetes mellitus, aleglitazar increased migration and colony-forming units in a concentration-dependent manner. Furthermore, oxidative stress-induced CAC apoptosis and expression of p53 were reduced, while telomerase activity and expression of phospho-eNOS and phospho-Akt were elevated. Comparative agonist and inhibitor experiments revealed that aleglitazar's effects on CAC migration and colony-forming units were mediated by both PPARα and PPARγ signalling and required Akt. CONCLUSIONS AND IMPLICATIONS Aleglitazar augments the number, function and survival of CAC, which correlates with improved vascular function, enhanced arteriogenesis and prevention of atherosclerosis in mice.
Collapse
Affiliation(s)
- C M Werner
- Klinik für Innere Medizin III (Kardiologie, Angiologie und Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Endothelial progenitor cells (EPCs) are primitive endothelial precursors which are known to functionally contribute to the pathogenesis of disease. To date a number of distinct subtypes of these cells have been described, with differing maturation status, cellular phenotype, and function. Although there is much debate on which subtype constitutes the true EPC population, all subtypes have endothelial characteristics and contribute to neovascularisation. Vasculogenesis, the process by which EPCs contribute to blood vessel formation, can be dysregulated in disease with overabundant vasculogenesis in the context of solid tumours, leading to tumour growth and metastasis, and conversely insufficient vasculogenesis can be present in an ischemic environment. Importantly, it is widely known that transcription factors tightly regulate cellular phenotype and function by controlling the expression of particular target genes and in turn regulating specific signalling pathways. This suggests that transcriptional regulators may be potential therapeutic targets to control EPC function. Herein, we discuss the observed EPC subtypes described in the literature and review recent studies describing the role of a number of transcriptional families in the regulation of EPC phenotype and function in normal and pathological conditions.
Collapse
|
19
|
Wang Z, Moran E, Ding L, Cheng R, Xu X, Ma JX. PPARα regulates mobilization and homing of endothelial progenitor cells through the HIF-1α/SDF-1 pathway. Invest Ophthalmol Vis Sci 2014; 55:3820-32. [PMID: 24845641 DOI: 10.1167/iovs.13-13396] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The mechanism for the antiangiogenic activity of peroxisome proliferator-activated receptor alpha (PPARα) remains incompletely understood. Endothelial progenitor cells (EPC) are known to participate in neovascularization (NV). The purpose of this study was to investigate whether PPARα regulates EPC during retinal NV. METHODS Retinal NV was induced by oxygen-induced retinopathy (OIR). Mice with OIR were injected intraperitoneally with the PPARα agonist fenofibric acid (FA) or with adenovirus expressing PPARα (Ad-PPARα). Flow cytometry was used to quantify circulating and retinal EPC. Serum stromal cell-derived factor 1 (SDF-1) levels were measured by ELISA. Hypoxia was induced in primary human retinal capillary endothelial cells (HRCEC) and mouse brain endothelial cells (MBEC) by CoCl2. Levels of SDF-1 and hypoxia-inducible factor 1 alpha (HIF-1α) were measured by Western blotting. RESULTS Fenofibric acid and overexpression of PPARα attenuated the increase of circulating and retinal EPC, correlating with suppressed retinal NV in OIR mice at P17. The PPARα knockout enhanced the OIR-induced increase of circulating and retinal EPC. Fenofibric acid decreased retinal HIF-1α and SDF-1 levels as well as serum SDF-1 levels in the OIR model. In HRCEC, PPARα inhibited HIF-1α nuclear translocation and SDF-1 overexpression induced by hypoxia. Further, MBEC from PPARα(-/-) mice showed more prominent activation of HIF-1α and overexpression of SDF-1 induced by hypoxia, compared with the wild-type (WT) MBEC. PPARα failed to block SDF-1 overexpression induced by a constitutively active mutant of HIF-1α, suggesting that regulation of SDF-1 by PPARα was through blockade of HIF-1α activation. CONCLUSIONS Peroxisome proliferator-activated receptor alpha suppresses ischemia-induced EPC mobilization and homing through inhibition of the HIF-1α/SDF-1 pathway. This represents a novel molecular mechanism for PPARα's antiangiogenic effects.
Collapse
Affiliation(s)
- Zhongxiao Wang
- Department of Ophthalmology, Shanghai First People's Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Elizabeth Moran
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Lexi Ding
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Rui Cheng
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Xun Xu
- Department of Ophthalmology, Shanghai First People's Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China
| | - Jian-xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
20
|
Schiro A, Wilkinson FL, Weston R, Smyth JV, Serracino-Inglott F, Alexander MY. Endothelial microparticles as conveyors of information in atherosclerotic disease. Atherosclerosis 2014; 234:295-302. [PMID: 24721189 DOI: 10.1016/j.atherosclerosis.2014.03.019] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/19/2022]
Abstract
Endothelial microparticles (EMPs) are complex submicron membrane-shed vesicles released into the circulation following endothelium cell activation or apoptosis. They are classified as either physiological or pathological, with anticoagulant or pro-inflammatory effects respectively. Endothelial dysfunction caused by inflammation is a key initiating event in atherosclerotic plaque formation. Athero-emboli, resulting from ruptured carotid plaques are a major cause of stroke. Current clinical techniques for arterial assessment, angiography and carotid ultrasound, give accurate information about stenosis but limited evidence on plaque composition, inflammation or vulnerability; as a result, patients with asymptomatic, or fragile carotid lesions, may not be identified and treated effectively. There is a need to discover novel biomarkers and develop more efficient diagnostic approaches in order to stratify patients at most risk of stroke, who would benefit from interventional surgery. Increasing evidence suggests that EMPs play an important role in the pathogenesis of cardiovascular disease, acting as a marker of damage, either exacerbating disease progression or triggering a repair response. In this regard, it has been suggested that EMPs have the potential to act as biomarkers of disease status. In this review, we will present the evidence to support this hypothesis and propose a novel concept for the development of a diagnostic device that could be implemented in the clinic.
Collapse
Affiliation(s)
- A Schiro
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK; Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK.
| | - F L Wilkinson
- Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - R Weston
- Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - J V Smyth
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK
| | - F Serracino-Inglott
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK; Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK
| | - M Y Alexander
- Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK; Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| |
Collapse
|
21
|
Thuma F, Zöller M. Outsmart tumor exosomes to steal the cancer initiating cell its niche. Semin Cancer Biol 2014; 28:39-50. [PMID: 24631836 DOI: 10.1016/j.semcancer.2014.02.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 02/22/2014] [Indexed: 12/14/2022]
Abstract
Exosomes are small vesicles that derive from endosomes and are delivered by many cells, including tumor cells that are a particular rich source of exosomes. Exosomes are suggested to be the most potent intercellular communicators. Being recovered in all body fluids, they can communicate with neighboring as well as distant cells. The latter was first described for dendritic cell exosomes that can initiate T cell activation. However, tumor exosomes (TEX) may impede this crosstalk. Besides with hematopoietic cells, TEX communicate with the tumor cell itself, but also with host stroma cells and endothelial cells. This crosstalk received much attention as there is strong evidence that TEX account for angiogenesis and premetastatic niche formation, which may proceed directly via binding and uptake of TEX by cells in the premetastatic organ or indirectly via TEX being taken up by hematopoietic progenitors in the bone marrow (BM), which mature toward lineages with immunosuppressive features or are forced toward premature release from the BM and homing into premetastatic organs. Knowing these deleterious activities of TEX, it becomes demanding to search for modes of therapeutic interference. I here introduce our hypothesis that metastasis formation may be hampered by tailored exosomes that outsmart TEX. The essential prerequisites are an in depth knowledge on TEX binding, uptake, binding-initiated signal transduction and uptake-promoted target cell reprogramming.
Collapse
Affiliation(s)
- Florian Thuma
- Department of Tumor Cell Biology, University Hospital of Surgery and German Cancer Research Center, Heidelberg, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery and German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
22
|
C/EBPβ mediates osteoclast recruitment by regulating endothelial progenitor cell expression of SDF-1α. PLoS One 2014; 9:e91217. [PMID: 24618682 PMCID: PMC3949754 DOI: 10.1371/journal.pone.0091217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/10/2014] [Indexed: 12/12/2022] Open
Abstract
Integration of tissue-engineered bone grafts with the host bone is vital for the healing of critical-size bone defects. An important aspect of this process is bone resorption, which must be carried out by osteoclasts derived from the host. However, the mechanism underlying recruitment of host osteoclast precursors to graft sites remains unclear. Endothelial progenitor cells (EPCs) mobilize from the bone marrow into the circulation and home to sites of angiogenesis such as tissue remodeling. Since EPCs express SDF-1, and C/EBPβ is known to regulate SDF-1α expression, we hypothesized that EPCs may recruit CXCR4-expressing host osteoclast precursors to the repair area and that this recruitment may be mediated through C/EBPβ signaling. Using an inflammatory EPC model we showed that EPCs upregulate protein levels of both SDF-1α and C/EBPβ. A luciferase assay confirmed that C/EBPβ acts on the SDF-1α promoter in these cells, and that binding is increased under conditions of inflammation, while silencing of C/EBPβ reduces expression of SDF-1 α and C/EBPβ. Using RAW264.7 cells as a model of osteoclastic monocyte precursors, we investigated their responses to migratory factors in EPC conditioned medium. We showed that RAW264.7 cells migrate towards conditioned medium from EPCs treated with IL-1β, an effect which could be abolished by silencing C/EBPβ in EPCs, and was almost completely blocked by silencing CXCR4 in RAW264.7 cells. These findings show that EPCs respond to inflammatory stimuli by signaling to osteoclast precursors via SDF-1, and that C/EBPβ mediates this response.
Collapse
|
23
|
Blumenschein GR, Saintigny P, Liu S, Kim ES, Tsao AS, Herbst RS, Alden C, Lee JJ, Tang X, Stewart DJ, Kies MS, Fossella FV, Tran HT, Mao L, Hicks ME, Erasmus J, Gupta S, Girard L, Peyton M, Diao L, Wang J, Davis SE, Minna J, Wistuba I, Hong WK, Heymach JV, Lippman SM. Comprehensive biomarker analysis and final efficacy results of sorafenib in the BATTLE trial. Clin Cancer Res 2013; 19:6967-75. [PMID: 24166906 PMCID: PMC3905243 DOI: 10.1158/1078-0432.ccr-12-1818] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To report the clinical efficacy of sorafenib and to evaluate biomarkers associated with sorafenib clinical benefit in the BATTLE (Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination) program. PATIENTS AND METHODS Patients with previously treated non-small cell lung cancer (NSCLC) received sorafenib until progression or unacceptable toxicity. Eight-week disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) were assessed. Prespecified biomarkers included K-RAS, EGFR, and B-RAF mutations, and EGFR gene copy number. Gene expression profiles from NSCLC cell lines and patient tumor biopsies with wild-type EGFR were used to develop a sorafenib sensitivity signature (SSS). RESULTS A total of 105 patients were eligible and randomized to receive sorafenib. Among 98 patients evaluable for eight-week DCR, the observed DCR was 58.2%. The median PFS and OS were 2.83 [95% confidence interval (CI), 2.04-3.58] and 8.48 months (95% CI, 5.78-10.97), respectively. Eight-week DCR was higher in patients with wild-type EGFR than patients with EGFR mutation (P = 0.012), and in patients with EGFR gene copy number gain (FISH-positive) versus patients FISH-negative (P = 0.048). In wild-type EGFR tumors, the SSS was associated with improved PFS (median PFS 3.61 months in high SSS vs. 1.84 months in low SSS; P = 0.026) but not with eight-week DCR. Increased expression of fibroblast growth factor-1, NF-κB, and hypoxia pathways were identified potential drivers of sorafenib resistance. CONCLUSION Sorafenib demonstrates clinical activity in NSCLC, especially with wild-type EGFR. SSS was associated with improved PFS. These data identify subgroups that may derive clinical benefit from sorafenib and merit investigation in future trials.
Collapse
Affiliation(s)
- George R. Blumenschein
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Pierre Saintigny
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Suyu Liu
- Biostatistics and Applied Mathematics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Edward S. Kim
- Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Charlotte, North Carolina
| | - Anne S. Tsao
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Roy S. Herbst
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut
| | - Christine Alden
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - J. Jack Lee
- Biostatistics and Applied Mathematics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Ximing Tang
- Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - Merrill S. Kies
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Frank V. Fossella
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Hai T. Tran
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - L. Mao
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland
| | - Marshall E. Hicks
- Diagnostic Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeremy Erasmus
- Diagnostic Radiology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Sanjay Gupta
- Diagnostic Radiology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Luc Girard
- The Hamon Center for Therapeutic Oncology Research and Departments of Internal Medicine and Pharmacology at the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael Peyton
- The Hamon Center for Therapeutic Oncology Research and Departments of Internal Medicine and Pharmacology at the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lixia Diao
- Bioinformatics and Computational Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Bioinformatics and Computational Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Suzanne E. Davis
- Division of Cancer Medicine at The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - John Minna
- The Hamon Center for Therapeutic Oncology Research and Departments of Internal Medicine and Pharmacology at the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ignacio Wistuba
- Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Waun K. Hong
- Division of Cancer Medicine at The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - John V. Heymach
- Thoracic/Head&Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
24
|
Zöller M. Pancreatic cancer diagnosis by free and exosomal miRNA. World J Gastrointest Pathophysiol 2013; 4:74-90. [PMID: 24340225 PMCID: PMC3858795 DOI: 10.4291/wjgp.v4.i4.74] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/01/2013] [Accepted: 09/18/2013] [Indexed: 02/06/2023] Open
Abstract
Patients with pancreatic adenocarcinoma (PaCa) have a dismal prognosis. This is in part due to late diagnosis prohibiting surgical intervention, which provides the only curative option as PaCa are mostly chemo- and radiation resistance. Hope is raised on a reliable non-invasive/minimally invasive diagnosis that is still missing. Recently two diagnostic options are discussed, serum MicroRNA (miRNA) and serum exosomes. Serum miRNA can be free or vesicle-, particularly, exosomes-enclosed. This review will provide an overview on the current state of the diagnostic trials on free serum miRNA and proceed with an introduction of exosomes that use as a diagnostic tool in serum and other body fluids has not received sufficient attention, although serum exosome miRNA in combination with protein marker expression likely will increase the diagnostic and prognostic power. By their crosstalk with host cells, which includes binding-initiated signal transduction, as well as reprogramming target cells via the transfer of proteins, mRNA and miRNA exosomes are suggested to become a most powerful therapeutics. I will discuss which hurdles have still to be taken as well as the different modalities, which can be envisaged to make therapeutic use of exosomes. PaCa are known to most intensely crosstalk with the host as apparent by desmoplasia and frequent paraneoplastic syndromes. Thus, there is hope that the therapeutic application of exosomes brings about a major breakthrough.
Collapse
|
25
|
Shirinsky IV, Shirinsky VS. Treatment of erosive osteoarthritis with peroxisome proliferator-activated receptor alpha agonist fenofibrate: a pilot study. Rheumatol Int 2013; 34:613-6. [PMID: 23620259 DOI: 10.1007/s00296-013-2766-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/18/2013] [Indexed: 01/21/2023]
Abstract
Hand osteoarthritis (HOA) is a common condition associated with high disease burden and frequently accompanied by comorbidities including dyslipidemia, atherosclerosis and obesity. The most debilitating HOA phenotype is erosive HOA (EHOA), characterized by synovial inflammation, formation of erosions, and substantial decline in hand function. Currently, there is no proven symptomatic treatment for the EHOA. Due to their broad spectrum effects directed on lipid metabolism, inflammation and pain, the agonists of peroxisome proliferator-activated receptor alpha or fibrates are a candidate class of drugs for the treatment of EHOA. In this study, we assessed the influence of fenofibrate treatment on clinical efficacy parameters, in vivo cytokine and adipokine production and concentrations of endothelial progenitor cells (EPC) in patients with EHOA. Fourteen patients received treatment with 145 mg of fenofibrate/day for 12 weeks. Fenofibrate treatment was associated with significant decreases in pain score, tender joint count, duration of morning stiffness, disease activity score, Cochin index, and ESR. Eight (57.14 %) patients developed Outcome Measures in Rheumatology Clinical Trials-Osteoarthritis Research Society response at the end of treatment. Paracetamol consumption did not change during the treatment course. There was a significant reduction in triglyceride levels. No changes were detected in serum pro-inflammatory cytokine and adipokine concentrations while circulating IL-10 levels significantly decreased. There were no differences in circulating EPC numbers before and after the treatment. Fenofibrate was well tolerated, no patient experienced disease flare during the treatment. In conclusion, in EHOA patients, fenofibrate is associated with pleiotropic effects on pain, inflammation, and lipid profile. Larger, controlled studies are needed to confirm these results.
Collapse
Affiliation(s)
- Ivan V Shirinsky
- Laboratory of Clinical Immunopharmacology, Institute of Clinical Immunology RAMS, 6 Zalesskogo str., 630099, Novosibirsk, Russia,
| | | |
Collapse
|
26
|
Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY, Huang YC, Liao WL, Huang HC, Liu PL, Chen YH. Endothelial progenitor cell dysfunction in cardiovascular diseases: role of reactive oxygen species and inflammation. BIOMED RESEARCH INTERNATIONAL 2012; 2013:845037. [PMID: 23484163 PMCID: PMC3591199 DOI: 10.1155/2013/845037] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 11/13/2012] [Indexed: 12/31/2022]
Abstract
Endothelial progenitor cells (EPCs) move towards injured endothelium or inflamed tissues and incorporate into foci of neovascularisation, thereby improving blood flow and tissue repair. Patients with cardiovascular diseases have been shown to exhibit reduced EPC number and function. It has become increasingly apparent that these changes may be effected in response to enhanced oxidative stress, possibly as a result of systemic and localised inflammatory responses. The interplay between inflammation and oxidative stress affects the initiation, progression, and complications of cardiovascular diseases. Recent studies suggest that inflammation and oxidative stress modulate EPC bioactivity. Clinical medications with anti-inflammatory and antioxidant properties, such as statins, thiazolidinediones, angiotensin II receptor 1 blockers, and angiotensin-converting enzyme inhibitors, are currently administered to patients with cardiovascular diseases. These medications appear to exert beneficial effects on EPC biology. This review focuses on EPC biology and explores the links between oxidative stress, inflammation, and development of cardiovascular diseases.
Collapse
Affiliation(s)
- Chih-Pei Lin
- Department of Biotechnology and Laboratory Science in Medicine and Institute of Biotechnology in Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine, School of Medicine, Taipei Medical University and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Po-Hsun Huang
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wen-Chi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Yi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Chuen Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Chun Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
27
|
Shirinsky I, Polovnikova O, Kalinovskaya N, Shirinsky V. The effects of fenofibrate on inflammation and cardiovascular markers in patients with active rheumatoid arthritis: a pilot study. Rheumatol Int 2012; 33:3045-8. [PMID: 23263548 DOI: 10.1007/s00296-012-2613-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/09/2012] [Indexed: 11/29/2022]
Abstract
Peroxisome proliferator-activated receptors α (PPARα) agonists, or fibrates, are used in the treatment for dyslipidemia. Experimental data suggest that fibrates have anti-inflammatory properties, and PPARα is essential for the differentiation of endothelial progenitor cells (EPC) which diminished pool in rheumatoid arthritis (RA) contributes to accelerated atherosclerosis. The data on fibrates' effects in patients with RA are limited. The aim of this study was to investigate changes in disease activity, inflammatory markers, lipid profile, and circulating EPC in active RA patients treated with fenofibrate. Twenty-seven patients with active RA taking traditional disease-modifying antirheumatic drugs (DMARDs) were prescribed fenofibrate (145 mg/day) for 3 months. All patients received background traditional DMARDs in stable doses. The outcomes measured were clinical disease activity variables, circulating cytokines, adipokines, lipids, and EPC. Twenty-five patients completed the study. At the end of treatment, there was a significant reduction in DAS28, all individual DAS28 components except tender joint count, the duration of morning stiffness, and in the patient's assessment of disease activity. Fifteen (60 %) patients achieved good/moderate EULAR response, while 10 (40 %) patients satisfied ACR20 response criteria. Treatment with fenofibrate resulted in significant decrease in CRP and IL-6 concentrations and improvement in lipid profile. There was no change in the concentrations of circulating EPC. In conclusion, fenofibrate treatment is associated with reduced inflammation and improved lipid profile in RA patients. Large randomized controlled studies are needed to confirm these results and to define the role of fibrates in the treatment for RA.
Collapse
|
28
|
Baron M, Boulanger CM, Staels B, Tailleux A. Cell-derived microparticles in atherosclerosis: biomarkers and targets for pharmacological modulation? J Cell Mol Med 2012; 16:1365-76. [PMID: 22050954 PMCID: PMC3823207 DOI: 10.1111/j.1582-4934.2011.01486.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular diseases remain an important cause of morbi-mortality. Atherosclerosis, which predisposes to cardiovascular disorders such as myocardial infarction and stroke, develops silently over several decades. Identification of circulating biomarkers to evaluate cardiovascular event risk and pathology prognosis is of particular importance. Microparticles (MPs) are small vesicles released from cells upon apoptosis or activation. Microparticles are present in blood of healthy individuals. Studies showing a modification of their concentrations in patients with cardiovascular risk factors and after cardiovascular events identify MPs as potential biomarkers of disease. Moreover, the pathophysiological properties of MPs may contribute to atherosclerosis development. In addition, pharmacological compounds, used in the treatment of cardiovascular disease, can reduce plasma MP concentrations. Nevertheless, numerous issues remain to be solved before MP measurement can be applied as routine biological tests to improve cardiovascular risk prediction. In particular, prospective studies to identify the predictive values of MPs in pathologies such as cardiovascular diseases are needed to demonstrate whether MPs are useful biomarkers for the early detection of the disease and its progression.
Collapse
|
29
|
Mamidi MK, Pal R, Dey S, Bin Abdullah BJJ, Zakaria Z, Rao MS, Das AK. Cell therapy in critical limb ischemia: current developments and future progress. Cytotherapy 2012; 14:902-16. [DOI: 10.3109/14653249.2012.693156] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Dragoni S, Laforenza U, Bonetti E, Lodola F, Bottino C, Berra-Romani R, Carlo Bongio G, Cinelli MP, Guerra G, Pedrazzoli P, Rosti V, Tanzi F, Moccia F. Vascular endothelial growth factor stimulates endothelial colony forming cells proliferation and tubulogenesis by inducing oscillations in intracellular Ca2+ concentration. Stem Cells 2012; 29:1898-907. [PMID: 21905169 DOI: 10.1002/stem.734] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial progenitor cells (EPCs) home from the bone marrow to the site of tissue regeneration and sustain neovascularization after acute vascular injury and upon the angiogenic switch in solid tumors. Therefore, they represent a suitable tool for cell-based therapy (CBT) in regenerative medicine and provide a novel promising target in the fight against cancer. Intracellular Ca(2+) signals regulate numerous endothelial functions, such as proliferation and tubulogenesis. The growth of endothelial colony forming cells (ECFCs), which are EPCs capable of acquiring a mature endothelial phenotype, is governed by store-dependent Ca(2+) entry (SOCE). This study aimed at investigating the nature and the role of VEGF-elicited Ca(2+) signals in ECFCs. VEGF induced asynchronous Ca(2+) oscillations, whose latency, amplitude, and frequency were correlated to the growth factor dose. Removal of external Ca(2+) (0Ca(2+)) and SOCE inhibition with N-(4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl)-4-methyl-1,2,3-thiadiazole-5-carboxamide (BTP-2) reduced the duration of the oscillatory signal. Blockade of phospholipase C-γ with U73122, emptying the inositol-1,4,5-trisphosphate (InsP(3))-sensitive Ca(2+) pools with cyclopiazonic acid (CPA), and inhibition of InsP(3) receptors with 2-APB prevented the Ca(2+) response to VEGF. VEGF-induced ECFC proliferation and tubulogenesis were inhibited by the Ca(2+)-chelant, BAPTA, and BTP-2. NF-κB activation by VEGF was impaired by BAPTA, BTP-2, and its selective blocker, thymoquinone. Thymoquinone, in turn, suppressed VEGF-dependent ECFC proliferation and tubulogenesis. These data indicate that VEGF-induced Ca(2+) oscillations require the interplay between InsP(3)-dependent Ca(2+) release and SOCE, and promote ECFC growth and tubulogenesis by engaging NF-κB. This novel signaling pathway might be exploited to enhance the outcome of CBT and chemotherapy.
Collapse
Affiliation(s)
- Silvia Dragoni
- Department of Physiology, University of Pavia, Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jung C, Sörensson P, Saleh N, Arheden H, Rydén L, Pernow J. Circulating endothelial and platelet derived microparticles reflect the size of myocardium at risk in patients with ST-elevation myocardial infarction. Atherosclerosis 2012; 221:226-31. [DOI: 10.1016/j.atherosclerosis.2011.12.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 12/17/2011] [Accepted: 12/19/2011] [Indexed: 12/19/2022]
|
32
|
Soleti R, Martinez MC. Sonic Hedgehog on microparticles and neovascularization. VITAMINS AND HORMONES 2012; 88:395-438. [PMID: 22391314 DOI: 10.1016/b978-0-12-394622-5.00018-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neovascularization represents a pivotal process consisting in the development of vascular network during embryogenesis and adult life. Postnatally, it arises mainly through angiogenesis, which has physiological and pathological roles in health and disease. Blood vessel formation results as tightly regulated multistep process which needs coordination and precise regulation of the balance of proangiogenic and antiangiogenic factors. Sonic Hedgehog (SHH), a morphogen belonging to Hedgehog (HH) family proteins, is implicated in a remarkably wide variety of process, including vessel development. Recent evidence demonstrate that, in addition to the classic factors, microvesicles (MVs), both microparticles (MPs) and exosomes, small vesicles released distinct cellular compartments, are involved in modulation of neovascularization. MPs generated from T lymphocytes undergoing both activation and apoptosis harbor at their surface SHH and play a crucial role in modulation of neovascularization. They are able to modulate the different steps implicated in angiogenesis process in vitro and to enhance postischemic neovascularization in vivo. As the consequence, we suggest that the MPs carrying SHH contribute to generation of a vascular network and may represent a new therapeutic approach to treat pathologies associated with failed angiogenesis.
Collapse
|
33
|
Saito H, Yamamoto Y, Yamamoto H. Diabetes alters subsets of endothelial progenitor cells that reside in blood, bone marrow, and spleen. Am J Physiol Cell Physiol 2011; 302:C892-901. [PMID: 22159079 DOI: 10.1152/ajpcell.00380.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) derived from the bone marrow (BM) participate in maintaining endothelial integrity and vascular homeostasis. Reduced EPC number and function result in vascular complications in diabetes. EPCs are a population of cells existing in various differentiation stages, and their cell surface marker profiles change during the process of mobilization and maturation. Hence, a generally accepted marker combination and a standardized protocol for the quantification of EPCs remain to be established. To determine the EPC subsets that are affected by diabetes, we comprehensively analyzed 32 surface marker combinations of mouse peripheral blood (PB), BM, and spleen cells by multicolor flow cytometry. Ten subsets equivalent to previously reported mouse EPCs significantly declined in number in the PB of streptozotocin-induced diabetic mice, and this reduction was reversed by insulin treatment. The PI(-)Lin(-)c-Kit(-)Sca-1(+)Flk-1(-)CD34(-)CD31(+) EPC cluster, which can differentiate into mature endothelial cells in vitro, was the highest population in the PB, BM, and spleen and occurred 61 times more in the spleen than in the PB. The cell number significantly decreased in the BM as well as in the PB but paradoxically increased in the spleen under diabetic conditions. Insulin treatment reversed the decrease of EPC subsets in the BM and PB and reversed their increase in spleen. A similar tendency was observed in some of the major cell populations in db/db mice. To the best of our knowledge, we are the first to report spatial population changes in mouse EPCs by diabetes in the blood and in the BM across the spleen. Diminished circulating EPC supply by diabetes may be ascribed to impaired EPC production in the BM and to decreased EPC mobilization from the spleen, which may contribute to vascular dysfunction in diabetic conditions.
Collapse
Affiliation(s)
- Hidehito Saito
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Japan
| | | | | |
Collapse
|
34
|
Rautou PE, Vion AC, Amabile N, Chironi G, Simon A, Tedgui A, Boulanger CM. Microparticles, Vascular Function, and Atherothrombosis. Circ Res 2011; 109:593-606. [DOI: 10.1161/circresaha.110.233163] [Citation(s) in RCA: 291] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Membrane-shed submicron microparticles (MPs) are released after cell activation or apoptosis. High levels of MPs circulate in the blood of patients with atherothrombotic diseases, where they could serve as a useful biomarker of vascular injury and a potential predictor of cardiovascular mortality and major adverse cardiovascular events. Atherosclerotic lesions also accumulate large numbers of MPs of leukocyte, smooth muscle cell, endothelial, and erythrocyte origin. A large body of evidence supports the role of MPs at different steps of atherosclerosis development, progression, and complications. Circulating MPs impair the atheroprotective function of the vascular endothelium, at least partly, by decreased nitric oxide synthesis. Plaque MPs favor local inflammation by augmenting the expression of adhesion molecule, such as intercellular adhesion molecule -1 at the surface of endothelial cell, and monocyte recruitment within the lesion. In addition, plaque MPs stimulate angiogenesis, a key event in the transition from stable to unstable lesions. MPs also may promote local cell apoptosis, leading to the release and accumulation of new MPs, and thus creating a vicious circle. Furthermore, highly thrombogenic plaque MPs could increase thrombus formation at the time of rupture, together with circulating MPs released in this context by activated platelets and leukocytes. Finally, MPs also could participate in repairing the consequences of arterial occlusion and tissue ischemia by promoting postischemic neovascularization.
Collapse
Affiliation(s)
- Pierre-Emmanuel Rautou
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Anne-Clémence Vion
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Nicolas Amabile
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Gilles Chironi
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Alain Simon
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Alain Tedgui
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Chantal M. Boulanger
- From the INSERM (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), U970, Paris Cardiovascular Research Center PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité (P.E.R., A.C.V., N.A., G.C., A.S., A.T., C.M.B.), UMR-S970, Paris, France; Service de Cardiologie (N.A.), Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France; Centre de Médecine Préventive Cardiovasculaire (G.C., A.S.), AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
35
|
Microparticles: targets and tools in cardiovascular disease. Trends Pharmacol Sci 2011; 32:659-65. [PMID: 21794929 DOI: 10.1016/j.tips.2011.06.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 01/04/2023]
Abstract
Cells communicate with other cells not only via direct cell-cell contact and the production of signaling molecules but also through release of microparticles (MPs). MPs are small vesicles released from stimulated and/or apoptotic cells. They harbor membrane proteins that are characteristic of the original parent cell and intracellular components involved in cell signaling. MPs are considered to be both biomarkers and effectors of cell signaling that maintain and/or initiate cell dysfunction. Thus, MPs can evoke endothelial dysfunction by decreasing nitric oxide (NO) production and promoting vascular inflammation which favor the prothrombotic state in atherosclerosis. Novel pharmacological approaches targeting MP production or properties could be used to treat cardiovascular pathologies. Paradoxically, another useful approach might be to employ engineered MPs with modified compositions as therapeutic agents to correct cardiovascular pathologies. This review is focused on the mechanisms of MP formation and their effects on target cells under physiological or pathophysiological conditions.
Collapse
|
36
|
Chen J, Chen S, Chen Y, Zhang C, Wang J, Zhang W, Liu G, Zhao B, Chen Y. Circulating endothelial progenitor cells and cellular membrane microparticles in db/db diabetic mouse: possible implications in cerebral ischemic damage. Am J Physiol Endocrinol Metab 2011; 301:E62-71. [PMID: 21505143 PMCID: PMC3129837 DOI: 10.1152/ajpendo.00026.2011] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For determining the implications of circulating endothelial progenitor cells (cEPCs) and cellular membrane microparticles (MPs) in diabetic stroke, levels of EPCs, EPC-MPs, and endothelium-derived MPs (EMPs) and their correlations with blood glucose concentration, cerebral microvascular density (cMVD), and ischemic damage were investigated in type 2 diabetic db/db and db/+ (wild-type control) mice. Therapeutic efficacy of EPC infusion (preincubated with MPs) was also explored. Ischemic stroke was induced by middle cerebral artery occlusion (MCAO) surgery. Ischemic damage and cMVD were determined using histological analyses. The levels of cEPCs and MPs were determined using flow cytometric analyses. EPC generation and functions were evaluated by in vitro cell cultures. Results showed the following. 1) In db/db mice, the basal level of cEPCs was less and cMVDs were lower, but the levels of circulating EPC-MPs and EMPs were more; 2) MCAO induced a larger infarct volume and less of an increase in cEPCs in db/db mice; 3) the level of cEPCs correlated with blood glucose concentration (negatively), cMVD (positively), and ischemic damage (negatively), but the levels of EPC-MPs and EMPs correlated inversely with those parameters; 4) EPCs were reduced and dysfunctional in db/db mice, and preincubation with db/db MPs impaired EPC functions; and 5) infusion of EPCs preincubated with db/+ MPs increased the level of cEPCs and reduced ischemic damage, and these beneficial effects were reduced or lost in EPCs preincubated with db/db MPs. These data suggest that reduced cEPCs, impaired EPC generation/function, and increased production of MPs might be the mechanisms responsible for increased ischemic damage seen in db/db mice.
Collapse
MESH Headings
- Animals
- Blood Cells/pathology
- Blood Cells/physiology
- Brain Ischemia/blood
- Brain Ischemia/etiology
- Brain Ischemia/pathology
- Cell-Derived Microparticles/pathology
- Cells, Cultured
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Endothelial Cells/pathology
- Endothelial Cells/physiology
- Homozygote
- Hypoxia, Brain/blood
- Hypoxia, Brain/etiology
- Hypoxia, Brain/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Risk Factors
- Stem Cells/pathology
- Stem Cells/physiology
Collapse
Affiliation(s)
- Ji Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Considered during the past decades as cell dust, microparticles are now deemed true biomarkers and vectors of biological information between cells. Depending on their origin, the composition of microparticles varies and the subsequent message transported by them, such as proteins, mRNA, or miRNA, can differ. Recent studies have described microparticles as “cargos” of deleterious information in blood vessel wall under pathological situations such as hypertension, myocardial infarction, and metabolic syndrome. In addition, it has been reported that depending on their origin, microparticles also possess a therapeutic potential regarding angiogenesis. Microparticles can act directly through the interaction ligand/receptor or indirectly on angiogenesis by modulating soluble factor production involved in endothelial cell differentiation, proliferation, migration, and adhesion; by reprogramming endothelial mature cells; and by inducing changes in levels, phenotype, and function of endothelial progenitor cells. This results in an increase in formation of in vitro capillary-like tubes and the generation of new vessels in vivo under ischemic conditions, for instance. Taking into consideration these properties of microparticles, recent evidence provides new basis to expand the possibility that microparticles might be used as therapeutic tools in pathologies associated with an alteration of angiogenesis.
Collapse
Affiliation(s)
- M. Carmen Martinez
- From the INSERM U694, Mitochondrie Régulations et Pathologie, Université d'Angers, Rue des Capucins, Angers, France
| | - Ramaroson Andriantsitohaina
- From the INSERM U694, Mitochondrie Régulations et Pathologie, Université d'Angers, Rue des Capucins, Angers, France
| |
Collapse
|
38
|
Circulating microparticles: new insights into the biochemical basis of microparticle release and activity. Basic Res Cardiol 2011; 106:911-23. [PMID: 21691898 DOI: 10.1007/s00395-011-0198-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/06/2011] [Accepted: 06/08/2011] [Indexed: 02/07/2023]
Abstract
Circulating microparticles released from various cell types are present in healthy individuals and the number and composition of their membrane vary in different disorders. Long considered to be cellular debris, microparticles have been recently identified as regulatory vectors of intercellular cross-talk. Indeed, circulating microparticles represent a heterogeneous mixture of spheroids of diverse surface membrane glycoproteins and lipids, with diverse cytoplasm components, the pattern of which depends on the type of stimulation and pathophysiology of parental cells. Despite extensive research into the procoagulant and proinflammatory properties of microparticles, there are few data that can provide information on the mechanism(s) of their formation and biological effects. Although several mechanisms of microparticle release have been suggested, the precise order of the events associated with key features of microparticle formation, transmembrane phosphatidylserine redistribution and cytoskeleton disruption remain to be clarified. In this review, we provide an overview of the molecular mechanisms involved in microparticle formation, as well as the diverse physiological and pathological roles they are able to undertake. Understanding the mechanism(s) governing microparticle release processes may be critical to understanding their precise role in various pathophysiological processes and thus indicate new potential routes to therapy.
Collapse
|
39
|
Targeting Nuclear Hormone Receptors: PPARα Agonists as Potential Disease-Modifying Drugs for Rheumatoid Arthritis. Int J Rheumatol 2011; 2011:937843. [PMID: 21760804 PMCID: PMC3134189 DOI: 10.1155/2011/937843] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 04/12/2011] [Accepted: 04/26/2011] [Indexed: 01/21/2023] Open
Abstract
In recent years, peroxisome proliferator-activated receptors (PPARs) have received growing interest due to the broad spectrum of their biological activities. PPARα, an isoform of PPAR, plays an important role in lipid homeostasis and inflammation, which makes it a potential target for the treatment of chronic inflammatory disorders, including RA. This paper reviews studies on the properties of PPARα agonists which may be pertinent to the treatment of RA. These properties include effects on lipid metabolism, inflammation, and angiogenesis, as well as interference with glucocorticoid effects, and a potential role in gender dimorphism of autoimmune disorders. However, current clinical experience with this class of drugs in RA is limited. New studies are needed to elucidate whether PPARα agonism may be an effective treatment strategy for RA patients.
Collapse
|
40
|
Abstract
The peroxisome proliferator-activated receptors (PPARs) and the retinoid X receptors (RXRs) are ligand-activated transcription factors that coordinately regulate gene expression. This PPAR-RXR transcriptional complex plays a critical role in energy balance, including triglyceride metabolism, fatty acid handling and storage, and glucose homeostasis: processes whose dysregulation characterize obesity, diabetes, and atherosclerosis. PPARs and RXRs are also involved directly in inflammatory and vascular responses in endothelial and vascular smooth muscle cells. New insights into fundamental aspects of PPAR and RXR biology, and their actions in the vasculature, continue to appear. Although RXRs are obligate heterodimeric partners for PPAR action, the part that RXRs, and their endogenous retinoid mediators, exert in the vessel wall is less well understood. Biological insights into PPAR-RXRs may help inform interpretation of clinical trials with synthetic PPAR agonists and prospects for future PPAR therapeutics. Importantly, the extensive data establishing a key role for PPARs and RXRs in energy balance, inflammation, and vascular biology stands separately from the clinical experience with any given synthetic PPAR agonist. Both the basic science data and the clinical experience with PPAR agonists identify the need to better understand these important transcriptional regulators.
Collapse
Affiliation(s)
- Jorge Plutzky
- From Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|