1
|
Kidney Mesenchymal Stem Cell-derived Extracellular Vesicles Engineered to Express Erythropoietin Improve Renal Anemia in Mice with Chronic Kidney Disease. Stem Cell Rev Rep 2021; 18:980-992. [PMID: 33651336 DOI: 10.1007/s12015-021-10141-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2021] [Indexed: 12/29/2022]
Abstract
Extracellular vesicles (EVs) shed from kidney mesenchymal stem cells (KMSCs) show protective effects against acute kidney injury and progressive kidney fibrosis via mRNA transfer. Previous studies report improvement of renal anemia following administration of genetically modified MSCs or peritoneal mesothelial cells that secrete erythropoietin (EPO). Here, we determined whether EPO-secreting KMSC-derived EVs (EPO(+)-EVs) can improve renal anemia in mouse models of chronic kidney disease (CKD). The mouse CKD and renal anemia model was induced by electrocoagulation of the right renal cortex and sequential left nephrectomy. At six weeks post-nephrectomy, we observed significantly lower hemoglobin (10.4 ± 0.2 vs. 13.2 ± 0.2 g/dL) and significantly higher blood urea nitrogen and serum creatinine levels in CKD mice relative to controls (60.5 ± 0.5 and 0.37 ± 0.09 mg/dL vs. 19.9 ± 0.5 and 0.12 ± 0.02 mg/dL, respectively). Genetically engineered EPO(+)-KMSCs secreted 71 IU/mL EPO/106 cells/24 h in vitro, and EPO(+)-EVs isolated by differential ultracentrifugation expressed EPO mRNA and horizontally transferred EPO mRNA into target cells in vitro and in vivo. Furthermore, at two weeks post-injection of EPO(+)-KMSCs or EPO(+)-EVs into CKD mice with renal anemia, we observed significant increases in hemoglobin levels (11.7 ± 0.2 and 11.5 ± 0.2 vs. 10.1 ± 0.2 g/dL, respectively) and significantly lower serum creatinine levels at eight weeks in comparison to mice receiving vehicle control (0.30 ± 0.00 and 0.23 ± 0.03 vs. 0.43 ± 0.06 mg/dL, respectively). These results demonstrate that intraperitoneal administration of EPO(+)-EVs significantly increased hemoglobin levels and renal function in CKD mice, suggesting the efficacy of these genetically engineered EVs as a promising novel strategy for the treatment of renal anemia.
Collapse
|
2
|
Miyazaki M, Obata Y, Abe K, Furusu A, Koji T, Tabata Y, Kohno S. Gene Transfer Using Nonviral Delivery Systems. Perit Dial Int 2020. [DOI: 10.1177/089686080602600603] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In peritoneal dialysis, loss of peritoneal function is a major factor in treatment failure. The alterations in peritoneal function are related to structural changes in the peritoneal membrane, including peritoneal sclerosis with increased extracellular matrix. Although peritoneal sclerosis is considered reversible to some extent through peritoneal rest, which improves peritoneal function and facilitates morphological changes, there has been no therapeutic intervention and no drug against the development and progression of peritoneal sclerosis. Using recent biotechnological advances in genetic engineering, a strategy based on genetic modification of the peritoneal membrane could be a potential therapeutic maneuver against peritoneal sclerosis and peritoneal membrane failure. Before this gene therapy may be applied clinically, a safe and effective gene delivery system as well as the selection of a gene therapy method must be established. There are presently two kinds of gene transfer vectors: viral and nonviral. Viral vectors are used mainly as a gene delivery system in the field of continuous ambulatory peritoneal dialysis research; however, they have several problems such as immunogenicity and toxicity. On the other hand, nonviral vectors have several advantages over viral vectors. We review here gene transfer using nonviral vector systems in the peritoneum: electroporation, liposomes, and cationized gelatin microspheres. In the field of peritoneal dialysis, gene therapy research using nonviral vectors is presently limited. Improvement in delivery methods together with an intelligent design of targeted genes has brought about large degrees of enhancement in the efficiency, specificity, and temporal control of nonviral vectors.
Collapse
Affiliation(s)
- Masanobu Miyazaki
- Second Department of Internal Medicine, Department of Histology, Kyoto, Japan
- Miyazaki-Furukawa Clinic, Nagasaki
| | - Yoko Obata
- Second Department of Internal Medicine, Department of Histology, Kyoto, Japan
| | - Katsushige Abe
- Second Department of Internal Medicine, Department of Histology, Kyoto, Japan
| | - Akira Furusu
- Second Department of Internal Medicine, Department of Histology, Kyoto, Japan
| | - Takehiko Koji
- Cell Biology, Nagasaki University School of Medicine, Kyoto, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shigeru Kohno
- Second Department of Internal Medicine, Department of Histology, Kyoto, Japan
| |
Collapse
|
3
|
Herrick SE, Mutsaers SE. The Potential of Mesothelial Cells in Tissue Engineering and Regenerative Medicine Applications. Int J Artif Organs 2018; 30:527-40. [PMID: 17628854 DOI: 10.1177/039139880703000611] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Injury to the serosa through injurious agents such as radiation, surgery, infection and disease results in the loss of the protective surface mesothelium and often leads to fibrous adhesion formation. Mechanisms that increase the rate of mesothialisation are therefore actively being investigated in order to reduce the formation of adhesions. These include intraperitoneal delivery of cultured mesothelial cells as well as administration of factors that are known to increase mesothelial proliferation and migration. An exciting alternative that has only recently received attention, is the possible role of mesothelial progenitor cells in the repair and regeneration of denuded serosal areas. Accumulating evidence suggests that such a population exists and under certain conditions is able to form a number of defined cell types indicating a degree of plasticity. Such properties may explain the extensive use of mesothelial cells in various tissue engineering applications including the development of vascular conduits and peripheral nerve replacements. It is likely that with the rapid explosion in the fields of tissue engineering and regenerative medicine, a greater understanding of the potential of mesothelial progenitor cells to repair, replace and possibly regenerate damaged or defective tissue will be uncovered.
Collapse
Affiliation(s)
- S E Herrick
- School of Medicine, Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
| | | |
Collapse
|
4
|
Kawanishi K. Mesothelial cell transplantation: history, challenges and future directions. Pleura Peritoneum 2016; 1:135-143. [PMID: 30911617 PMCID: PMC6419540 DOI: 10.1515/pp-2016-0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022] Open
Abstract
Mesothelial cells line the surface of the pleura, pericardium, peritoneum and internal reproductive organs. One of their main functions is to act as a non-adhesive barrier to protect against physical damage, however, over the past decades their physiological and pathological properties have been revealed in association with a variety of conditions and diseases. Mesothelium has been used in surgical operations in clinical settings, such as omental patching for perforated peptic ulcers and in glutaraldehyde-treated autologous pericardium for aortic valve reconstruction. Various methods for mesothelial cell transplantation have also been established and developed, particularly within the area of tissue engineering, including scaffold and non-scaffold cell sheet technologies. However, the use of mesothelial cell transplantation in patients remains challenging, as it requires additional operations under general anesthesia in order to obtain enough intact cells for culture. Moreover, the current methods of mesothelial cell transplantation are expensive and are not yet available in clinical practice. This review firstly summarizes the history of the use of mesothelial cell transplantation in tissue engineering, and then critically discusses the barriers for the clinical application of mesothelial cell transplantation. Finally, the recent developments in xenotransplantation technologies are discussed to evaluate other feasible alternatives to mesothelial cell transplantation.
Collapse
Affiliation(s)
- Kunio Kawanishi
- Department of Cellular and Molecular Medicine, University of California, San Diego,9500 Gilman Drive, La Jolla, CA 92093–0687, USA
- Department of Surgical Pathology, Tokyo Women’s Medical University, 8–1, Kawada-cho, Shinjuku-ku, 162–8666, Tokyo, Japan
| |
Collapse
|
5
|
Gyabaah K, Aboushwareb T, Guimaraes Souza N, Yamaleyeva L, Varner A, Wang HJ, Atala A, Yoo JJ. Controlled regulation of erythropoietin by primary cultured renal cells for renal failure induced anemia. J Urol 2012; 188:2000-6. [PMID: 22999549 DOI: 10.1016/j.juro.2012.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Indexed: 11/19/2022]
Abstract
PURPOSE Renal failure induced anemia develops as a result of inadequate production of erythropoietin, which is the primary regulator of red blood cell production. We previously noted that culture expanded primary renal cells stably express erythropoietin and suggested that these cells may be used as a potential treatment for renal failure induced anemia. We investigated whether these cells are able to regulate erythropoietin expression in a controlled manner under different oxygen and environmental conditions. MATERIALS AND METHODS Primary rat renal cells were exposed to different hypoxic (0.1% to 1% O(2)) and normoxic environments. Erythropoietin expression was assessed using reverse transcriptase-polymerase chain reaction. Erythropoietin production was measured in culture medium using Meso Scale Discovery® assays. Results were plotted to compare different levels of production to the control. RESULTS Cultured renal cells expressed high levels of erythropoietin under hypoxia for up to 24 hours with a gradual decrease thereafter. However, erythropoietin expression was decreased when cells were switched from a hypoxic to a normoxic environment within the initial 24 hours. This indicated that cultured renal cells have the capacity to sense environmental oxygen tension and regulate erythropoietin expression accordingly. In addition, erythropoietin release in medium followed a pattern similar to that of gene expression under normoxic and hypoxic conditions. CONCLUSIONS These findings indicate that primary renal cells have the ability to regulate erythropoietin gene expression and release through environment dependent mechanisms. This also suggests that with further study the possibility exists of developing these cells as a potential method to treat renal failure induced anemia.
Collapse
Affiliation(s)
- Kenneth Gyabaah
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Scheidemann F, Therrien JP, Vogel J, Pfützner W. In vivosynthesis and secretion of erythropoietin by genetically modified primary human keratinocytes grafted onto immunocompromised mice. Exp Dermatol 2010; 19:289-97. [DOI: 10.1111/j.1600-0625.2009.00984.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
7
|
Nakav S, Naamani O, Chaimovitz C, Shaked G, Czeiger D, Zlotnik M, Douvdevani A. Regulation of adenosine system at the onset of peritonitis. Nephrol Dial Transplant 2009; 25:931-9. [PMID: 19861313 DOI: 10.1093/ndt/gfp542] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Adenosine, a potent regulator of inflammation, is produced under stressful conditions due to degradation of ATP/ADP by the ectoenzymes CD39 and CD73. Adenosine is rapidly degraded by adenosine deaminase (ADA) or phosphorylated in the cell by adenosine kinase (AK). From four known receptors to adenosine, A(1) (A(1)R) promotes inflammation by a G(i)-coupled receptor. We have previously shown that A(1)R is up-regulated in the first hours following bacterial inoculation. The aim of the current study is to characterize the inflammatory mediators that regulate adenosine-metabolizing enzymes and A(1)R at the onset of peritonitis. METHODS Peritonitis was induced in CD1 mice by intraperitoneal injection of Escherichia coli. TNFalpha and IL-6 levels were determined in peritoneal fluid by enzyme-linked immunosorbent assay. Adenosine-metabolizing enzymes and the A(1)R mRNA or protein levels were analyzed by quantitative PCR or by Western blot analysis, respectively. RESULTS We found that CD39 and CD73 were up-regulated in response to bacterial stimuli (6-fold the basal levels), while AK and ADA mRNA levels were down-regulated. Cytokine production and leukocyte recruitment were enhanced (2.5-fold) by treatment with an A(1)R agonist (2-chloro-N(6)-cyclopentyladenosine, 0.1 mg/kg) and reduced (2.5-3-fold) by the A(1)R antagonist (8-cyclopentyl-1, 3-dipropylxanthine, 1 mg/kg). In contrast to lipopolysaccharide, IL-1, TNF and IFNgamma, only low IL-6 levels (0.01 ng/ml), in the presence of its soluble IL-6R (sIL-6R), were found to promote A(1)R expression on mesothelial cells. In mice, administration of neutralizing antibody to IL-6R or soluble gp130-Fc (sgp130-Fc) blocked peritoneal A(1)R up-regulation following inoculation. CONCLUSION Bacterial products induce the production of adenosine by up-regulation of CD39 and CD73. Low IL-6-sIL-6R up-regulates the A(1)R to promote efficient inflammatory response against invading microorganisms.
Collapse
Affiliation(s)
- Sigal Nakav
- Department of Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | | | | | | | | | | | | |
Collapse
|
8
|
Nakav S, Kachko L, Vorobiov M, Rogachev B, Chaimovitz C, Zlotnik M, Douvdevani A. Blocking adenosine A2A receptor reduces peritoneal fibrosis in two independent experimental models. Nephrol Dial Transplant 2009; 24:2392-9. [PMID: 19221177 DOI: 10.1093/ndt/gfp041] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Long-term peritoneal dialysis (PD) is associated with peritoneal fibrosis and loss of function. It has been shown that activation of the adenosine A(2A) receptor (A(2A)R) promotes tissue repair, wound healing and extracellular matrix (ECM) production. We have previously shown that adenosine is a potent regulator of inflammation in the peritoneum. In the current study, we explored the role of adenosine and the A(2A)R in two experimental models. METHODS Collagen deposition was evaluated in primary peritoneal fibroblasts following treatment with an A(2A)R agonist and antagonist. In addition, peritoneal fibrosis was induced by i.p. injection of either chlorhexidine gluconate for 2 weeks or 4.25% glucose peritoneal dialysis fluid (PDF) for 1 month. The development of fibrosis was compared between wild-type (WT) and WT mice treated with caffeine (an A(2A)R antagonist) in drinking water or between (A(2A)R(+/+)) mice and A(2A)R-deficient mice (A(2A)R(-/-)). RESULTS Adenosine or the A(2A)R agonist CGS21680 stimulated collagen production by peritoneal fibroblasts in vitro and A(2A)R antagonists (ZM241385 and caffeine) blocked this effect. Consistent with these results, caffeine-treated WT or A(2A)R(-/-) mice had reduced submesothelial thickness, collagen deposition and mRNA levels of fibroblast-specific protein (FSP-1) and connective tissue growth factor (CTGF). In addition, treatment with caffeine in vitro and in vivo diminished A(2A)R and A(2B)R mRNA levels induced by CG or PDF while it upregulated A(1)R levels. CONCLUSION Our data suggest that adenosine through its A(2A)R promotes peritoneal fibrosis and therefore should be considered as a target for pharmacological intervention.
Collapse
Affiliation(s)
- Sigal Nakav
- Department of Clinical Biochemistry, Soroka Medical Center and Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | | | | | | | | | | |
Collapse
|
9
|
Ruvinov E, Sharabani-Yosef O, Nagler A, Einbinder T, Feinberg MS, Holbova R, Douvdevani A, Leor J. Transplantation of genetically engineered cardiac fibroblasts producing recombinant human erythropoietin to repair the infarcted myocardium. FIBROGENESIS & TISSUE REPAIR 2008; 1:7. [PMID: 19014419 PMCID: PMC2585557 DOI: 10.1186/1755-1536-1-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2008] [Accepted: 11/03/2008] [Indexed: 11/29/2022]
Abstract
Background Erythropoietin possesses cellular protection properties. The aim of the present study was to test the hypothesis that in situ expression of recombinant human erythropoietin (rhEPO) would improve tissue repair in rat after myocardial infarction (MI). Methods and results RhEPO-producing cardiac fibroblasts were generated ex vivo by transduction with retroviral vector. The anti-apoptotic effect of rhEPO-producing fibroblasts was evaluated by co-culture with rat neonatal cardiomyocytes exposed to H2O2-induced oxidative stress. Annexin V/PI assay and DAPI staining showed that compared with control, rhEPO forced expression markedly attenuated apoptosis and improved survival of cultured cardiomyocytes. To test the effect of rhEPO on the infarcted myocardium, Sprague-Dawley rats were subjected to permanent coronary artery occlusion, and rhEPO-producing fibroblasts, non-transduced fibroblasts, or saline, were injected into the scar tissue seven days after infarction. One month later, immunostaining identified rhEPO expression in the implanted engineered cells but not in controls. Compared with non-transduced fibroblasts or saline injection, implanted rhEPO-producing fibroblasts promoted vascularization in the scar, and prevented cell apoptosis. By two-dimensional echocardiography and postmortem morphometry, transplanted EPO-engineered fibroblasts did not prevent left ventricular (LV) dysfunction and adverse LV remodeling 5 and 9 weeks after MI. Conclusion In situ expression of rhEPO enhances vascularization and reduces cell apoptosis in the infarcted myocardium. However, local EPO therapy is insufficient for functional improvement after MI in rat.
Collapse
Affiliation(s)
- Emil Ruvinov
- Neufeld Cardiac Research Institute, Sheba Medical Center, Tel-Aviv University, Tel-Hashomer, Israel
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Vorobiov M, Malki M, Shnaider A, Basok A, Rogachev B, Lewis EC, Chaimovitz C, Zlotnik M, Douvdevani A. Erythropoietin Prevents Dialysis Fluid-Induced Apoptosis of Mesothelial Cells. Perit Dial Int 2008. [DOI: 10.1177/089686080802800618] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background In peritoneal dialysis (PD)-treated patients, denudation of the mesothelium correlates with peritoneal fibrosis and vascular changes. Since recombinant human erythropoietin (rHuEPO) induces a range of cytoprotective cellular responses, rHuEPO treatment may reduce PD fluid (PDF)-induced damage. Methods To investigate the antiapoptotic effect and mechanism of rHuEPO in peritoneal mesothelial cells (PMCs), isolated mice PMCs were used for in vitro characterization of rHuEPO effects. To confirm the in vitro effects, active caspase-3 was analyzed in imprints of liver visceral peritoneum of mice pretreated overnight with rHuEPO (5000 U/kg intraperitoneally) and exposed to PDF (Dianeal 4.25%; Baxter Healthcare, Deerfield, Illinois, USA) for 4 hours. Results Mouse PMCs expressed EPO-receptor mRNA and protein. Short exposure to rHuEPO (5 U/mL) induced phosphorylation of JAK2, STAT5, and ERK1/2. PMCs pretreated for 1 hour with rHuEPO showed reduced PDF-induced caspase-3 activation (49.6%) and DNA fragmentation (38.4%) in comparison to cells treated by PDF alone ( p < 0.05). rHuEPO treatment induced an increase in ERK1/2 phosphorylation and reduced levels of PDF-induced phospho-P38. PD98059, a specific inhibitor of ERK activation, fully blocked the protective effect of rHuEPO. In mice, rHuEPO reduced the apoptotic effect of PDF, as assessed by the level of active caspase-3. Conclusions Our study presents new insights into clinical use of rHuEPO in the setting of PD. We found that rHuEPO provides ERK1/2-dependent protection to PMCs from PDF-induced apoptosis. The use of rHuEPO, or any of its new derivatives that do not stimulate erythropoiesis, should be considered for peritoneal preservation.
Collapse
Affiliation(s)
- Marina Vorobiov
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Myriam Malki
- Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alla Shnaider
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ana Basok
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Boris Rogachev
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eli C. Lewis
- Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Cidio Chaimovitz
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moshe Zlotnik
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amos Douvdevani
- Departments of Nephrology Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
11
|
Nakav S, Chaimovitz C, Sufaro Y, Lewis EC, Shaked G, Czeiger D, Zlotnik M, Douvdevani A. Anti-inflammatory preconditioning by agonists of adenosine A1 receptor. PLoS One 2008; 3:e2107. [PMID: 18461129 PMCID: PMC2329854 DOI: 10.1371/journal.pone.0002107] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Accepted: 03/23/2008] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Adenosine levels rise during inflammation and modulate inflammatory responses by engaging with four different G protein-coupled receptors. It is suggested that adenosine exhibits pro-inflammatory effects through its A(1) receptor (A(1)R), and anti-inflammatory effects through A(2A) receptor (A(2A)R). Therefore, understanding of the mechanisms that govern adenosine receptor regulation may advance treatment of various inflammatory disorders. We previously reported that peak A(1)R expression during leukocyte recruitment, is followed by a peak in A(2A)R during inflammation resolution. PRINCIPAL FINDINGS Here, we examined whether A(1)R activation sequentially induces A(2A)R expression and by this reverses inflammation. The effect of adenosine on A(1)R mediated A(2A)R expression was examined in peritoneal macrophages (PMPhi) and primary peritoneal mesothelial cells (PMC) in vitro. Induction of A(2A)R was inhibited by pertussis toxin (PTX) and partly dependent on A(2A)R stimulation. Administration of A(1)R agonists to healthy mice reduced A(1)R expression and induced A(2A)R production in PMC. Mice that were preconditioned with A(1)R agonists 24 hours before E. coli inoculation exhibited decreased TNFalpha and IL-6 sera levels and reduced leukocytes recruitment. Preconditioning was blocked by pretreatment with A(1)R antagonist, as well as, or by late treatment with A(2A)R antagonist, and was absent in A(2A)R(-/-) mice. CONCLUSIONS Our data suggest that preconditioning by an A(1)R-agonist promotes the resolution of inflammation by inducing the production of A(2A)R. Future implications may include early treatment during inflammatory disorders or pretreatment before anticipated high risk inflammatory events, such as invasive surgery and organ transplantation.
Collapse
Affiliation(s)
- Sigal Nakav
- Department of Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Cidio Chaimovitz
- Department of Nephrology, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yuval Sufaro
- Department of Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eli C. Lewis
- Department of Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gad Shaked
- Department of General Surgery, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - David Czeiger
- Department of General Surgery, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moshe Zlotnik
- Department of Nephrology, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amos Douvdevani
- Department of Clinical Biochemistry, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Nephrology, Soroka Medical University Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
- * E-mail:
| |
Collapse
|
12
|
Elmadbouh I, Michel JB, Chachques JC. Mesothelial cell transplantation in myocardial infarction. Int J Artif Organs 2007; 30:541-9. [PMID: 17628855 DOI: 10.1177/039139880703000612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Mesothelial cells (MCs) are accessible in human patients by excision and digestion of epiploon or from peritoneal fluid or lavage. MCs are easy to culture to obtain large quantities in vitro and they can be genetically modified with interesting therapeutic genes. The important potential of MCs in tissue engineering has been shown during epiplooplasty to different organs and also in creating artificial blood conduits. MC of epicardium is probably the precursor of coronary arteries during embryogenesis. MCs secrete a broad spectrum of angiogenic cytokines, growth factors and extracellular matrix, which could be useful for repairing damaged tissues. MCs are transitional mesodermal-derived cells and considered as progenitor stem cell, have similar morphological and functional properties with endothelial cells and conserve properties of transdifferentiation. MC therapy in myocardial infarction induced neoangiogenesis in infarcted scar and preserved heart function. In conclusion, a potential therapeutic strategy would be to implant or re-implant genetically modified MCs in post-infarction injury to enhance tissue repair and healing. Imparting therapeutic target genes such as angiogenic genes would also be useful for inducing neovascularization.
Collapse
Affiliation(s)
- I Elmadbouh
- INSERM unit 698, Cardiovascular Remodelling, CHU Xavier Bichat-Claude Bernard, Paris, France
| | | | | |
Collapse
|
13
|
Oh TK, Quan GH, Kim HY, Park F, Kim ST. Correction of anemia in uremic rats by intramuscular injection of lentivirus carrying an erythropoietin gene. Am J Nephrol 2006; 26:326-34. [PMID: 16825758 DOI: 10.1159/000094401] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Accepted: 06/07/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Anemia is an inevitable consequence of chronic renal failure. Gene therapy using lentiviral vector (LV) would be an effective tool to treat anemia associated with renal failure. METHODS A LV carrying the erythropoietin (EPO) cDNA was administered to skeletal muscle of partially nephrectomized rats, which is a model of uremia. The red blood cell production and serum EPO levels were temporally monitored in these rats. Polymerase chain reaction assays were done to validate the presence of the LV in the experimental rats. RESULTS After a single intramuscular injection of LV at a dose of 55 microg p24 Gag antigen (approximately 5 x 10(7) transducing units), blood hematocrit (Hct) levels increased and peaked at 3 weeks (47.8 +/- 4.2%, p < 0.01, n = 8) with the levels being maintained for at least 20 weeks (duration of study; 44.9 +/- 3.3%, p < 0.01, n = 3). The control rats receiving LV expressing lacZ had Hct levels of 36.9 +/- 4.1% (n = 8) at 3 weeks and 33.1 +/- 3.7% (n = 4) at 20 weeks, respectively. The serum EPO levels in the rats injected with the LV expression EPO significantly increased (p < 0.01) to 156.3 +/- 3.0 mU/ml compared to the control rats (63.9 +/- 1.7 mU/ml). Polymerase chain reaction analysis of the isolated genomic DNA from the LV-injected rats showed specific positive detection of the LV in only the skeletal muscle tissue at the site of injection, whereas the other tissues, including the liver, spleen, and kidney, were negative. CONCLUSIONS This study demonstrates that intramuscular injection of LV can produce highly efficient and sustained EPO secretion in uremic rats, and suggests that this approach could be an effective tool to deliver secretable proteins at therapeutic levels in various animal disease models.
Collapse
Affiliation(s)
- Tae Keun Oh
- Department of Internal Medicine, Chungbuk National University College of Medicine, Hungdok-Gu, Cheongju, Korea
| | | | | | | | | |
Collapse
|
14
|
Rogachev B, Ziv NY, Mazar J, Nakav S, Chaimovitz C, Zlotnik M, Douvdevani A. Adenosine is upregulated during peritonitis and is involved in downregulation of inflammation. Kidney Int 2006; 70:675-81. [PMID: 16788688 DOI: 10.1038/sj.ki.5001609] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Loss of function of the peritoneal membrane is associated with peritonitis. Adenosine levels in sites of inflammation were shown to increase and exhibit immunoregulatory effects. Our aim was to elucidate the regulatory role of adenosine during peritonitis and to test the involvement of peritoneal mesothelial cells (PMC) in adenosine regulation. In a mice model of Escherichia coli peritonitis, the adenosine A(2A) receptor (A(2A)R) agonist (CGS21680) prevented leukocyte recruitment and reduced tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6) levels. Peritonitis induced the elevation of adenosine with a peak at 24 h. Analysis of adenosine receptor levels on peritoneum showed that A(1) receptor (A(1)R) protein levels peak at 12 h after inoculation and then return to baseline at 24 h, whereas high affinity A(2A)R protein levels peak at 24 h concomitantly with the peak of adenosine concentration. Low affinity A(2B) receptor (A(2B)R) levels elevated slowly, remaining elevated up to 48 h. In human PMC (HPMC), the early cytokines, IL-1-alpha, and TNF-alpha upregulated the A(2B) and A(2A) receptors. However, interferon-gamma (IFN-gamma) upregulated the A(2B)R and decreased A(2A)R levels. Treatment with the A(2A)R agonist reduced IL-1-dependent IL-6 secretion from HPMC. In conclusion, the kinetics of adenosine receptors suggest that at early stage of peritonitis, the A(1)R dominates, and later its dominance is replaced by the G stimulatory (Gs) protein-coupled A(2A)R that suppresses inflammation. Early proinflammatory cytokines are an inducer of the A(2A)R and this receptor reduces their production and leukocyte recruitment. Future treatment with adenosine agonists should be considered for attenuating the damage to mesothelium during the course of acute peritonitis.
Collapse
Affiliation(s)
- B Rogachev
- Department of Nephrology, Soroka Medical Center and Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | | | | | | | | | | |
Collapse
|
15
|
Eliopoulos N, Gagnon RF, Francois M, Galipeau J. Erythropoietin delivery by genetically engineered bone marrow stromal cells for correction of anemia in mice with chronic renal failure. J Am Soc Nephrol 2006; 17:1576-84. [PMID: 16672321 DOI: 10.1681/asn.2005101035] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The goal of this research was to develop a strategy to couple stem cell and gene therapy for in vivo delivery of erythropoietin (Epo) for treatment of anemia of ESRD. It was shown previously that autologous bone marrow stromal cells (MSCs) can be genetically engineered to secrete pharmacologic amounts of Epo in normal mice. Therefore, whether anemia in mice with mild to moderate chronic renal failure (CRF) can be improved with Epo gene-modified MSCs (Epo+MSCs) within a subcutaneous implant was examined. A cohort of C57BL/6 mice were rendered anemic by right kidney electrocoagulation and left nephrectomy. In these CRF mice, the hematocrit (Hct) dropped from a prenephrectomy baseline of approximately 55% to 40% after induction of renal failure. MSCs from C57BL/6 donor mice were genetically engineered to secrete murine Epo at a rate of 3 to 4 units of Epo/10(6) cells per 24 h, embedded in a collagen-based matrix, and implanted subcutaneously in anemic CRF mice. It was observed that Hct increased after administration of Epo+MSCs, according to cell dose. Implants of 3 million Epo+MSCs per mouse had no effect on Hct, whereas 10 million led to a supraphysiologic effect. The Hct of CRF mice that received 4.5 or 7.5 million Epo+MSCs rose to a peak 54+/-4.0 or 63+/-5.5%, respectively, at 3 wk after implantation and remained above 48 or 54% for >19 wk. Moreover, mice that had CRF and received Epo+MSCs showed significantly greater swimming exercise capacity. In conclusion, these results demonstrate that subcutaneous implantation of Epo-secreting genetically engineered MSCs can correct anemia that occurs in a murine model of CRF.
Collapse
Affiliation(s)
- Nicoletta Eliopoulos
- Lady Davis Institute for Medical Research, McGill University Health Center, and Division of Hematology/Oncology, Jewish General Hospital, 3755 Cote Sainte-Catherine Road, Montreal, Quebec, Canada H3T 1E2
| | | | | | | |
Collapse
|
16
|
Hekking LHP, Zweers MM, Keuning ED, Driesprong BAJ, de Waart DR, Beelen RHJ, van den Born J. Apparent successful mesothelial cell transplantation hampered by peritoneal activation. Kidney Int 2006; 68:2362-7. [PMID: 16221241 DOI: 10.1111/j.1523-1755.2005.00698.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Mesothelial cell transplantation has been suggested to improve mesothelial repair after surgery, recurrent peritonitis and peritoneal dialysis. METHODS In this study we evaluated mesothelial cell transplantation during the resolution phase of experimentally thioglycollate-induced peritonitis in rats. To this end 4 x 10(6) DiO-labeled autologous mesothelial cells were transplanted 1 week after peritonitis induction. Peritoneal inflammation and permeability characteristics were evaluated after another week. RESULTS Mesothelial cell transplantation after peritonitis resulted in incorporation of these cells in the parietal mesothelial lining, leading to an acute transient submesothelial thickening which was not seen in transplanted animals without prior peritonitis induction. Long-term functioning of these repopulated mesothelial cells leaded to peritoneal activation as evidenced by a approximately twofold increase in peritoneal lymphocytes (P < 0.01) and omental mast cell counts (P < 0.05), accompanied by the induction of inflammation markers monocyte chemoattractant protein-1 (MCP-1) (P < 0.01) and hyaluronan (P < 0.01) in the transplanted peritonitis group, but not in rats with peritonitis without mesothelial cell transplantation or in control rats without mesothelial cell transplantation (all four parameters P < 0.01). In addition, trapping of transplanted mesothelial cells in the milky spots of omental tissue and lymphatic stomata of the diaphragm both in control and thioglycollate rats seems to increase microvascular permeability, reflected by apparent increased diffusion rates of small solutes and proteins. CONCLUSION Altogether, our data underscore the importance of controlling peritoneal (patho)physiology and function in mesothelial transplantation protocols.
Collapse
Affiliation(s)
- Liesbeth H P Hekking
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|