Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1538 word(s) 1538 2021-10-31 07:29:17 |
2 Format Change Meta information modification 1538 2021-11-11 03:22:20 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Yang, C.; Wu, B. Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury. Encyclopedia. Available online: https://encyclopedia.pub/entry/15873 (accessed on 18 April 2024).
Yang C, Wu B. Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury. Encyclopedia. Available at: https://encyclopedia.pub/entry/15873. Accessed April 18, 2024.
Yang, Chih-Yu, Bo-Sheng Wu. "Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury" Encyclopedia, https://encyclopedia.pub/entry/15873 (accessed April 18, 2024).
Yang, C., & Wu, B. (2021, November 10). Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury. In Encyclopedia. https://encyclopedia.pub/entry/15873
Yang, Chih-Yu and Bo-Sheng Wu. "Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury." Encyclopedia. Web. 10 November, 2021.
Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury
Edit

Acute kidney injury (AKI) causes a lot of harm to human health but is treated by only supportive therapy in most cases. Recent evidence shows that mesenchymal stem cells (MSCs) benefit kidney regeneration through releasing paracrine factors and extracellular vesicles (EVs) to the recipient kidney cells and are considered to be promising cellular therapy for AKI. To develop more efficient, precise therapies for AKI, we review the therapeutic mechanism of MSCs and MSC-derived EVs in AKI and look for a better understanding of molecular signaling and cellular communication between donor MSCs and recipient kidney cells. We also review recent clinical trials of MSC-EVs in AKI. This review summarizes the molecular mechanisms of MSCs’ therapeutic effects on kidney regeneration, expecting to comprehensively facilitate future clinical application for treating AKI.

acute kidney injury mesenchymal stem cells extracellular vesicles

1. Mesenchymal Stem Cell-Based Therapy in Acute Kidney Injury

1.1. Different Stem Cell Sources in Acute Kidney Injury

Mechanisms involved in self-renewal, differentiation, and repair after kidney injury is still unclear. Evidence showed that stem cell therapies have protective effects on AKI [1]. It was found that the resident renal tubular cells in the injured denudated tissue expressed CD133, an important stem cell marker and a molecule for Wnt/beta-catenin signaling for cell proliferation and tissue repair. Embryonic renal cells express CD133 and maintain uneven expression on the scattered tubular cells in different segments. These resident scattered renal progenitor CD133+ cells act like stem cells, capable of self-renewal and differentiation [2]. Besides, endothelial progenitor cells (EPCs) from bone marrow can also act as a source of regenerative progenitor cells for AKI via the surface protein expression of L-selectin, an adhesion molecule that promotes EPCs migration to injured tissue [3]. Different stem cell sources, such as EPCs [3][4], human liver stem cells (HLSCs) [5], and MSCs [6][7], had demonstrated protective effects in murine AKI models. Among different stem cell sources, MSCs have the most extensive preclinical evidence and can be harvested from variable tissues, e.g., bone marrow, umbilical cord, adipose tissue, etc. Herrera et al. found that exogenous MSCs migrated to the injured kidney through the interaction between MSC surface CD44 and its ligand, hyaluronic acid), which promoted kidney regeneration [8]. Ullah et al. summarized molecular mechanisms of MSC homing steps: (1) tethering by selectins, (2) activating by cytokines, (3) arresting by integrins, (4) transmigrating by matrix remodelers, (5) extravascular migrating by chemokine gradients [9]. Besides, human induced pluripotent stem cells (hiPSCs) and embryonic stem cells were both demonstrated to promote kidney regeneration [10]. Schubert et al. had demonstrated the MSC migration and homing by tracking the adipose-derived MSC (ADSC) with bioluminescence imaging (BLI) versus quantitative reverse transcription-polymerase chain reaction (qRT-PCR) in the cisplatin-induced AKI model [11]. They detected the labeled Luc-specific mRNA in the injured kidney tissue using qRT-PCR; however, they only detected Luc+-ADSCs in the lung but not in the kidney, suggesting that Luc-based qRT-PCR might be a better tool than BLI to track the transplanted MSCs. In 2021, Chen et al. reported a novel therapy combined with plerixafor (AMD3100, as an immunostimulant) and granulocyte-colony stimulating factor (G-CSF). The combinative therapy of AMD3100 and G-CSF could mobilize bone marrow-derived mesenchymal stem cells (BM-MSCs) to the injured tissue and ameliorate kidney function deterioration in the cisplatin-induced AKI model [12].

2. Mesenchymal Stem Cells and MSC-Derived Extracellular Vesicles  Protect from Acute Tubular Injury in Different Models

EV trafficking is multidirectional cell-to-cell communication in damaged tissues and has the effect of facilitating and reprogramming regenerative cells [13]. Additionally, the EVs contained various therapeutic mediators, including cytokines, proteins, and miRNAs [14]. The application of EVs for clinical use relies on efficient EV isolation and characterization [15][16].
Numerous studies in various experimental AKI models demonstrated that MSCs and MSC-derived EVs had the therapeutic activities to reverse AKI via cell-to-cell paracrine communication rather than MSCs per se transdifferentiation. Gatti et al. reported that human MSC-derived EVs could prevent AKI and CKD progression by inhibiting TEC-apoptosis while promoting TEC proliferation in the ischemia-reperfusion (I/R) AKI model [17]. A similar therapeutic effect for AKI prevention had also been observed by using EVs from renal glomerular and tubular progenitor CD133+ cells [18]. Besides, Chen et al. found MVs extracted from Wharton’s jelly MSCs of the umbilical cord could mitigate renal fibrosis, induce cell proliferation, and inhibit cell apoptosis via ERK1/2 signal pathway to initial G2/M cell cycle arrest in the I/R AKI model [19]. MSC-derived MVs administered in the cisplatin-AKI model have been found to upregulate anti-apoptotic genes, including Bcl2, Bcl-xL, and BIRC8. The down-regulation of apoptosis genes, e.g., Casp1, Casp8, and LTA, was also noted [20]. HLSC, with MSC surface markers but without hematopoietic/endothelial markers, and HLSC-derived EVs enhanced renal tubular regeneration in a murine glycerol-induced AKI model [5]. Furthermore, human MSC-conditioned media (MSC-CM) also possess regenerative properties for tissue injury due to MSC-secreted products, such as proteins, lipids, cytokines, or EVs, etc. Overath et al. found that MSC-CM from ADSCs preincubated in a hypoxic environment contains more protective factors and had better therapeutic effects for cisplatin-induced AKI mice than ordinary MSC-CM [21]. These studies show that MSC-derived EVs can be an innovative therapy for AKI.

3. MSCs and MSC-Derived EVs Protect from Acute Glomerular Injury in Different Models

The pathophysiology of AKI is multifactorial and complex. Definite diagnosis of AKI etiologies is sometimes challenging because multiple kidney injuries may coexist. The two major causes of acute tubular damage are ischemic and nephrotoxic [22]. Another important AKI etiology is glomerular damage, including primary rapidly progressive glomerulonephritis (RPGN), or glomerulonephritis secondary to systemic lupus erythematosus, or bacterial endocarditis [23][24]. The epidemiology, clinical phenotypes, and pathophysiology are different between acute tubular and glomerular injuries [25]. We summarized MSC-derived EVs in experimental AKI models of glomerular and tubular damage and listed the potential involved factors and main effects of EV-related kidney repair (Table 1).
Table 1. MSC-derived EVs in experimental models of AKI and the potential mechanisms of EV-induced renal tissue repair.
Histology Authors/Year Reference EV Sources EV Types Experimental Model Species EV Factors Molecular Response Functional Modulation
Acute Tubular Injury Herrera et al., 2007 [8] BM-MSCs NM In vitro/in vivo, glycerol-induced AKI Mouse NM ↑CD44 and hyaluronic acid (major ligand of CD44) interactions ↑exogenous MSC migration and homing
  Gatti et al., 2011 [17] BM-MSCs MVs In vivo, I/R induced acute tubular injury Rat NM NM ↓tubular cell apoptosis,
↑TEC proliferation
  Bruno et al., 2012 [20] BM-MSCs MVs In vitro/in vivo, cisplatin-induced acute tubular injury Mouse Human POLR2E mRNA ↑anti-apoptotic genes, Bcl-xL, Bcl2, and BIRC8,
↓apoptosis genes, Casp1, Casp8, and LTA
↑renal function, morphology, and survival
  Mb et al., 2014 [5] hLSCs NM In vitro/in vivo, intra-muscle glycerol induced AKI Mouse NM ↑PCNA expression ↑tubular cell proliferation,
↑renal function,
↑morphology
  Chen et al., 2017 [19] hWJMSCs MVs In vitro/in vivo, I/R-induced renal fibrosis Rat NM ↑ERK1/2 signaling
↓EMT–related protein, TGF-β1
↑cell cycle-related proteins, CDK 1 and CyclinB1
↑proliferation, ↓apoptosis,
↓collagen deposition,
↑cells in G2/M cell cycle,
↓fibrosis, ↓EMT
  Ranghino et al., 2017 [18] Gl-MSCs
T-CD133+ cells
Gl-MSC-EVs
T-CD133+-EVs
In vivo, I/R induced acute tubular injury Mouse 62 group of miRNAs NM ↑TEC proliferation
  Overath et al., 2016 [21] ADSC-pCM pCM In vitro/in vivo, cisplatin-induced acute tubular injury Mouse 64 expressed proteins ↓inflammatory cytokines, IL-1β, IL-6 ↑ survival
↓ serum Cr and N-GAL
Acute Glomerular Injury Tsuda et al., 2010 [26] FM-MSCs NM In vitro/in vivo, anti-Thy1 nephritis rats NM ↓TNF and MCP-1 through a PGE2-dependent mechanism. ↓Proteinuria
↓mesangial matrix/cell proliferation,
↓glomerular monocyte/macrophage infiltration,
  Zoja et al., 2012 [27] BM-MSCs NM In vitro/in vivo, Adriamycin-induced crescentic nephritis rats NM ↑VEGF expression
↑nephrin and CD2AP
↓monocyte infiltration,
↓podocyte apoptosis,
↓microvascular rarefaction
  Iseri et al., 2016 [28] hMSC-CM CM In vitro/in vivo, anti-glomerular basement membrane nephritis rats NM ↓proinflammatory cytokines TNF-α, IL-1-β, MCP-1, and IL-6 ↑M2 macrophage polarization,
↓proteinuria and crescent formation
Abbreviations: ADSC-pCM, adipose-derived MSC-preconditioned media; AKI, acute kidney injury; BM-MSCs, bone marrow-derived MSCs; CDK, cyclin-dependent protein kinase; EMT, epithelial-mesenchymal transition; EVs, extracellular vesicles; FM-MSCs, fetal membrane-derived MSCs; Gl-MSCs, MSCs derived from human glomeruli; I/R, ischemia/reperfusion; hLSCs, human liver stem cells; hMSC-CM, human MSC-conditioned media; hWJMSCs, human Wharton’s jelly-MSCs; IGF-1R, insulin-like growth factor-1 receptor; MAC, membrane attack complex; MCP-1, monocyte chemoattractant protein 1; MSC, mesenchymal stem cell; MVs, microvesicles; N-GAL, neutrophil gelatinase-associated lipocalin; NM, not mentioned; PCNA, proliferating cell nuclear antigen; T-CD133+ cells, T-CD133+ progenitor cells from human renal tubules tissue.
 
 
 
An experimental glomerulonephritis rat model was created by intravenous infusion of anti-Thy1.1 antibody through complement-mediated mesangial cell damage, impairing glomerular angiogenesis. Tsuda et al. found that allogeneic fetal membrane-derived MSCs (FM-MSCs) decreased urinary protein excretion by inhibiting glomerular monocyte infiltration and mesangial matrix hyperplasia histologically in the rats with anti-Thy1 glomerulonephritis. The mechanism was evident by in vitro experiment showing that FM-MSC conditioned medium contributed to the healing process in injured kidney tissue by inhibiting prostaglandin E2-dependent expression of TNF-α and monocyte chemoattractant protein 1 (MCP-1; a key chemokine for monocyte activation) [26]. In another glomerulonephritis rat model induced by Adriamycin, Zoja et al. found that multiple administrations of BM-MSCs increased VEGF expression and reduced monocyte infiltration, podocyte apoptosis, and microvascular rarefaction and subsequently attenuated glomerular sclerosis [27]. Moreover, Iseri et al. reported that human MSC-CM treatment reduced TNF-α-related proinflammatory cytokine and modulated the glomerular macrophage polarization. The shift to anti-inflammatory M2 subsequently reduced proteinuria and crescent formation in the rat AKI model with anti-glomerular basement membrane RPGN [28]. These studies demonstrated the MSCs significantly decrease proteinuria and regain renal function through various immunomodulatory pathways.

References

  1. Humphreys, B.D.; Bonventre, J.V. Mesenchymal stem cells in acute kidney injury. Annu. Rev. Med. 2008, 59, 311–325.
  2. Brossa, A.; Papadimitriou, E.; Collino, F.; Incarnato, D.; Oliviero, S.; Camussi, G. Role of CD133 molecule in Wnt response and renal repair. Stem Cells Transl. Med. 2018, 7, 283–294.
  3. Biancone, L.; Cantaluppi, V.; Duò, D.; Deregibus, M.C.; Torre, C.; Camussi, G. Role of L-selectin in the vascular homing of peripheral blood-derived endothelial progenitor cells. J. Immunol. 2004, 173, 5268–5274.
  4. Cantaluppi, V.; Medica, D.; Mannari, C. Endothelial progenitor cell-derived extracellular vesicles protect from complement-mediated mesangial inury in experimental anti-Thy 1.1 glomerulonephritis. Nephrol. Dial. Transplant. 2015, 30, 410–422.
  5. Sanchez, M.B.H.; Bruno, S.; Grange, C.; Tapparo, M.; Cantaluppi, V.; Tetta, C. Human liver stem cells and derived extracellular vesicles improve recovery in a murine model of acute kidney injury. Stem Cell Res Ther. 2014, 5, 124.
  6. Cao, Z.; Zhang, G.; Wang, F.; Liu, H.; Liu, L.; Han, Y. Protective effects of mesenchymal stem cells with CXCR4 up-regulation in a rat renal transplantation model. PLoS ONE 2013, 8, e82949.
  7. Bai, M.; Zhang, L.; Fu, B.; Bai, J.; Zhang, Y.; Cai, G. IL-17A improves the efficacy of mesenchymal stem cells in ischemic-reperfusion renal injury by increasing Treg percentages by the COX-2/PGE2 pathway. Kidney Int. 2018, 93, 814–825.
  8. Herrera, M.B.; Bussolati, B.; Bruno, S.; Morando, L.; Mauriello-Romanazzi, G.; Sanavio, F. Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int. 2007, 72, 430–441.
  9. Ullah, M.; Liu, D.D.; Thakor, A.S. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience 2019, 15, 421–438.
  10. Humphreys, B.D. Kidney injury, stem cells and regeneration. Curr. Opin. Nephrol. Hypertens. 2014, 23, 25–31.
  11. Schubert, R.; Sann, J.; Frueh, J.T.; Ullrich, E.; Geiger, H.; Baer, P.C. Tracking of Adipose-Derived Mesenchymal Stromal/Stem Cells in a Model of Cisplatin-Induced Acute Kidney Injury: Comparison of Bioluminescence Imaging versus qRT-PCR. Int. J. Mol. Sci. 2018, 19, 2564.
  12. Chen, Z.; Ren, X.; Ren, R.; Wang, Y.; Shang, J. The combination of G-CSF and AMD3100 mobilizes bone marrow-derived stem cells to protect against cisplatin-induced acute kidney injury in mice. Stem Cell Res. Ther. 2021, 12, 209.
  13. Medica, D.; Dellepiane, S.; Cantaluppi, V. Regenerative Role of Stem Cell-Derived Extracellular Vesicles in Acute Kidney Injury. Nephron 2020, 144, 1–6.
  14. Cha, J.M.; Shin, E.K.; Sung, J.H.; Moon, G.J.; Kim, E.H.; Cho, Y.H. Efficient scalable production of therapeutic microvesicles derived from human mesenchymal stem cells. Sci. Rep. 2018, 8, 1171.
  15. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750.
  16. Doyle, L.M.; Wang, M.Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells 2019, 8, 727.
  17. Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusioninduced acute and chronic kidney injury. Nephrol. Dial. Transplant. 2011, 26, 1474–1483.
  18. Ranghino, A.; Bruno, S.; Bussolati, B. The effects of glomerular and tubular renal progenitors and derived extracellular vesicles on recovery from acute kidney injury. Stem Cell Res. Ther. 2017, 8, 24.
  19. Chen, W.; Yan, Y.; Song, C.; Ding, Y.; Du, T. Microvesicles derived from human Wharton’s Jellymesenchymal stem cells ameliorate ischemia-reperfusion-induced renal fibrosis by releasing from G2/M cell cycle arrest. Biochem. J. 2017, 474, 4207–4218.
  20. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS ONE 2012, 7, e33115.
  21. Overath, J.M.; Gauer, S.; Obermüller, N.; Schubert, R.; Schäfer, R.; Geiger, H.; Baer, P.C. Short-term preconditioning enhances the therapeutic potential of adipose-derived stromal/stem cell-conditioned medium in cisplatin-induced acute kidney injury. Exp. Cell Res. 2016, 342, 175–183.
  22. Basile, D.P.; Anderson, M.D.; Sutton, T.A. Pathophysiology of acute kidney injury. Compr. Physiol. 2012, 2, 1303–1353.
  23. Finn, W.F. Acute Renal Failurea Companion to Brenner and Rector’s The Kidney; John Wiley and Sons Ltd.: Hoboken, NJ, USA, 2001.
  24. Lieberthal, W.; Nigam, S.K. Acute renal failure. I. Relative importance of proximal vs. distal tubular injury. Am. J. Physiol. Ren. Physiol. 1998, 275, F623–F632.
  25. Makris, K.; Spanou, L. Acute Kidney Injury: Definition, Pathophysiology and Clinical Phenotypes. Clin. Biochem. Rev. 2016, 37, 85–98.
  26. Tsuda, H.; Yamahara, K.; Ishikane, S.; Otani, K.; Nakamura, A.; Sawai, K.; Ichimaru, N.; Sada, M.; Taguchi, A.; Hosoda, H.; et al. Allogenic fetal membrane-derived mesenchymal stem cells contribute to renal repair in experimental glomerulonephritis. Am. J. Physiol. Ren. Physiol. 2010, 299, F1004–F1013.
  27. Zoja, C.; Garcia, P.B.; Rota, C.; Conti, S.; Gagliardini, E.; Corna, D.; Zanchi, C.; Bigini, P.; Benigni, A.; Remuzzi, G.; et al. Mesenchymal stem cell therapy promotes renal repair by limiting glomerular podocyte and progenitor cell dysfunction in adriamycin-induced nephropathy. Am. J. Physiol. Ren. Physiol. 2012, 303, F1370–F1381.
  28. Iseri, K.; Iyoda, M.; Ohtaki, H.; Matsumoto, K.; Wada, Y.; Suzuki, T.; Yamamoto, Y.; Saito, T.; Hihara, K.; Tachibana, S.; et al. Therapeutic effects and mechanism of conditioned media from human mesenchymal stem cells on anti-GBM glomerulonephritis in WKY rats. Am. J. Physiol. Ren. Physiol. 2016, 310, F1182–F1191.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 378
Revisions: 2 times (View History)
Update Date: 11 Nov 2021
1000/1000