Next Article in Journal
Functional Roles of FGF Signaling in Early Development of Vertebrate Embryos
Previous Article in Journal
Anti-Inflammatory Effects of C1q/Tumor Necrosis Factor-Related Protein 3 (CTRP3) in Endothelial Cells
Previous Article in Special Issue
Angiogenic CD34 Stem Cell Therapy in Coronary Microvascular Repair—A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Stem Cells to the Rescue: Development and Application of Cell-Based Therapy for Microvascular Repair

1
Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
2
Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
*
Authors to whom correspondence should be addressed.
Cells 2021, 10(8), 2144; https://doi.org/10.3390/cells10082144
Submission received: 17 August 2021 / Accepted: 18 August 2021 / Published: 20 August 2021
The microcirculation includes an invisible network of micro-vessels that are up to a few hundred microns in diameter. The microcirculation in each organ is largely responsible for end organ perfusion and the transfer of oxygen from red blood cells to the cells of the parenchyma to meet their energy requirements. In addition, the microcirculation drives a dynamic and demand-driven exchange of solutes between the intra-vascular and extra-vascular spaces, as well as the delivery of nutrients and blood-born humoral substances to tissues. Moreover, the microcirculation may exert these effects via the regulation of vascular tone.
In many tissues, the structural or functional loss of micro-vessels characterizes disease progression and correlates with ultimate organ failure [1]. Damage to the microcirculation can be caused by ischemia, inflammation, or hypoxia [2], which may induce endothelial activation, with the adhesion and extravasation of leukocytes into tissues. Microvascular integrity is maintained by a careful balance between endothelium-derived vasodilators such as nitric-oxide and vasoconstrictors such as endothelin. Exposure to cardiovascular risk, genetic, and epigenetic factors can cause dysfunction or physical injury to the endothelial layer, such as the loss of its integrity, and therefore the impairment of myriad functions attributed to the microcirculation [3], including insulin sensitivity [4]. In addition, the loss of angiogenic signaling, development of tissue fibrosis, prolonged periods of vasoconstriction, and inflammation can cause microvascular obliteration.
One of the mechanisms by which the microcirculation retains its integrity involves a continuous process of endothelial cell repair and replacement, as well as ongoing stimuli to facilitate and foster its expansion. The endogenous cellular repair system plays an important role in the maintenance of the endothelium, as well as parenchymal cells. This system includes bone marrow-derived blood-borne endothelial progenitor cells that continuously patrol the microcirculation to repair and replace injured endothelial cells and micro-vessels whenever and wherever encountered [5]. Upon injury, signals are released from the injured organ to increase the number of endothelial progenitor cells in the peripheral circulation and the cells home to injury sites [6]. In addition to circulating cells, many tissues contain niches of resident stem cells, such as mesenchymal stem/stromal cells, that respond to local cues and are recruited upon injury to repair tissue.
However, these potent reparative systems can be injured or lost during development of systemic or local tissue disease, which may render them ineffective when a tissue requires reparative activities [7]. This apparent structural and functional loss of the microcirculation, together with the inability of the endogenous repair system to restore it, has generated the impetus to exogenously replenish this system. Endothelial progenitor cells seem to have a particularly robust pro-angiogenic activity [8]. Nevertheless, MSCs show potent anti-inflammatory and immunomodulatory activities, and they can both directly and indirectly restore the microcirculation and improve its function [9]. Therefore, both these interventions have been used and gained popularity for the purpose of microvascular repair.
Being a fundamental pathogenic mechanism underlying tissue damage, microvascular repair has been positioned at the center of a number of therapeutic interventions. This Special Issue of Cells focuses on the development and application of cell-based therapy for microvascular repair. The ubiquitous nature of microvascular damage as a fundamental mechanism of tissue injury is reflected in the variety of organs in which cell-based therapy has been attempted. The disease entities described in this Special Issue include renovascular disease associated with ischemic nephropathy in both humans [10] and pigs [11], lung injury induced with Influenza A virus in mice [12], coronary microvascular repair [13], and ischemic stroke [14,15]. Novel approaches to achieve microvascular repair via the co-transplantation of kidney tissue-forming cells with vessel-forming cells, as well as sophisticated imaging techniques that are capable of illustrating the morphology and function of the renal microcirculation [16], are described [17]. Importantly, a review article also provides some cautionary notes and describes the reality of clinical development and the limitations that it imposes on the field [18].
Clearly, additional development is needed for the field of cell-based and cell-derived therapy, which is rapidly moving forward. For example, the use of stem cell-derived extracellular vesicles may alleviate some safety concerns associated with the delivery of live replicating cells [19], and the delivery of organoids might allow for the replacement of basic functional units of different tissues in forms that might expedite the restoration of organ function [20]. Novel scaffolds are being designed to ensure long-term residence of the stem-cells in tissues and increase their ability to release favorable paracrine mediators and endow healing benefits [21]. The genetic engineering of stem cells [22] or their daughter extracellular vesicles [23] may improve tissue targeting and enable the utilization of these cells and cellular products as vehicles to deliver additional drugs or diagnostic features [24].
Overall, teamwork, scientific collaboration, and knowledge dissemination, as exemplified in the current Special Issue of Cells, will hopefully propel this field forward in the coming years.

Funding

Partly supported by NIH grants numbers DK120292, DK122734, and AG062104.

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

Lilach O. Lerman is an advisor AstraZeneca, Janssen Pharmaceuticals, and Butterfly Biosciences. Amir Lerman serves on the advisory board of Itamar medical and CohBar, Inc. The authors declare no conflict.

References

  1. Jo, Y.S.; Moon, H.; Park, K. Different Microcirculation Response Between Culprit and Non-Culprit Vessels in Patients With Acute Coronary Syndrome. J. Am. Heart Assoc. 2020, 9, e015507. [Google Scholar] [CrossRef]
  2. Sorop, O.; Olver, T.D.; van de Wouw, J.; Heinonen, I.; van Duin, R.W.; Duncker, D.J.; Merkus, D. The microcirculation: A key player in obesity-associated cardiovascular disease. Cardiovasc. Res. 2017, 113, 1035–1045. [Google Scholar] [CrossRef]
  3. Escaned, J.; Lerman, L.O. Coronary microcirculation and hypertensive heart failure. Eur. Heart J. 2020, 41, 2376–2378. [Google Scholar] [CrossRef] [PubMed]
  4. Sara, J.D.; Taher, R.; Kolluri, N.; Vella, A.; Lerman, L.O.; Lerman, A. Coronary microvascular dysfunction is associated with poor glycemic control amongst female diabetics with chest pain and non-obstructive coronary artery disease. Cardiovasc. Diabetol. 2019, 18, 22. [Google Scholar] [CrossRef]
  5. Chade, A.R.; Zhu, X.; Lavi, R.; Krier, J.D.; Pislaru, S.; Simari, R.D.; Napoli, C.; Lerman, A.; Lerman, L.O. Endothelial progenitor cells restore renal function in chronic experimental renovascular disease. Circulation 2009, 119, 547–557. [Google Scholar] [CrossRef]
  6. Chade, A.R.; Zhu, X.Y.; Krier, J.D.; Jordan, K.L.; Textor, S.C.; Grande, J.P.; Lerman, A.; Lerman, L.O. Endothelial Progenitor Cells Homing and Renal Repair in Experimental Renovascular Disease. Stem Cells 2010, 28, 1039–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Gossl, M.; Khosla, S.; Zhang, X.; Higano, N.; Jordan, K.L.; Loeffler, D.; Enriquez-Sarano, M.; Lennon, R.J.; Lerman, L.O.; Lerman, A. Role of circulating osteogenic progenitor cells in calcific aortic stenosis. J. Am. Coll. Cardiol. 2012, 60, 1945–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Zhu, X.Y.; Urbieta-Caceres, V.; Krier, J.D.; Textor, S.C.; Lerman, A.; Lerman, L.O. Mesenchymal stem cells and endothelial progenitor cells decrease renal injury in experimental swine renal artery stenosis through different mechanisms. Stem Cells 2013, 31, 117–125. [Google Scholar] [CrossRef] [Green Version]
  9. Eirin, A.; Zhu, X.Y.; Krier, J.D.; Tang, H.; Jordan, K.L.; Grande, J.P.; Lerman, A.; Textor, S.C.; Lerman, L.O. Adipose tissue-derived mesenchymal stem cells improve revascularization outcomes to restore renal function in swine atherosclerotic renal artery stenosis. Stem Cells 2012, 30, 1030–1041. [Google Scholar] [CrossRef] [Green Version]
  10. Textor, S.C.; Abumoawad, A.; Saad, A.; Ferguson, C.; Dietz, A. Stem Cell Therapy for Microvascular Injury Associated with Ischemic Nephropathy. Cells 2021, 10, 765. [Google Scholar] [CrossRef]
  11. Ferguson, C.M.; Farahani, R.A.; Zhu, X.-Y.; Tang, H.; Jordan, K.L.; Saadiq, I.M.; Lerman, A.; Lerman, L.O.; Eirin, A. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Elicit Better Preservation of the Intra-Renal Microvasculature Than Renal Revascularization in Pigs with Renovascular Disease. Cells 2021, 10, 763. [Google Scholar] [CrossRef] [PubMed]
  12. Bogatcheva, N.V.; Coleman, M.E. Concentrated Secretome of Adipose Stromal Cells Limits Influenza A Virus-Induced Lung Injury in Mice. Cells 2021, 10, 720. [Google Scholar] [CrossRef] [PubMed]
  13. Rai, B.; Shukla, J.; Henry, T.D.; Quesada, O. Angiogenic CD34 Stem Cell Therapy in Coronary Microvascular Repair—A Systematic Review. Cells 2021, 10, 1137. [Google Scholar] [CrossRef] [PubMed]
  14. Davis, C.; Savitz, S.I.; Satani, N. Mesenchymal Stem Cell Derived Extracellular Vesicles for Repairing the Neurovascular Unit after Ischemic Stroke. Cells 2021, 10, 767. [Google Scholar] [CrossRef] [PubMed]
  15. Saft, M.; Gonzales-Portillo, B.; Park, Y.J.; Cozene, B.; Sadanandan, N.; Cho, J.; Garbuzova-Davis, S.; Borlongan, C.V. Stem Cell Repair of the Microvascular Damage in Stroke. Cells 2020, 9, 2075. [Google Scholar] [CrossRef] [PubMed]
  16. Apelt, K.; Bijkerk, R.; Lebrin, F.; Rabelink, T.J. Imaging the Renal Microcirculation in Cell Therapy. Cells 2021, 10, 1087. [Google Scholar] [CrossRef]
  17. Namestnikov, M.; Pleniceanu, O.; Dekel, B. Mixing Cells for Vascularized Kidney Regeneration. Cells 2021, 10, 1119. [Google Scholar] [CrossRef]
  18. Povsic, T.J.; Gersh, B.J. Stem Cells in Cardiovascular Diseases: 30,000-Foot View. Cells 2021, 10, 600. [Google Scholar] [CrossRef]
  19. Zhao, Y.; Zhu, X.; Zhang, L.; Ferguson, C.M.; Song, T.; Jiang, K.; Conley, S.M.; Krier, J.D.; Tang, H.; Saadiq, I.; et al. Mesenchymal Stem/Stromal Cells and their Extracellular Vesicle Progeny Decrease Injury in Poststenotic Swine Kidney Through Different Mechanisms. Stem Cells Dev. 2020, 29, 1190–1200. [Google Scholar] [CrossRef]
  20. Lawlor, K.T.; Vanslambrouck, J.M.; Higgins, J.W.; Chambon, A.; Bishard, K.; Arndt, D.; Er, P.X.; Wilson, S.B.; Howden, S.E.; Tan, K.S.; et al. Cellular extrusion bioprinting improves kidney organoid reproducibility and conformation. Nat. Mater. 2021, 20, 260–271. [Google Scholar] [CrossRef]
  21. Zhou, C.; Zhou, L.; Liu, J.; Xu, L.; Xu, Z.; Chen, Z.; Ge, Y.; Zhao, F.; Wu, R.; Wang, X.; et al. Kidney extracellular matrix hydrogel enhances therapeutic potential of adipose-derived mesenchymal stem cells for renal ischemia reperfusion injury. Acta Biomater. 2020, 115, 250–263. [Google Scholar] [CrossRef] [PubMed]
  22. Serra, J.; Alves, C.P.A.; Brito, L.; Monteiro, G.A.; Cabral, J.M.S.; Prazeres, D.M.F.; da Silva, C.L. Engineering of Human Mesenchymal Stem/Stromal Cells with Vascular Endothelial Growth Factor-Encoding Minicircles for Angiogenic Ex Vivo Gene Therapy. Hum. Gene Ther. 2019, 30, 316–329. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, X.J.; Jiang, K.; Ferguson, C.M.; Tang, H.; Zhu, X.; Lerman, A.; Lerman, L.O. Augmented efficacy of exogenous extracellular vesicles targeted to injured kidneys. Signal Transduct. Target Ther. 2020, 5, 199. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, M.; Jeong, M.; Hur, S.; Cho, Y.; Park, J.; Jung, H.; Seo, Y.; Woo, H.A.; Nam, K.T.; Lee, K.; et al. Engineered ionizable lipid nanoparticles for targeted delivery of RNA therapeutics into different types of cells in the liver. Sci. Adv. 2021, 7, eabf4398. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lerman, L.O.; Lerman, A. Stem Cells to the Rescue: Development and Application of Cell-Based Therapy for Microvascular Repair. Cells 2021, 10, 2144. https://doi.org/10.3390/cells10082144

AMA Style

Lerman LO, Lerman A. Stem Cells to the Rescue: Development and Application of Cell-Based Therapy for Microvascular Repair. Cells. 2021; 10(8):2144. https://doi.org/10.3390/cells10082144

Chicago/Turabian Style

Lerman, Lilach O., and Amir Lerman. 2021. "Stem Cells to the Rescue: Development and Application of Cell-Based Therapy for Microvascular Repair" Cells 10, no. 8: 2144. https://doi.org/10.3390/cells10082144

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop