ArticlePDF AvailableLiterature Review

The Continuing Story of Renal Repair with Stem Cells

Authors:
  • Nakayamadera Imai Clinic
The Continuing Story of Renal
Repair with Stem Cells
Enyu Imai* and Hirotsugu Iwatani
*Department of Nephrology, Osaka University Graduate School of
Medicine, Suita, and
Department of Medicine, Osaka Koseinen-
kin Hospital, Osaka, Japan
J Am Soc Nephrol 18: 2423–2428, 2007.
doi: 10.1681/ASN.2007070769
Renal stem cells are still a hot topic, but their location and
mechanism of action continue to elude. The therapeutic po-
tential of intrinsic stem cells to the recovery of damaged kid-
ney has been suggested by a number of recent experiments,
whereas the role of various other kinds of administered stem
cells is still controversial. There are two large questions to
address regarding the role of stem cells in kidney repair. The
first is whether the restoration of nephrons requires either
intrinsic renal progenitor cells, bone marrow– derived stem
cells, or both. The second pertains to the role of stem cells in
the repair process itself, specifically whether stem cells replace
damaged tubular cells by transdifferentiation or accelerate
repair by an indirect mechanism.
In the early 2000s, pluripotent bone marrow– derived
stem cells were thought to contribute directly to regeneration
of the kidney.
1,2
Bone marrow contains at least three stem cell
lineages: Hematopoietic stem cells (HSC), mesenchymal
stem/stromal cells (BMSC), and endothelial progenitor cells.
Crude preparations of bone marrow– derived stem cells seem
to have a high capacity for transdifferentiation and therefore
are able to replace damaged renal tissue with tubular epithe-
lial cells, mesangial cells, endothelial cells, and even podo-
cytes. In these studies, double staining of green fluorescence
protein or Y chromosomes from bone marrow– derived stem
cells in addition to kidney cell–specific surface markers pro-
vided evidence of transdifferentiation and repair, although
the issue of cell fusion looms large and is still controversial.
In other studies, the injection of BMSC protects the kid-
ney from toxin or ischemia/reperfusion injury and attenuates
lost renal function, whereas injected HSC do not have the
same effect.
3
In both approaches, however, bone marrow–
derived stem cells seemed to contribute relatively small num-
bers of cells (3 to 22%) to regenerating renal tubular
2
and
glomerular cell populations
1
; that is, the majority of repara-
tive cells were derived from intrinsic kidney cells. In studies
from this period, the notion of transdifferentiation was used
with loosely defined meaning. The definition of transdiffer-
entiation in the stem cell field is the complete conversion of a
cell from one lineage to another lineage with clearly altered
tissue-specific markers and functions. However, the com-
pleteness of this definition has not been demonstrated in
many studies.
If recent studies argue against the direct differentiation of
most bone marrow– derived stem cells into kidney cells, then
some other mechanism must contribute to kidney repair.
Duffield et al.,
4
for example, reported that adoptively trans-
ferred BMSC are not detected in the kidney, lung, or spleen,
whereas injection of BMSC ameliorated repair of the kidney
in the ischemia/reperfusion model. Togel et al.
5
also demon-
strated that administration of BMSC significantly improved
renal function in the ischemia/reperfusion model, but none
of the cells differentiated into tubular or endothelial cells.
In this issue of JASN,Biet al.
6
provide another mecha-
nism. They demonstrate that adoptive transfer of BMSC into
the peritoneal cavity or passive transfer of condition medium
from cultured BMSC accelerates recovery from cisplatin-in-
duced acute renal failure in mice. This is new evidence that
humoral factors from BMSC—and not BMSC per se—are
necessary for recovery from acute kidney injury. Further-
more, injected BMSC localize in the vasculature of the lung
but not in the kidney at all. These results suggest that the local
presence of BMSC in the kidney is not necessary for repair
and that the humoral factors secreted by BMSC at remote
locations are sufficient to protect or repair injured nephrons.
Although many of us assume that tissue regeneration after
acute kidney injury may subtly orchestrate the temporal and
spatial expression of various growth factors or cytokines as in
the developmental stage, it is of great interest that systemic
administration of condition medium from BMSC affords
this renoprotective effect as well, presumably activating in-
trinsic stem cells in the kidney through endocrine or para-
crine action. What these intrinsic renal cells are and what in
the conditioned medium are the critical mediators of this
signaling are not yet clear.
The candidate factors are plentiful. We know from other
experiments that the therapeutic effects of angiogenic growth
factors such as vascular endothelial growth factor (VEGF)
7
and hepatocyte growth factor (HGF)
8
have been reported in
tissue injury followed by fibrosis. VEGF and HGF are se-
creted by BMSC, and administration of VEGF or HGF im-
proves hemodynamics, increases capillary density, acceler-
ates tissue repair, and inhibits fibrosis in injured tissues.
BMSC also seem to have immunomodulatory properties and
inhibit alloantigen-induced differentiation, maturation, and
Published online ahead of print. Publication date available at www.jasn.org.
Correspondence: Dr. Enyu Imai, Department of Nephrology, Osaka Univer-
sity Graduate School of Medicine, 2–2 Yamadaoka, Suita 565-0871, Japan.
Phone: 81-6-6879-3632; Fax: 81-6-6879-3639; E-mail imai@medone.med.
osaka-u.ac.jp
Copyright © 2007 by the American Society of Nephrology
UP FRONT MATTERS
EDITORIALS www.jasn.org
J Am Soc Nephrol 18: 2423–2428, 2007 ISSN : 1046-6673/1809-2423 2423
activation of dendritic cells. They also decrease IL-2 produc-
tion and IL-2 receptor expression in activated T cells; induce
regulatory T cells; and suppress the activation, proliferation,
chemotaxis, and antibody production of B cells.
9
These immu-
nosuppressive actions of BMSC are not fully understood; how-
ever, some speculate that secreted, soluble factors suppress in-
flammation and mediate the beneficial actions in tissue repair.
What are the cellular targets of this conditioned medium?
Presumably they are renal stem cells or progenitor cells. Kid-
ney stem cells have been described in the renal papilla,
10,11
Bowman’s capsule,
12
and the S3 segment of the proximal tu-
bule.
13–15
CD133
/CD24
stem cells in or near Bowman’s
capsule can differentiate into epithelial and endothelial cells.
Oct4-, Pax-2–, and CD90-expressing cells in the proximal tu-
bules differentiate into tubular cells.
15
Multipotent renal pro-
genitor cells expressing Pax-2, Sca-1, and Musashi-1 have been
isolated from microdissected S3 segments.
14
Taking advantage
of the slow cycling of stem cells, a population of proximal tu-
bular epithelial cells have also been isolated in important ex-
periments.
13
Further studies, of course are necessary to identify the ben-
eficial effects of soluble factors in the condition medium of
BMSC on kidney repair, and confirmatory experiments are
needed in other models of acute kidney injury and experimen-
tal glomerulonephritis. For the future, more vertical research
on mechanisms of kidney regeneration is needed, and a high
priority should be placed on the identification of the therapeu-
tic factors and target cells in and around the nephron.
DISCLOSURES
None.
REFERENCES
1. Poulsom R, Forbes S, Hodivala-Dike K, Ryan E, Wyles S, Navarat-
narasah S, Jeffery R, Hunt T, Alison M, Cook T, Pusey C, Wright NA:
Bone marrow contributes to renal parenchymal turnover and regen-
eration. J Pathol 195: 229–235, 2001
2. Kale S, Karihaloo A, Clark P, Kashgarian M, Krause D, Cantley L: Bone
marrow stem cells contribute to repair of the ischemically injured renal
tubule. J Clin Invest 112: 42–49, 2003
3. Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, Abbate M, Rottoli
D, Angioletti S, Benigni A, Perico N, Alison M, Remuzzi G: Mesenchy-
mal stem cells are renotropic, helping to repair the kidney and im-
prove function in acute renal failure. J Am Soc Nephrol 15: 1794
1804, 2004
4. Duffield JS, Park KM, Hsiao LL, Kelley VR, Scadden DT, Ichimura T,
Bonventre JV: Restoration of tubular epithelial cells during repair of
the post-ischemic kidney occurs independently of bone marrow-de-
rived stem cells. J Clin Invest 115: 1743–1755, 2005
5. Toegel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C: Adminis-
tered mesenchymal stem cells protect against ischemic acute renal
failure through differentiation-independent mechanisms. Am J Physiol
289: F31–F42, 2005
6. Bi B, Schmitt R, Israilova M, Nishio H, Cantley L: Stromal cells protect
against acute tubular injury via an endocrine effect. J Am Soc Nephrol
18: 2486–2496, 2007
7. Shimizu A, Masuda Y, Mori T, Kitamura H, Ishizaki M, Sugisaki Y,
Fukuda Y: Vascular endothelial growth factor165 resolves glomerular
inflammation and accelerates glomerular capillary repair in rat anti-
glomerular basement membrane glomerulonephritis. J Am Soc Neph-
rol 15: 2655–2665, 2004
8. Matsumoto K, Nakamura T: Hepatocyte growth factor; renotropic role
and potential therapeutics for renal diseases. Kidney Int 59: 2023–
2038, 2001
9. Mctaggart S, Atkinson K: Mesenchymal stem cells: Immunobiology
and therapeutic potential in kidney disease. Nephrology 12: 44–52,
2006
10. Oliver J, Maarouf O, Cheema F, Martens T, Al-Awqati Q: The renal
papilla is a niche for adult kidney stem cells. J Clin Invest 114: 795–
804, 2004
11. Dekel B, Zangi L, Shezen E, Reich-Zeliger S, Eventov-Friedman S,
Katchman H, Jacob-Hirsch J, Amariglio N, Rechavi G, Margalit R,
Reisner Y: Isolation and characterization of nontubular Sca-1Lin
multipotent stem/progenitor cells from adult mouse kidney. JAmSoc
Nephrol 17: 3300–3314, 2006
12. Sagrinati C, Nett G, Mazzinghi B, Lazzeri E, Liotta F, Frosali F, Ronconi
E, Meini C, Gacci M, Squecco R, Carini M, Gesualdo L, Francini F,
Maggi E, Annunziato F, Lasagni L, Serio M, Romagnani S, Romagnani
P: Isolation and characterization of multipotent progenitor cells from
the Bowman’s capsule of adult human kidneys. J Am Soc Nephrol 17:
2443–2456, 2006
13. Maeshima A, Yamashita S, Nojima Y: Identification of renal progenitor-
like tubular cells that participate in the regeneration processes of the
kidney. J Am Soc Nephrol 14: 3138–3146, 2003
14. Kitamura S, Yamasaki Y, Kinomura M, Sugaya T, Sugiyama H, Mae-
shima Y, Makino H: Establishment and characterization of renal pro-
genitor like cells from S3 segment of nephron in rat adult kidney.
FASEB J 19: 1789–1797, 2005
15. Gupta S, Verfaillie C, Chmielewski D, Kren S, Eidman K, Connaire J,
Heremans Y, Lund T, Blackstad M, Jiang Y, Luttun A, Rosenberg ME:
Isolation and characterization of kidney-derived stem cells. J Am Soc
Nephrol 17: 3028–3040, 2006
See the related article, “Stromal Cells Protect against Acute Tubular Injury via an
Endocrine Effect,” on pages 2486–2496.
Anti–Endothelial Cell
Antibodies in Vasculitis
Caroline O.S. Savage and Julie M. Williams
Division of Immunity and Infection, Medical School, University of
Birmingham, Birmingham, United Kingdom
J Am Soc Nephrol 18: 2425–2426, 2007.
doi: 10.1681/ASN.2007070767
Anti– endothelial cell antibodies have been described in asso-
ciation with small vessel systemic vasculitides since the late
1980s. Opinions have waxed and waned about their impor-
tance. An early study from this group suggested they were
present in 59% of 168 samples from patients with Wegener’s
granulomatosis or microscopic polyangiitis,
1
while a contem-
poraneous study by Varagunam using a similar patient cohort
EDITORIALS www.jasn.org
2424 Journal of the American Society of Nephrology J Am Soc Nephrol 18: , 2007
... Mesenchymal stem cell (MSC) research is a key topic in current stem cell research. Multiple previous studies have demonstrated that MSCs have therapeutic effects on acute kidney injury (AKI), immunoglobulin A nephropathy and other renal diseases (1)(2)(3). AKI is recognized as a common disease which is expensive to manage, prolongs hospitalization and is associated with increased mortality. The mortality rate for AKI has been reported at 24% in hospital patients, and increases with the severity of AKI (4). ...
... Clinically, novel and more effective strategies for the treatment of AKI are expected. MSCs have been applied and provided satisfactory therapeutic effects for AKI in animals and humans (2,3,5); there are multiple mechanisms involved in the therapeutic actions of MSC in AKI treatment (1). ...
Article
Full-text available
Stem cells are used with increasing success in the treatment of renal tubular injury. However, whether mesenchymal stem cells (MSC) differentiate into renal tubular epithelial cells remains controversial. The aims of the present study were to observe the localization of human embryonic MSCs (hMSCs) in the kidneys of newborn mice, and to investigate hMSC differentiation into tubular epithelium. Primary culture hMSCs were derived from 4-7-week-old embryos and labeled with the cell membrane fluorescent dye PKH-26. The degree of apoptosis, cell growth, differentiation and localization of hMSCs with and without this label were then determined using immunohistochemical methods and fl ow cytometry. hMSCs and PKH26-labeled hMSCs were revealed to differentiate into chondrocytes and adipocytes, and were demonstrated to have similar proliferative capability. In the two cell types, the antigens CD34 and CD45, indicative of hematopoietic lineages, were not expressed; however, the expression of the mesenchymal markers CD29 and CD90 in MSCs, was significantly increased. During a 4-week culture period, laser confocal microscopy revealed that PKH26-labeled hMSCs in the kidneys of newborn mice gradually dispersed. Two weeks after the injection of the PKH26-labeled cells, the percentage of PKH26-labeled hMSCs localized to the renal tubules was 10±2.1%. In conclusion, PKH26 labeling has no effect on hMSC differentiation, proliferation and mesenchymal cell surface features, and hMSCs injected into the kidneys of newborn mice may transform to renal tubule epithelium.
Article
Full-text available
The liver in an animal adult healthy body maintains a balance between cell renew and cell loss. The cell damage can stimulate the new cell growth and in liver it is hepatocyte self-replication. The transplanted hepatocyte can undergo clonal expansion which shows that the hepatocytes themselves can play the function as stem cells in the liver. Severe liver injury can activate a potential stem cell compartment located within the intrahepatic biliary tree, giving rise to cords of bipotential within the lobules that can differentiate into hepatocytes and biliary epithelial cells. A third population of stem cells with hepatic potential resides in the bone marrow; these haematopoietic stem cells can contribute to regeneration and restore normal liver function. [Ma H, Young M, Yang Y. Hepatic Stem Cell. Stem Cell. 2015;6(1):5-12] (ISSN 1545-4570). http://www.sciencepub.net/stem. 2
Chapter
Today, most patients who require renal replacement therapy are conventionally treated using dialysis membranes. New developments have been made in the last decade to improve the outcomes for chronic dialysis patients. Cellulose membranes have been replaced by synthetic polymeric membranes with improved biocompatibility, and the development of high-flux membranes and more efficient treatment modes such as hemodiafiltration (HDF) has resulted in improved removal rates for uremic toxins.
Article
Purpose: This study examined the effect of mesenchymal stem cells' conditioned media on the severity of acute kidney injury. Materials and methods: Acute kidney injury was induced in male rats with 100 mg/kg of gentamicin for six consecutive days intraperitoneally. After inducing the standard model of acute kidney injury, the conditioned medium of 5 × 106 cells was calculated for each kilogram of body weight of the rats. Then, it was injected in three different injection patterns other than the baseline injection of gentamicin. The rats were randomly divided into four groups: control group (n = 18) that did not receive any treatment, gentamicin group (n = 18) that received gentamicin at a dosage of 100 mg/kg for six consecutive days intraperitoneally, sham group (n = 54) that received gentamicin for six consecutive days, and an experimental group (n = 54) that received gentamicin for six consecutive days. Serum biochemical analysis and histological changes were studied and analyzed in all groups. Results: Although human mesenchymal stem cells' conditioned media did not improve serum and tissue markers in the treatment groups, a relative improvement was observed in some indicators of tissue damage. Conclusion: Secretory factors of human mesenchymal stem cells can be partly protective against gentamicin-induced nephrotoxicity.&nbsp.
Article
The definition of stem cell is "an unspecialized cell that gives rise to a specific specialized cell, such as a blood cell". Stem Cell is the original of life. All cells come from stem cells. Serving as a repair system for the living body, the stem cells can divide without limit to replenish other cells as long as the living body is still alive. When a stem cell divides, each new cell has the potential to either remain a stem cell situation or become another type of cell with a more specialized function, such as a muscle cell, a red blood cell, a bone cell, a nerve cell, or a brain cell. Stem cell research is a typical and important topic of life science. This material collects some literatures on stem cell and granulocyte colony-stimulating factor (GCSF) and stem cell.
Article
Today, most patients who require renal-replacement therapy (RRT) are conventionally treated using dialysis membranes. To improve the outcome of chronic dialysis patients, unique developments have been made in the past decades. Cellulose membranes have been replaced by synthetic polymeric membranes with improved biocompatibility. The development of high-flux membranes and more efficient treatment modes, for example, hemodiafiltration (HDF), have resulted in improved removal rates of uremic toxins.In 2007, approximately 160 million dialyzers (with an average surface area of 1.7m²) were manufactured for the treatment of 1.6 million dialysis patients worldwide. This large membrane-surface area requires highly automated manufacturing plants to produce high-quality and safe products.Apart from membranes used for conventional hemodialysis (HD), new membranes with increased pore size have been developed for specific treatments. These specialized membranes allow removal of higher-molecular-weight molecules, such as mediators of sepsis/inflammation, or the removal of nephro-toxic light chains of immunoglobulins derived from multiple myelomas (MMs) (bone marrow cancer).Membranes have also been utilized in cell-based extracorporeal systems to treat patients with kidney disease. In addition, specialized, highly efficient membranes have been developed for stem-cell growth in hollow fiber bioreactors as a technological prerequisite for cellular therapies and other applications.
Chapter
The kidney is one of the few organs that undergo mesenchymaleepithelial transition during their development. Structures present in the adult kidney arise from reciprocal interactions between two discrete embryonic appendages: the ureteric bud and metanephric mesenchyme. The adult kidney contains more than 24 mature cell types arranged in distinct vascular, interstitial, glomerular, and tubular compartments. This unique organogenesis and structural complexity of the adult kidney is the cause of many challenges to the identification and characterization of kidney stem cells. Remission of disease and regression of renal lesions are widely observed in experimental animals and even in humans. The repair of injured renal tissue in mammals is not sustained by the generation of new nephrons and frequently leads to a non-functioning mass of fibrotic tissue. A large body of evidence has recently shown that the parietal epithelium of Bowman's capsule represents a reservoir of renal progenitors in adult kidney deputed to replacement and regeneration of podocytes, as well as of proximal tubular cells. This finding provides a new point of view for the understanding of renal physiology and pathophysiology. The first main outcome of the discovery of renal stem cells is that they encourage regeneration and promote functional repair of glomerular injury, and even prevention and treatment of glomerulosclerosis may be possible. The discovery that renal progenitors of Bowman's capsule can not only regenerate podocytes but also cause hyperplastic glomerular lesions in crescentic glomerulonephritis, collapsing glomerulopathy, FSGS or the tip lesion, suggests that glomerular disorders may at least in part result from abnormal or inefficient regenerative responses to podocyte injury.
Article
Chronic glomerulonephritis is a major cause of morbidity and mortality in feline chronic kidney diseases. Mesenchymal stem cells are multipotent stem cells found in the stroma of bone marrow, adipose tissue and umbilical cord. Mesenchymal stem cells can be induced to differentiate into a wide array of cells types including osteoblasts, chondrocytes, myocytes, adipocytes, endothelium and neuronal cells. The effects of mesenchymal stem cell therapy have been investigated in rodent chronic renal failure models including genetic disease, glomerulo-nephritis and experimentally-induced chronic kidney diseases. The purpose of this study was to assess the feasibility of intra-renal artery mesenchymal stem cells transfer in cat with chronic glomerulonephritis. Researchers, hypothesized that mesenchymal stem cells could be safely administered to cats with chronic glomerulo-nephritis. Intra-renal artery mesenchymal stem cells injection would improve the function of the injected kidney. This clinical result showed the optimal direction that the patient demonstrated blood urea nitrogen and creatinine improvement after intra-renal artery mesenchymal stem cells transfer.
Article
Full-text available
Kidney is a highly complex organ which maintains blood pressure, pH and red blood cell count by producing key hormones. Ischemia injury is a common cause of acute renal failure which affects up to 5% of the hospitalized patients. This results in the loss of kidneys function and the patient needs dialysis or a kidney transplant. The renal transplant is a successful treatment but the main disadvantage is the serious shortage of donor organs. Therefore, the use of stem cells for the treatment of kidney diseases represents a critical clinical target. There are four types of stem cells which are used for kidney regeneration: human embryonic stem cells (hESCs), renal adult stem
Article
Full-text available
Kidney is a highly complex organ which maintains blood pressure, pH and red blood cell count by producing key hormones. Ischemia injury is a common cause of acute renal failure which affects up to 5% of the hospitalized patients. This results in the loss of kidneys function and the patient needs dialysis or a kidney transplant. The renal transplant is a successful treatment but the main disadvantage is the serious shortage of donor organs. Therefore, the use of stem cells for the treatment of kidney diseases represents a critical clinical target. There are four types of stem cells which are used for kidney regeneration: human embryonic stem cells (hESCs), renal adult stem cells, non renal adult stem cells and bone marrow derived stem cells (BMSC). The hESCs are derived from the early mammalian embryo and have the ability of undifferentiating and can differentiate to kidney cells but it has two major problems, the ethical problem and the immune rejection. Therefore, we use adult stem cells for the regeneration of human kidney. This review summarizes the current literature on the physiological role of some of the stem cells responsible for renal regeneration.
Article
Full-text available
The paradigm for recovery of the renal tubule from acute tubular necrosis is that surviving cells from the areas bordering the injury must migrate into the regions of tubular denudation and proliferate to re-establish the normal tubular epithelium. However, therapies aimed at stimulating these events have failed to alter the course of acute renal failure in human trials. In the present study, we demonstrate that Lin-Sca-1+ cells from the adult mouse bone marrow are mobilized into the circulation by transient renal ischemia and home specifically to injured regions of the renal tubule. There they differentiate into renal tubular epithelial cells and appear to constitute the majority of the cells present in the previously necrotic tubules. Loss of stem cells following bone marrow ablation results in a greater rise in blood urea nitrogen after renal ischemia, while stem cell infusion after bone marrow ablation reverses this effect. Thus, therapies aimed at enhancing the mobilization, propagation, and/or delivery of bone marrow stem cells to the kidney hold potential as entirely new approaches for the treatment of acute tubular necrosis.
Article
Full-text available
Many adult organs contain stem cells, which are pluripotent and are involved in organ maintenance and repair after injury. In situ, these cells often have a low cycling rate and locate in specialized regions (niches). To detect such cells in the kidney, we administered a pulse of the nucleotide bromodeoxyuridine (BrdU) to rat and mouse pups and, after a long (more than 2-month) chase, examined whether the kidney contained a population of low-cycling cells. We found that in the adult kidney, BrdU-retaining cells were very sparse except in the renal papilla, where they were numerous. During the repair phase of transient renal ischemia, these cells entered the cell cycle and the BrdU signal quickly disappeared from the papilla, despite the absence of apoptosis in this part of the kidney. In vitro isolation of renal papillary cells showed them to have a plastic phenotype that could be modulated by oxygen tension and that when injected into the renal cortex, they incorporated into the renal parenchyma. In addition, like other stem cells, papillary cells spontaneously formed spheres. Single-cell clones of these cells coexpressed mesenchymal and epithelial proteins and gave rise to myofibroblasts, cells expressing neuronal markers, and cells of uncharacterized phenotype. These data indicate that the renal papilla is a niche for adult kidney stem cells.
Article
Full-text available
Kidney is thought to be a regenerative organ in terms of repair from acute tubular injury. It is unknown whether cell population contributes to repair disordered kidney. We attempted to identify and isolate highly proliferative cells from a single cell. We dissected a single nephron from adult rat kidney. Isolated nephrons were separated into segments and cultured. Outgrowing cells were replated after limiting dilution so that each well contained a single cell. One of cell line which was the most potent to grow was designated as rKS56. rKS56 cells showed cobblestone appearance and expressed immature cell markers relating to kidney development and mature tubular cell markers. rKS56 cells grew exponentially and could be maintained for 300 days without transformation. In different culture conditions, rKS56 cells differentiated into mature tubular cells defined by aquaporin-1, 2 expression, and responsiveness to parathyroid hormone or vasopressin. Engrafted to kidney in rat ischemic reperfusion model, rKS56 cells replaced in injured tubules in part after implantation and improved renal function. These results suggest rKS56 cells possess character such as self-renewal, multi-plasticity and capability of tissue repair. rKS56 may possibly contribute to the future development of cell therapy for renal regeneration.
Article
Hepatocyte growth factor (HGF), a ligand for the c-Met receptor tyrosine kinase, has mitogenic, motogenic, anti-apoptotic, and morphogenic (for example, induction of branching tubulogenesis) activities for renal tubular cells, while it has angiogenic and angioprotective actions for endothelial cells. Stromal cells such as mesangial cells, endothelial cells, and macrophages are sources of renal HGF; thus, HGF mediates epithelial-stromal and endothelial-mesangial interactions in the kidney. In response to acute renal injury, the expression of HGF increases in the injured kidney and in distant intact organs such as the lung and spleen. Locally and systemically increased HGF supports renal regeneration, possibly not only by enhancing cell growth but also by promoting morphogenesis of renal tissue. During progression of chronic renal failure/renal fibrosis, the expression of HGF decreases in a manner reciprocal to the increase in expression of transforming growth factor-beta (TGF-beta), a key player in tissue fibrosis. A decrease in endogenous HGF, as well as increase in TGF-beta, augments susceptibility to the onset of chronic renal failure/renal fibrosis. On the other hand, supplements of exogenous HGF have preventive and therapeutic effects in cases of acute and chronic renal failure/renal fibrosis in laboratory animals. HGF prevents epithelial cell death and enhances regeneration and remodeling of renal tissue with injury or fibrosis. A renotropic system underlies the vital potential of the kidney to regenerate, while an impaired renotropic system may confer susceptibility to the onset of renal diseases. Thus, HGF supplementation may be one therapeutic strategy to treat subjects with renal diseases, as it enhances the intrinsic ability of the kidney to regenerate.
Article
In order to establish whether extra-renal cells contribute to the turnover and repair of renal tissues, this study examined kidneys of female mice that had received a male bone marrow transplant and kidney biopsies from male patients who had received kidney transplants from female donors. By using in situ hybridization to detect Y-chromosomes it could be demonstrated that circulating stem cells frequently engraft into the kidney and differentiate into renal parenchymal cells. In the human renal grafts it was confirmed that some of the recipient-derived cells within the kidney exhibited a tubular epithelial phenotype, by combining in situ hybridization with immunostaining for the epithelial markers CAM 5.2 and the lectin Ulex europaeus. Female mouse recipients of male bone marrow grafts showed co-localization of Y-chromosomes and tubular epithelial markers Ricinus communis and Lens culinaris, and a specific cytochrome P450 enzyme (CYP1A2) indicating an appropriate functional capability of clustered newly formed marrow-derived tubular epithelial cells. Y-chromosome-containing cells were observed within glomeruli, with morphology and location appropriate for podocytes. Within the murine kidney, these Y-chromosome-positive cells were negative for the mouse macrophage marker F4/80 antigen and leukocyte common antigen, but were vimentin-positive. The presence of bone marrow-derived cells was noted in both histologically normal mouse kidneys and in human transplanted kidneys suffering damage from a variety of causes. These data indicate that bone marrow cells contribute to both normal turnover of renal epithelia and regeneration after damage, and it is suggested that this could be exploited therapeutically.
Article
The present study was conducted to explore renal progenitor-like cells that are actively engaged in tubular regeneration after injury. For addressing this issue, the existence of label-retaining cells (LRC; slow-cycling cells) in normal rat kidneys by in vivo bromodeoxyuridine (BrdU) labeling was examined. LRC were scattering among renal epithelial tubular cells of normal rat kidneys. During the recovery after renal ischemia, LRC underwent cell division and most of them became positive for proliferating cell nuclear antigen. In contrast, proliferating cell nuclear antigen-positive but BrdU-negative tubular cells were rarely observed, suggesting that cells proliferating during tubular regeneration are essentially derived from LRC. At an early phase of tubular regeneration, descendants of LRC expressed a mesenchymal marker, vimentin, and eventually became positive for an epithelial marker, E-cadherin, after multiple cell divisions. These findings suggested that LRC function as a source of regenerating cells to replace injured cells. Collectively, it was concluded that LRC are renal progenitor-like tubular cells that provide regenerating cells, which actively proliferate and eventually differentiate into epithelial cell, during tubular regeneration. It may be possible to regenerate renal tubules in vivo through the activation of LRC.
Article
Injury to a target organ can be sensed by bone marrow stem cells that migrate to the site of damage, undergo differentiation, and promote structural and functional repair. This remarkable stem cell capacity prompted an investigation of the potential of mesenchymal and hematopoietic stem cells to cure acute renal failure. The model of renal injury induced in mice by the anticancer agent cisplatin was chosen. Injection of mesenchymal stem cells of male bone marrow origin remarkably protected cisplatin-treated syngeneic female mice from renal function impairment and severe tubular injury. Y chromosome-containing cells localized in the context of the tubular epithelial lining and displayed binding sites for Lens culinaris lectin, indicating that mesenchymal stem cells engraft the damaged kidney and differentiate into tubular epithelial cells, thereby restoring renal structure and function. Mesenchymal stem cells markedly accelerated tubular proliferation in response to cisplatin-induced damage, as revealed by higher numbers of Ki-67-positive cells within the tubuli with respect to cisplatin-treated mice that were given saline. Hematopoietic stem cells failed to exert beneficial effects. These results offer a strong case for exploring the possibility that mesenchymal stem cells by virtue of their renotropic property and tubular regenerative potential may have a role in the treatment of acute renal failure in humans.
Article
Vascular endothelial growth factor (VEGF) is essential for maintenance of the glomerular capillary network. The present study investigated the effects of VEGF in rats with progressive crescentic glomerulonephritis (GN). Necrotizing and crescentic GN was induced in rats by injection of anti-rat glomerular basement membrane (GBM) antibody. The alterations of glomerular capillaries and glomerular VEGF expression were assessed. In addition, the effects of continuous VEGF165 administration (10 microg/100 g per d) on glomerular capillaries, glomerular inflammation, and the course of crescentic GN were examined. The appropriate timing of VEGF administration in progressive GN also was evaluated. In anti-GBM GN, necrotizing and crescentic glomerular lesions occurred by day 7, and newly formed necrotizing lesions reoccurred by week 3. Expression of VEGF was markedly reduced in necrotizing and crescentic lesions. Capillary repair was impaired after capillary destruction in necrotizing and crescentic glomeruli, which rapidly progressed to sclerotic glomeruli with chronic renal failure. In contrast, in the rats that received VEGF165 administration from day 7, the necrotizing and crescentic lesions recovered and renal function significantly improved in week 4. This was evident by proliferating endothelial cells and glomerular capillary repair. In addition, VEGF administration decreased intercellular adhesion molecule-1 and monocyte chemoattractant protein-1 expression in glomeruli (particularly on endothelial cells), reduced glomerular infiltrating CD8-postive and ED-1-positive cells, and inhibited the newly formed necrotizing lesions. VEGF administration was apparently effective against both the inflammatory and necrotizing glomerular lesions. These results suggest that VEGF administration resolves glomerular inflammation and accelerates glomerular recovery in the progressive necrotizing and crescentic GN. The therapeutic application of VEGF may be clinically useful for severe GN accompanied by extensive glomerular inflammation and endothelial injury.
Article
Severe acute renal failure (ARF) remains a common, largely treatment-resistant clinical problem with disturbingly high mortality rates. Therefore, we tested whether administration of multipotent mesenchymal stem cells (MSC) to anesthetized rats with ischemia-reperfusion-induced ARF (40-min bilateral renal pedicle clamping) could improve the outcome through amelioration of inflammatory, vascular, and apoptotic/necrotic manifestations of ischemic kidney injury. Accordingly, intracarotid administration of MSC (approximately 10(6)/animal) either immediately or 24 h after renal ischemia resulted in significantly improved renal function, higher proliferative and lower apoptotic indexes, as well as lower renal injury and unchanged leukocyte infiltration scores. Such renoprotection was not obtained with syngeneic fibroblasts. Using in vivo two-photon laser confocal microscopy, fluorescence-labeled MSC were detected early after injection in glomeruli, and low numbers attached at microvasculature sites. However, within 3 days of administration, none of the administered MSC had differentiated into a tubular or endothelial cell phenotype. At 24 h after injury, expression of proinflammatory cytokines IL-1beta, TNF-alpha, IFN-gamma, and inducible nitric oxide synthase was significantly reduced and that of anti-inflammatory IL-10 and bFGF, TGF-alpha, and Bcl-2 was highly upregulated in treated kidneys. We conclude that the early, highly significant renoprotection obtained with MSC is of considerable therapeutic promise for the cell-based management of clinical ARF. The beneficial effects of MSC are primarily mediated via complex paracrine actions and not by their differentiation into target cells, which, as such, appears to be a more protracted response that may become important in late-stage organ repair.
Article
Ischemia causes kidney tubular cell damage and abnormal renal function. The kidney is capable of morphological restoration of tubules and recovery of function. Recently, it has been suggested that cells repopulating the ischemically injured tubule derive from bone marrow stem cells. We studied kidney repair in chimeric mice expressing GFP or bacterial beta-gal or harboring the male Y chromosome exclusively in bone marrow-derived cells. In GFP chimeras, some interstitial cells but not tubular cells expressed GFP after ischemic injury. More than 99% of those GFP interstitial cells were leukocytes. In female mice with male bone marrow, occasional tubular cells (0.06%) appeared to be positive for the Y chromosome, but deconvolution microscopy revealed these to be artifactual. In beta-gal chimeras, some tubular cells also appeared to express beta-gal as assessed by X-gal staining, but following suppression of endogenous (mammalian) beta-gal, no tubular cells could be found that stained with X-gal after ischemic injury. Whereas there was an absence of bone marrow-derived tubular cells, many tubular cells expressed proliferating cell nuclear antigen, which is reflective of a high proliferative rate of endogenous surviving tubular cells. Upon i.v. injection of bone marrow mesenchymal stromal cells, postischemic functional renal impairment was reduced, but there was no evidence of differentiation of these cells into tubular cells of the kidney. Thus, our data indicate that bone marrow-derived cells do not make a significant contribution to the restoration of epithelial integrity after an ischemic insult. It is likely that intrinsic tubular cell proliferation accounts for functionally significant replenishment of the tubular epithelium after ischemia.