Introduction

There are lineage-committed and relatively more differentiated cells as well as primitive stem cells in the bone marrow. Bone marrow stem cells are classified into hematopoietic stem cells (HSC) that give rise to all cells of the hematopoietic system and mesenchymal stem cells (MSC) that support hematopoiesis. In addition to their role in hematopoiesis, both HSC and MSC have been shown to have the ability to differentiate into many other types of cells in vivo or in culture conditions. In the last few years, reports of bone marrow cell conversion into various types of renal cells have emerged. MSC have also been shown to provide renal protection by paracrine effects. The diverse developmental potential of bone marrow stem cells as well as the easy access to HSC and expandable nature of MSC in cultures opens the possibility of stem cell-based therapy for kidney diseases. The purpose of this review article is to prove a general overview of bone marrow stem cell biology and discuss strategies in developing bone marrow stem cell-based therapy for the treatment of kidney diseases.

Hematopoietic stem cells and stem cell niche

HSC discovery and isolation

The presence of HSC was first suggested by James Till and Ernest McCulloch in a landmark study in 1961 [1]. In studying radiation sensitivity of mouse bone marrow cells, they discovered that bone marrow contained a unique population of cells that were capable of forming macroscopic cell colonies in the spleen of the transplant recipient mice. The cells were then called “spleen colony-forming units” (CFU-S) and were later found to be able to self-renew and differentiate into all types of blood cells [2]. This discovery led to the definition of the characteristics of all stem cells: self-renewal and lineage differentiation. Because of their pioneering stem cell research, Till and McCulloch received the Lasker Basic Medical Research Award in 2005 [3]. The definitive isolation of mouse HSC was in 1988 by the research team led by Irving Weissman [4]. After immunodepletion of lineage-differentiated cells and staining lineage undifferentiated cells (Lin- cells) with cell-surface markers Sca-1 and c-kit, Lin-Sca-1+c-kit+ cells were isolated by fluorescence-activated cell sorter (FACS) and shown to have stem cell properties [4, 5]. The balance between self-renewal and lineage differentiation of HSC ensures the lifetime supply of all types of blood cells. Based on their ability of self-renewal, HSC can be divided into long-term repopulating HSC (LT-HSC) that can self-renew and reconstitute the hematopoietic system for life, and short-term repopulating HSC (ST-HSC) that have limited or no self-renewal ability and can only sustain hematopoiesis for a few weeks [6]. LT-HSC are considered true stem cells and account for about 1 in 105 of bone marrow cells in mice [7, 8].

The discovery of cell-surface markers provides the advantage in isolation and further study of HSC. For example, CD34 is another commonly used cell-surface marker for isolation of human as well murine HSC, a finding that followed the discovery that a small fraction of human bone marrow cells express CD34 [9]. However, recent research indicates that CD34+ stem cells can only give short-term repopulation of the hematopoietic systems in the recipients, whereas the CD34- population is actually more primitive [6] and can self-renew to give long-term repopulation of blood cells [10]. Alternatively, HSC can be isolated using their physical properties. Based on low staining intensity with DNA dye Hoechst 33342 or mitochondrial membrane dye rhodamine 123, side population (SP) cells, or Rhlo cells with stem cell characteristics have been isolated, respectively [11, 12].

HSC development

The hematopoietic system in vertebrates is mesoderm-derived. During embryogenesis, the first blood cells in mice and men appear in the yolk sac. The primitive nucleated red cells enter the circulation when the embryonic and extraembryonic vessels link together [13]. It was originally thought that adult HSC originated in the yolk sac, migrated to the fetal liver, and subsequently colonized the adult bone marrow. More recent studies have identified the aorta–gonad–mesonephros (AGM) region and more specifically, the dorsal aorta, as the first site for adult HSC formation [14, 15]. The development of stem cells to lineage-differentiated mature cells is a continuum. HSC are a functionally heterogeneous population of cells, even when they are isolated based on their uniformed expression of surface markers. The heterogeneity is reflected by their homing efficiency, the ability to self-renew and to repopulate irradiated hosts, and differences in lineage-biased differentiation [1618]. This heterogeneity has been documented during development and aging. For example, aged HSC produce more myeloid progeny than their younger counterparts. This lineage bias is only partially corrected by exposure to a younger environment [19, 20]. The mechanisms of stem cell heterogeneity are not fully understood at the present time. The model of a single type of “mother of all HSC” that produces diverse daughter cells under the control of a variety of extrinsic and intrinsic factors has been suggested [2123]. Alternatively, the hypothesis of “many mothers” with various clonal diversities has been proposed, and there is increasing evidence favoring this hypothesis [17]. The heterogeneity of HSC may complicate the comparison of experimental results obtained by different research groups with different methods of cell isolation. Similarly, caution should be taken when interpreting microarray analysis data of HSC that are phenotypically homogenous but functionally heterogenous [24, 25].

Stem cell niche

The concept of a stem cell niche was first proposed by Ray Schofield in 1978 to describe a microenvironment in which HSC reside and maintain self-renewal potential. When HSC divides, one daughter cell remains in the niche as the primitive stem cell, whereas the other daughter cell leaves the niche and differentiates into more mature progeny [26]. The niche concept is supported by in vitro studies to demonstrate that bone marrow stromal cells are able of supporting hematopoiesis and maintaining HSC for long-term repopulation as a result of HSC self-renewal [27, 28]. In vivo stem cell niches were defined in Drosophila ovary and testis [2931] and in mammalian skin and gut [3235] before identification of HSC niche in mice in 2003. Zhang and Calvi independently reported the localization of an HSC niche to the endosteal space [36, 37]. Using genetic strategies, these investigators showed that osteoblasts were the important niche cells controlling the number of long-term repopulating HSC. LT-HSC colocalized with spindle-shaped osteoblasts lining the inner bone surface. The number of LT-HSC increased with the increasing number of osteoblasts induced by injection of parathyroid hormone (PTH) or in transgenic mice with conditional inactivation of bone morphogenetic protein (BMP) receptor type IA (BMPRIA). Furthermore, Adams et al. demonstrated that in response to high calcium concentration (as high as 40 mM) in the endosteal niche, HSC that expressed high levels of calcium-sensing receptor (CaR) migrated and engrafted to the endosteal niche [38]. Bone marrow is the preferred homing site for HSC. It is unknown whether chronically elevated levels of PTH observed in humans with renal failure has the same effect on HSC homing and mobilization as do cyclic levels of PTH created by PTH injection. However, the effects of PTH and BMP signaling on HSC provides potential targets for stem-cell-based therapy. More recently, endothelia of bone marrow sinusoidal vessels were found to be the vascular niche for HSC [39, 40].

Stem cell homing and mobilization

One of the most clinically relevant features of HSC is homing and mobilization. HSC homing involves the events of stem cell adhesion and rolling along the bone marrow sinusoidal endothelial cells, transendothelial migration, and lodgement in the bone marrow niches. HSC mobilization from bone marrow into extramedullary tissues is believed to be governed by similar mechanisms as HSC homing [4143]. The interaction of HSC with endothelial cells requires binding of integrins, sialomucins, and CD44 molecules expressed on the HSC with E- and P-selectin and vascular adhesion molecule-1 (VCAM-1) expressed by endothelial cells. Integrin binding, particularly α4β1 integrin or very late antigen-4 (VLA-4) with the counterreceptor VCAM-1, ensures firm HSC adhesion with endothelial cells. The important role of VLA-4 and VCAM-1 in stem cell homing is demonstrated by an early study in which preexposure of blocking antibodies to VLA-4 or VCAM-1 reduced the homing of transplanted murine bone marrow cells to the host bone marrow [44]. Other key players are stromal-derived-factor-1 (SDF-1) and its receptor CXCR4 [42]. SDF-1 belongs to the chemokine family and is expressed by a wide variety of cells, including bone marrow stromal cells and endothelial cells. It consists of cell-bound, extracellular matrix-bound, and soluble forms. SDF-1 binding with CXCR4 expressed on HSC and hematopoietic progenitor cells elicits hematopoietic cell adhesion and transmigration, which is mainly mediated via Gi-protein second messenger. Cooperation and compensation between VAL-4/VCAM-1 and SDF-1/CXCR4 in mediating HSC homing has recently been shown in mice. In that study, a combined blockade of α4 integrin/VCAM-1 and SDF-1/CXCR4 dramatically reduced bone marrow cell homing [45]. Similarly, studies in humans showed that inhibition of CXCR4 by a small molecule AMD3100 reduced homing and enhanced HSC mobilization into the circulation. As a result, a larger number of HSC can be collected for autologous stem cell transplantation. The study provides a good example of clinical usefulness by modulating HSC homing and mobilization [4648].

Discovery and therapeutic values of mesenchymal stem cells

MSC derivation

Mesenchymal stem cells are defined by two features: the ability to undergo sustained proliferation in vitro, and the potential to give rise to cells of mesenchymal lineages, including osteocytes, chondrocytes, and adipocytes [49, 50]. Most of our knowledge of MSC is obtained from cultures of the cells in vitro. Friedenstein et al. first cultured fibroblast precursor cells isolated from mouse bone marrow in 1976. The cells were adherent to a plastic surface and were able to differentiate into osteoblasts, adipocytes, and chondrocytes in vitro [51, 52]. Cells with similar phenotype were identified from cultures of human bone marrow cells [53] and more recently from many other sources, including fat, umbilical cord, scalp, placenta, and exfoliated deciduous teeth in humans and other species [5459]. The cells are generated by a variety of methods in culture medium supplemented with bovine or human serum to obtain plastic-adherent, spindle-shaped, and fibroblast-looking cells. A large number of cells can be achieved after several passages. The culture-derived mesenchymal cells are highly heterogeneous, and some of the cells possess mesenchymal stem cell properties. There are no specific markers to distinguish mesenchymal stem cells from the rest of stromal cells, and virtually all published results are obtained from cultured cells, with different methods of isolation and expansion. At present, MSC are defined by a combination of physical, phenotypical, and functional properties.

Nomenclature and criteria to define MSC

To clarify nomenclature and promote comparison and contrast of study outcomes, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy (ISCT) published a position statement in 2005 suggesting that “the fibroblast-like plastic-adherent cells, regardless of the tissue from which they are isolated, be termed multipotent mesenchymal stromal cells, while the term mesenchymal stem cells is used only for cells that meet the specific stem cell criteria. The widely recognized acronym, MSC, may be used for both mesenchymal stromal cells and mesenchymal stem cells” [60]. Investigators are encouraged to define MSC in specific studies. More recently, another position statement from ISCT provided minimal criteria for defining human multipotent mesenchymal stromal cells as follows: “First, MSC must be plastic-adherent when maintained in standard culture conditions. Second, MSC must express CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface molecules. Third, MSC must differentiate to osteoblasts, adipocytes and chondroblasts in vitro” [61]. These criteria set the standard for cell-surface determinants and phenotype of human MSC.

MSC origin and function

Despite the well-characterized properties of MSC in vitro, the exact origin, physiological function, and development of MSC in vivo have remained elusive. Very recently, using the technique of genetic marking and lineage tracing in mice, Takashima et al. showed the surprising results that during early embryogenesis, the first wave of MSC is derived from neuroepithelium and neural crest rather than mesoderm. However, MSC derived from this pathway are transient and are replaced by MSC from other unidentified sources later in the development. By 4 weeks of age, neuroepithelium- and neural-crest-derived MSC constitute an insignificant portion of MSC isolated from bone tissue. The number of MSC decreases with age [62]. That study demonstrates convincingly that MSC are actually present in vivo rather than an in vitro culture product.

MSC have diverse functions. They provide extracellular matrix, cytokines, and growth factors to support the growth and differentiation of hematopoietic cells in ex vivo culture [49, 51, 52, 6367]. Other well-demonstrated characteristics of MSC include direct differentiation into other cells of mesenchymal origin, such as osteoblasts, adipocytes, chondroblasts, and myoblasts. It has been demonstrated that MSC can differentiate into neurons and astrocytes, cardiomyocytes, and endothelial cells [6872]. Studies conducted by Verfaillie’s group showed in vitro differentiation into many types of cells with mesoderm, neuroectoderm, and endoderm characteristics by multipotent adult progenitor cells (MAPCs) derived from mice and rats. When injected into blastocysts, single MAPC contributed to most, if not all, somatic cell types [73]. The multipotent potential of MAPCs has been found by many scientists around the world to be difficult to replicate [74]. One major difference in deriving MAPCs from most methods of generating MSC is that MAPCs were derived after plating at low density and requiring special culture conditions. Since MAPCs were generated by long-term and low-density culture in vitro, the existence of such cells in vivo has been questioned. The issue of “nature vs. nurture” in derivation of MAPCs has not been settled.

MSC therapeutic values

The ability of in vivo differentiation and their role in immunomodulation suggest that MSC may have clinical implications [75]. Animal and preclinical studies led to clinical trials in testing the safety and efficacy of MSC in treating cardiovascular diseases such as acute myocardial infarction, end-stage ischemic heart disease, and vascular restenosis [7679]; skeletal and muscular diseases such as osteogenesis imperfecta [80, 81]; neurological diseases such as amyotrophic lateral sclerosis [82]; and lysosomal storage diseases such as Hurler syndrome and metachromatic leukodystrophy [83]. Intravenous, intracoronary, or intraspinal cord injection of MSC has proven to be safe, and some structural and/or functional improvement of the diseased organs has been observed with MSC treatment. The therapeutic values of MSC in treating hematological diseases are largely related to their paracrine effect and immunomodulation. MSC can produce cytokines to support hematopoiesis [8486]. Cotransplantation of HSC and MSC enhances bone marrow recovery from chemotherapy or radiation therapy [87]. The immunosuppressive property of MSC has been utilized for potential treatment of graft-versus-host disease (GVHD) [87].

Hematopoietic stem cells in renal repair after acute kidney injury (AKI)

AKI pathogenesis and consequences

The main pathogenesis of AKI resulting from ischemia-reperfusion injury (IRI) includes acute tubular necrosis and apoptosis, glomerular injury, and inflammation [88, 89]. Inflammation and leukocyte infiltration occurs within hours of renal injury and is mediated by adhesion molecules, including integrins, selectins, cytokines, chemokines, and eicosanoid metabolites [9092]. Subsets of leukocytes may exert different effects on the kidney. While neutrophils and macrophages are believed to cause injury to the kidney, the effect of lymphocyte infiltration on the kidney is controversial [91, 93, 94]. Kale et al. showed HSC mobilization into the circulation 24 h postrenal IRI, and transplantation of Lin- bone marrow cells resulted in acceleration of renal functional recovery [95]. In contrast, HSC mobilization with its associated granulocytosis led to worsening of renal structure and function in mice with renal IRI [96], suggesting that deleterious effects by infiltrating granulocytes may override the protective effects of HSC.

Renal injury results in decreased renal plasma flow and glomerular filtration rate (GFR) and triggers tubular regeneration. We and others have shown that both intrarenal cells (surviving mature tubular epithelial cells and renal stem/progenitor cells) and bone marrow stem cells (HSC and MSC) contribute to renal repair [95, 97101]. Following acute renal injury, surviving mature epithelial cells dedifferentiate, proliferate, and redifferentiate to reestablish tubule structure and function. A population of slow-cycling cells located in the renal papilla of rats and mice has been shown to resemble some stem cell properties and may migrate to the site of injury for renal repair [102]. Quantitatively, intrarenal cells were the main source for regenerating cells [98, 103]. AKI carries high morbidity and high mortality [88, 104, 105]. At the present time, the only treatment option for AKI is supportive measures while waiting for renal function to return. The recovery from AKI is incomplete, and long-term consequences of AKI can be severe [106108]. For example, in pediatric patients who survived AKI, 59% had chronic renal injury, and 9% developed end-stage renal disease in 3–5 years [109]. This calls for the development of new and more effective treatment for AKI. The following summarizes our understanding of the role of bone marrow stem cells in renal repair and discusses the strategies in using bone marrow stem cells as a unique cellular source for potential therapy for kidney diseases.

HSC conversion into renal cells

HSC have been shown to differentiate into many types of nonhematopoietic cells in the body or in the culture [110112]. We and others have shown that bone marrow stem cells have the ability to localize to the injured kidney and be converted to various types of renal cells, including tubular epithelial cells, interstitial cells, endothelial cells, mesangial cells, and podocytes [95, 97, 98, 103, 113117]. Using laser scanning confocal microscopy and 3-dimentional image reconstruction, we showed that unfractionated male-derived bone marrow cells could integrate into the proximal tubules, thick ascending limbs, distal tubules, and collecting ducts of the female postischemic kidney. An average of 1.8% tubular epithelial cells were bone-marrow-derived and were negative for the leukocyte marker CD45, indicating the absence of infiltrating white cells in the tubules 28 days postischemic injury [118]. Conversion of bone marrow cells to epithelial cells suggests that bone marrow cells are capable of responding to the signals generated from the microenvironment in the injured kidney and integrating into the tubules for renal repair. However, it remains unclear whether the conversion of bone marrow cells to renal cells is due to differentiation of primitive HSC or HSC progeny that develop in vivo.

In animal models of IRI, most renal repair occurs in the first week postinjury [119], and most regenerating cells originate from intrarenal cells [98, 103]. We showed that with a single injection of freshly isolated and unfractionated bone marrow cells, the earliest time point at which bone-marrow-derived cells could be detected in the tubules was 5 days postrenal IRI. At 7 days, only 1 or 2 tubular cells per kidney section were donor-derived. No improvement of blood urea nitrogen (BUN) was observed, indicating that this modality has not produced physiologically relevant improvement in renal function. In addition, more collagenous interstitial lesions were observed in the kidneys of mice 28 days postinjection with unfractionated bone marrow cells, suggesting that unfractionated bone marrow cells might be involved in interstitial fibrosis at a later stage. To use HSC for potential treatment of AKI, strategies need to be developed to promote early and sustained transmigration of HSC into the kidney, induce the cells to commit to a renal cells fate, and enhance HSC conversion into renal tubular cells within the first 2 weeks following injury, when most renal repair occurs. It is believed that lineage-differentiated hematopoietic cells, such as macrophages, can be involved in interstitial fibrosis following renal ischemic injury. Future studies to select specific population(s) of bone marrow cells may optimize the therapeutic potential while minimizing the adverse effect of the bone marrow hematopoietic cells.

Low frequency of fusion between bone marrow cells and renal cells

Utilization of HSC for cell therapy of AKI requires understanding the mechanisms of cell conversion. Fusion of bone marrow cells with embryonic stem (ES) cells in cultures has been reported. Fused cells changed to ES-like cells with pluripotent potential [120]. Bone marrow cells could also fuse with hepatocytes, Purkinje neurons, intestinal stem cells, cardiomyocytes, or endothelial cells [121127]. To examine whether cell fusion occurs in the postischemic kidneys, we used the strategies of cre/loxP and sex-mismatched systems by injecting bone marrow cells isolated from male R26R-EYFP mice into female creksp mice with renal IRI. After 28 days, cell fusion was detected by chromosome fluorescent in situ hybridization (FISH) for polysomy of the sex chromosomes and by quantitative real-time polymerase chain reaction (qRT-PCR) and immunostaining for enhanced yellow fluorescent protein (EYFP). X and Y chromosome FISH detected four cells containing 3X1Y chromosomes and five cells containing 2X1Y chromosomes among 14,168 total kidney cells examined in the transplanted female mouse kidneys. Five cells with polysomy were detected from a total of 132 bone-marrow-derived tubule cells (3.8%). Cell fusion was verified by the de novo expression of EYFP in the kidney tubules. The frequency of fusion between bone marrow cells and epithelial cells was 0.066% (∼7 per 104 tubular cells) using EYFP detection method. No cell fusion was observed in the contralateral or sham-operated kidneys, suggesting that ischemic injury was required for cell fusion. The requirement of injury for cell fusion was confirmed in cell cultures in vitro. Collectively, these results indicate that bone marrow cells can fuse with renal epithelial cells after ischemic injury, but the low frequency of cell fusion does not account for the majority of the observed conversion of bone marrow cells into kidney cells [118]. In comparison, when wild-type HSC were transplanted into mice with hereditary tyrosinemia, the transplanted cells fused with hepatocytes [122, 123]. The fused cells proliferated extensively and generated millions of functional, highly aneuploid progeny. In these studies, strong selection pressure (mutation of hepatocytes and drug therapy followed by drug withdrawal) favored engraftment of wild-type HSC. In our study, the low number of fused cells in the kidney suggests low rates of proliferation. Although cell fusion may be used as a vehicle for delivering genes or pharmacological agents into target organs, such as the liver, the low frequency of cell fusion in postischemic kidneys may make this approach ineffective in stimulating renal function recovery.

Mesenchymal stem cells in renal repair

MSC differentiation to tubular epithelial cells

The role of MSC in renal protection has been studied. Morigi et al. isolated MSC from mouse bone marrow and transplanted them into the mice with cisplatin-induced acute chemical injury of the kidneys. Improvement in renal structure was observed 4 days post-MSC transplantation, and significant increase in tubular cell proliferation was detected at 4 and 11 days post-MSC treatment, suggesting stimulation of tubular cell growth by factors released by MSC. Renal structure improvement was accompanied by the acceleration of renal function recovery indicated by lower levels of BUN. Kidney analysis at 4 and 29 days post-MSC injection showed MSC differentiation into tubular epithelial cells. In contrast, HSC transplantation led to minimal HSC incorporation into tubules at 4 days post cell injection and no improvement in renal function was detected [101]. This is one of the first studies demonstrating renal protection by MSC differentiation into tubular epithelial cells and possible trophic factor secretion by MSC to enhance renal repair. It is interesting to note that the recipient mice did not have bone marrow ablation with total body irradiation or chemical treatment to prevent rejection. Better MSC engraftment compared with that seen with HSC may be partially due to MSC immune privilege status. MSC differentiation to tubular epithelial cells and the effect of MSC in promoting epithelial proliferation were also reported in mice with glycerol-induced acute renal failure [128], providing further support that MSC might be a direct cellular source for tubular regeneration. However, in both studies, no quantitative assessment of MSC contribution to tubular repair was performed.

Paracrine effect of MSC in renal protection

Togel et al. reported renal protection by MSC in rats with renal IRI. However, renal protection was mediated through proinflammatory cytokine inhibition and anti-inflammatory cytokine stimulation in postischemic kidneys rather than MSC differentiation into renal tubular epithelial cells [100]. In vivo tracking of injected MSC labeled with iron dextran demonstrated that most MSC were localized to the glomerular capillaries, and no tubules showed iron labeling 3 days post-MSC injection [129], supporting the paracrine effect of MSC in renal protection. More recently, Togel et al. showed that MSC produced vascular endothelial growth factor (VEGF), human growth factor (HGF), and insulin-like growth factor (IGF)-1 in cultures. MSC-conditioned medium inhibited apoptosis and promoted growth in cultured endothelial cells. Intra-arterial infusion of MSC into mice with renal IRI decreased apoptosis in the kidneys in the areas there MSC were attached to the microvasculature. These results suggest that paracrine effect on renal vasculature may contribute to renal protection [130] and provide a good example of how exogenous stem cells may interact with intrarenal cells to enhance the potential of endogenous cells for renal repair. At the present time, it is not clear what other factors secreted by MSC can offer renal protection. Identification and purification of these factors would provide new avenues for pharmacological therapy of AKI and avoid injection of a large volume of MSC-conditioned medium.

MSC mobilization into injured kidneys

To understand the mechanism of intrarenal migration of MSC, Herrera et al. examined kidneys injured by intramuscular injection of glycerol and found that the expression of hyaluronic acid (HA) was increased in the kidneys. MSC isolated from mice lacking CD44, the receptor for HA, were unable to localize to the injured kidneys and lacked renal protection. The defect of intrarenal migration of CD44-null MSC was corrected by transfection of the cells with cDNA encoding wild-type CD44. Their results suggest that MSC migration into the injured kidney is mediated by elevated levels of renal HA, which binds to its receptor CD44 expressed on MSC [131]. Because CD44 has multiple functions, including regulation of cell survival, proliferation, and differentiation, future studies by overexpression of CD44 may provide answers as to whether CD44 modulation on MSC would result in an increased renal transmigration and greater enhancement of renal repair.

Summary and future directions

In summary, stem cells residing in bone marrow can be harvested easily and have the potential to repair injured organs, such as the kidney (Fig. 1). Animal studies have demonstrated that both HSC and MSC can migrate into the kidney and differentiate into tubular epithelial cells. Future research to understand the mechanisms of transmigration and conversion of the cells into renal cells will help in the design of new strategies in using bone marrow stem cells as direct cellular sources for renal repair. In addition, the unique features of immunomodulation and trophic effect of MSC can be further explored to decrease inflammation and injury and promote regeneration from intrinsic renal cells (Table 1). Bone marrow stem cells have been used in clinical trials to treat a broad range of diseases, including hematological, neurological, cardiac, and genetic and metabolic diseases. We believe that bone marrow stem cell-based therapy will eventually become a reality in regenerating kidneys and will create significant impact in improving the quality of life in humans with kidney diseases.

Fig. 1
figure 1

Pathogenesis of acute kidney injury and the role of bone marrow stem cells in renal repair. Acute kidney injury is demonstrated by inflammation, detachment of tubular epithelial cells from the basement membrane, and endothelial cell injury (yellow) in renal blood vessels (BV). Renal repair following acute injury is characterized by proliferation of intrarenal cells, including surviving tubular epithelial cells and potential renal stem/progenitor cells to reline the tubular structure. Hematopoietic stem cells (HSC) and mesenchymal stem cells (MSC) can transmigrate into the kidney and integrate into the tubules for conversion to tubular epithelial cells. MSC also decrease inflammation, prevent cell death, and promote proliferation of endothelial and tubular epithelial cells via paracrine mechanisms

Table 1 Renal protective effects of bone marrow stem cells. HSC hematopoietic stem cells, MSC mesenchymal stromal (stem) cells