Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Am Soc Nephrol ; 24(11): 1756-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23949798

ABSTRACT

In CKD, the risk of kidney failure and death depends on the severity of proteinuria, which correlates with the extent of podocyte loss and glomerular scarring. We investigated whether proteinuria contributes directly to progressive glomerulosclerosis through the suppression of podocyte regeneration and found that individual components of proteinuria exert distinct effects on renal progenitor survival and differentiation toward a podocyte lineage. In particular, albumin prevented podocyte differentiation from human renal progenitors in vitro by sequestering retinoic acid, thus impairing retinoic acid response element (RARE)-mediated transcription of podocyte-specific genes. In mice with Adriamycin nephropathy, a model of human FSGS, blocking endogenous retinoic acid synthesis increased proteinuria and exacerbated glomerulosclerosis. This effect was related to a reduction in podocyte number, as validated through genetic podocyte labeling in NPHS2.Cre;mT/mG transgenic mice. In RARE-lacZ transgenic mice, albuminuria reduced retinoic acid bioavailability and impaired RARE activation in renal progenitors, inhibiting their differentiation into podocytes. Treatment with retinoic acid restored RARE activity and induced the expression of podocyte markers in renal progenitors, decreasing proteinuria and increasing podocyte number, as demonstrated in serial biopsy specimens. These results suggest that albumin loss through the damaged filtration barrier impairs podocyte regeneration by sequestering retinoic acid and promotes the generation of FSGS lesions. Our findings may explain why reducing proteinuria delays CKD progression and provide a biologic rationale for the clinical use of pharmacologic modulators to induce regression of glomerular diseases.


Subject(s)
Albuminuria/complications , Podocytes/physiology , Regeneration , Tretinoin/metabolism , Albuminuria/pathology , Animals , Cells, Cultured , Female , Glomerulosclerosis, Focal Segmental/etiology , Humans , Mice , Mice, SCID , Response Elements/physiology , Tretinoin/pharmacology
2.
Am J Pathol ; 183(2): 431-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747509

ABSTRACT

Interferon (IFN)-α and IFN-ß are the central regulators of antiviral immunity but little is known about their roles in viral glomerulonephritis (eg, HIV nephropathy). We hypothesized that IFN-α and IFN-ß would trigger local inflammation and podocyte loss. We found that both IFNs consistently activated human and mouse podocytes and parietal epithelial cells to express numerous IFN-stimulated genes. However, only IFN-ß significantly induced podocyte death and increased the permeability of podocyte monolayers. In contrast, only IFN-α caused cell-cycle arrest and inhibited the migration of parietal epithelial cells. Both IFNs suppressed renal progenitor differentiation into mature podocytes. In Adriamycin nephropathy, injections with either IFN-α or IFN-ß aggravated proteinuria, macrophage influx, and glomerulosclerosis. A detailed analysis showed that only IFN-ß induced podocyte mitosis. This did not, however, lead to proliferation, but was associated with podocyte loss via podocyte detachment and/or mitotic podocyte death (mitotic catastrophe). We did not detect TUNEL-positive podocytes. Thus, IFN-α and IFN-ß have both common and differential effects on podocytes and parietal epithelial cells, which together promote glomerulosclerosis by enhancing podocyte loss while suppressing podocyte regeneration from local progenitors.


Subject(s)
Antiviral Agents/pharmacology , Glomerulonephritis/drug therapy , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Doxorubicin/toxicity , Epithelial Cells/drug effects , Female , Glomerulonephritis/physiopathology , HIV Infections/drug therapy , HIV Infections/physiopathology , Humans , Kidney Glomerulus/physiology , Mice , Mice, SCID , Podocytes/drug effects , Regeneration/drug effects
3.
Stem Cells ; 30(8): 1714-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22628275

ABSTRACT

Recent studies implicated the existence in adult human kidney of a population of renal progenitors with the potential to regenerate glomerular as well as tubular epithelial cells and characterized by coexpression of surface markers CD133 and CD24. Here, we demonstrate that CD133+CD24+ renal progenitors can be distinguished in distinct subpopulations from normal human kidneys based on the surface expression of vascular cell adhesion molecule 1, also known as CD106. CD133+CD24+CD106+ cells were localized at the urinary pole of Bowman's capsule, while a distinct population of scattered CD133+CD24+CD106- cells was localized in the proximal tubule as well as in the distal convoluted tubule. CD133+CD24+CD106+ cells exhibited a high proliferative rate and could differentiate toward the podocyte as well as the tubular lineage. By contrast, CD133+CD24+CD106- cells showed a lower proliferative capacity and displayed a committed phenotype toward the tubular lineage. Both CD133+CD24+CD106+ and CD133+CD24+CD106- cells showed higher resistance to injurious agents in comparison to all other differentiated cells of the kidney. Once injected in SCID mice affected by acute tubular injury, both of these populations displayed the capacity to engraft within the kidney, generate novel tubular cells, and improve renal function. These properties were not shared by other tubular cells of the adult kidney. Finally, CD133+CD24+CD106- cells proliferated upon tubular injury, becoming the predominating part of the regenerating epithelium in patients with acute or chronic tubular damage. These data suggest that CD133+CD24+CD106- cells represent tubular-committed progenitors that display resistance to apoptotic stimuli and exert regenerative potential for injured tubular tissue.


Subject(s)
Acute Kidney Injury/pathology , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules, Proximal/cytology , Kidney/cytology , Stem Cells/cytology , Animals , Disease Models, Animal , Female , Humans , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Mice , Mice, SCID , Microscopy, Confocal , Regeneration/physiology , Stem Cells/metabolism , Transplantation, Heterologous
4.
Am J Pathol ; 179(1): 116-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703397

ABSTRACT

Monocyte/ chemoattractant protein-1/chemokine ligand (CCL) 2 and stromal cell-derived factor-1/CXCL12 both contribute to glomerulosclerosis in mice with type 2 diabetes mellitus, through different mechanisms. CCL2 mediates macrophage-related inflammation, whereas CXCL12 contributes to podocyte loss. Therefore, we hypothesized that dual antagonism of these chemokines might have additive protective effects on the progression of diabetic nephropathy. We used chemokine antagonists based on structured l-enantiomeric RNA (so-called Spiegelmers) ie, the CCL2-specific mNOX-E36 and the CXCL12-specific NOX-A12. Male db/db mice, uninephrectomized at the age of 6 weeks, received injections of Spiegelmer, both Spiegelmers, nonfunctional control Spiegelmer, or vehicle from the age of 4 months for 8 weeks. Dual blockade was significantly more effective than monotherapy in preventing glomerulosclerosis. CCL2 blockade reduced glomerular leukocyte counts and renal-inducible nitric oxide synthase or IL-6 mRNA expression. CXCL12 blockade maintained podocyte numbers and renal nephrin and podocin mRNA expression. Consistently, CXCL12 blockade suppressed nephrin mRNA up-regulation in primary cultures of human glomerular progenitors induced to differentiate toward the podocyte lineage. All previously mentioned parameters were significantly improved in the dual-blockade group, which also suppressed proteinuria and was associated with the highest levels of glomerular filtration rate. Blood glucose levels and body weight were identical in all treatment groups. Dual chemokine blockade can have additive effects on the progression of diabetic kidney disease when the respective chemokine targets mediate different pathomechanisms of disease (ie, inflammation and progenitor differentiation toward the podocyte lineage).


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Chemokine CXCL12/antagonists & inhibitors , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/prevention & control , Glomerulonephritis/prevention & control , Animals , Blotting, Western , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CXCL12/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glomerular Filtration Rate , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immunoenzyme Techniques , Interleukin-6/genetics , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Podocytes/metabolism , Podocytes/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
5.
Stem Cells ; 28(9): 1674-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20680961

ABSTRACT

Glomerular diseases account for 90% of end-stage kidney disease. Podocyte loss is a common determining factor for the progression toward glomerulosclerosis. Mature podocytes cannot proliferate, but recent evidence suggests that they can be replaced by renal progenitors localized within the Bowman's capsule. Here, we demonstrate that Notch activation in human renal progenitors stimulates entry into the S-phase of the cell cycle and cell division, whereas its downregulation is required for differentiation toward the podocyte lineage. Indeed, a persistent activation of the Notch pathway induced podocytes to cross the G(2)/M checkpoint, resulting in cytoskeleton disruption and death by mitotic catastrophe. Notch expression was virtually absent in the glomeruli of healthy adult kidneys, while a strong upregulation was observed in renal progenitors and podocytes in patients affected by glomerular disorders. Accordingly, inhibition of the Notch pathway in mouse models of focal segmental glomerulosclerosis ameliorated proteinuria and reduced podocyte loss during the initial phases of glomerular injury, while inducing reduction of progenitor proliferation during the regenerative phases of glomerular injury with worsening of proteinuria and glomerulosclerosis. Taken altogether, these results suggest that the severity of glomerular disorders depends on the Notch-regulated balance between podocyte death and regeneration provided by renal progenitors.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Glomerulosclerosis, Focal Segmental/metabolism , Lupus Nephritis/metabolism , Podocytes/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Case-Control Studies , Cell Cycle , Cell Death , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dipeptides/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Doxorubicin , Female , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/pathology , Humans , Lupus Nephritis/pathology , Mice , Mice, SCID , Podocytes/drug effects , Podocytes/pathology , Proteinuria/metabolism , Proteinuria/pathology , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Severity of Illness Index , Stem Cells/drug effects , Stem Cells/pathology , Time Factors , Transfection
6.
Transpl Int ; 23(9): 914-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20302596

ABSTRACT

During kidney allograft rejection, CXC chemokine ligand 10 (CXCL10)-CXC chemokine receptor 3 (CXCR3) trafficking between peripheral blood and tissues initiates alloresponse and perpetuates a self-inflammatory loop; thus, CXCL10-CXCR3 axis could represent a pharmacologic target. In this perspective, immunosuppressors targeting graft-resident cells, beside immune cells, could be very advantageous. Vitamin D receptor (VDR) agonists exhibit considerable immunomodulatory properties. This study aimed to investigate whether elocalcitol and BXL-01-0029 could decrease the expression of CXCL10 in activated renal tubular cells in vitro and thus be useful in kidney allograft rejection treatment. Experiments were performed in human tubular renal cells stimulated with interferon-gamma + tumor necrosis factor-alpha with and without VDR agonists, tacrolimus, sirolimus, hydrocortisone, methylprednisolone, cyclosporin A and mycophenolate mofetil. CXCL10 protein secretion and gene expression were measured by ELISA and by quantitative PCR. Specific inhibitors were used to investigate intracellular pathways involved in tubular cells activation. For IC(50) determination and comparison, dose-response curves with VDR agonists, tacrolimus and mycophenolic acid were performed. Elocalcitol and BXL-01-0029 inhibited CXCL10 secretion by renal cells, without affecting cell viability, while almost all the immunosuppressors were found to be ineffective, except for tacrolimus and mycophenolate mofetil. BXL-01-0029 was the most potent drug and, notably, it was found to be capable of allowing reduction in tacrolimus-inhibitory doses. Our data suggest that BXL-01-0029 could potentially be a dose-reducing agent for conventional immunosuppressors in kidney rejection management.


Subject(s)
Calcitriol/analogs & derivatives , Chemokine CXCL10/biosynthesis , Gene Expression Regulation/drug effects , Immunosuppressive Agents/pharmacology , Kidney Tubules/metabolism , RNA/genetics , Receptors, Calcitriol/agonists , Adult , Calcitriol/pharmacology , Cells, Cultured , Chemokine CXCL10/drug effects , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , Graft Rejection/metabolism , Graft Rejection/prevention & control , Humans , Kidney Transplantation , Kidney Tubules/cytology , Kidney Tubules/drug effects , Polymerase Chain Reaction , Reference Values
7.
J Am Soc Nephrol ; 20(12): 2593-603, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19875807

ABSTRACT

Glomerular injury can involve excessive proliferation of glomerular epithelial cells, resulting in crescent formation and obliteration of Bowman's space. The origin of these hyperplastic epithelial cells in different glomerular disorders is controversial. Renal progenitors localized to the inner surface of Bowman's capsule can regenerate podocytes, but whether dysregulated proliferation of these progenitors contributes to crescent formation is unknown. In this study, we used confocal microscopy, laser capture microdissection, and real-time quantitative reverse transcriptase-PCR to demonstrate that hypercellular lesions of different podocytopathies and crescentic glomerulonephritis consist of three distinct populations: CD133(+)CD24(+)podocalyxin (PDX)(-)nestin(-) renal progenitors, CD133(+)CD24(+)PDX(+)nestin(+) transitional cells, and CD133(-)CD24(-)PDX(+)nestin(+) differentiated podocytes. In addition, TGF-beta induced CD133(+)CD24(+) progenitors to produce extracellular matrix, and these were the only cells to express the proliferation marker Ki67. Taken together, these results suggest that glomerular hyperplastic lesions derive from the proliferation of renal progenitors at different stages of their differentiation toward mature podocytes, providing an explanation for the pathogenesis of hyperplastic lesions in podocytopathies and crescentic glomerulonephritis.


Subject(s)
Adult Stem Cells/pathology , Glomerulonephritis/pathology , Kidney Glomerulus/pathology , Podocytes/pathology , AC133 Antigen , Adult Stem Cells/classification , Adult Stem Cells/metabolism , Antigens, CD/metabolism , Bowman Capsule/metabolism , Bowman Capsule/pathology , CD24 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Extracellular Matrix/pathology , Glomerulonephritis/classification , Glomerulonephritis/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Glycoproteins/metabolism , Humans , Hyperplasia , Intermediate Filament Proteins/metabolism , Kidney Glomerulus/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Peptides/metabolism , Phenotype , Podocytes/classification , Podocytes/metabolism , Sialoglycoproteins/metabolism
8.
Blood Purif ; 27(3): 261-70, 2009.
Article in English | MEDLINE | ID: mdl-19218794

ABSTRACT

Acute kidney injury (AKI) is characterized by a sudden impairment of kidney function, which results in the retention of urea and other nitrogenous waste products and in the perturbation of extracellular fluid volume as well as electrolyte and acid-base homeostasis. The dysfunction and apoptosis of tubular epithelial cells are of key importance for the pathophysiological consequences of AKI. However, a growing body of evidence supports the contribution of altered renal vascular structure and function in potentially initiating and extending the initial tubular injury. Vascular injury and dysfunction result in alterations of renal oxygenation and hemodynamics that may have long-term effects in regards to renal function, predisposing to chronic kidney disease. There is growing evidence that endothelial progenitor cells (EPCs) may improve vascular regeneration in different ischemic organs, and recent data suggest that EPCs are mobilized after acute renal ischemia and recruited in ischemic kidney areas and can ameliorate AKI through both paracrine effects and repair of injured microvasculature. The loss of endothelial cell function may represent an important therapeutic target, in which EPCs may show potential importance in ameliorating the acute and chronic effects of ischemic AKI.


Subject(s)
Acute Kidney Injury/physiopathology , Endothelial Cells/physiology , Kidney/physiopathology , Stem Cells/physiology , Endothelium, Vascular/physiopathology , Humans , Kidney/blood supply
9.
J Am Soc Nephrol ; 20(2): 322-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19092120

ABSTRACT

Depletion of podocytes, common to glomerular diseases in general, plays a role in the pathogenesis of glomerulosclerosis. Whether podocyte injury in adulthood can be repaired has not been established. Here, we demonstrate that in the adult human kidney, CD133+CD24+ cells consist of a hierarchical population of progenitors that are arranged in a precise sequence within Bowman's capsule and exhibit heterogeneous potential for differentiation and regeneration. Cells localized to the urinary pole that expressed CD133 and CD24, but not podocyte markers (CD133+CD24+PDX- cells), could regenerate both tubular cells and podocytes. In contrast, cells localized between the urinary pole and vascular pole that expressed both progenitor and podocytes markers (CD133+CD24+PDX+) could regenerate only podocytes. Finally, cells localized to the vascular pole did not exhibit progenitor markers, but displayed phenotypic features of differentiated podocytes (CD133-CD24-PDX+ cells). Injection of CD133+CD24+PDX- cells, but not CD133+CD24+PDX+ or CD133-CD24- cells, into mice with adriamycin-induced nephropathy reduced proteinuria and improved chronic glomerular damage, suggesting that CD133+CD24+PDX- cells could potentially treat glomerular disorders characterized by podocyte injury, proteinuria, and progressive glomerulosclerosis.


Subject(s)
Kidney Glomerulus/metabolism , Kidney/cytology , Podocytes/metabolism , Regeneration , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Bowman Capsule/metabolism , CD24 Antigen/biosynthesis , Female , Glomerulosclerosis, Focal Segmental/metabolism , Glycoproteins/biosynthesis , Humans , Kidney/metabolism , Kidney Glomerulus/pathology , Mice , Mice, SCID , Peptides , Podocytes/pathology , Proteinuria/metabolism , Stem Cells
10.
Trends Mol Med ; 14(7): 277-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18554984

ABSTRACT

With the increasing rate of end-stage renal failure and limited alternatives for its treatment, stem cell (SC) therapy for kidney injury is urgently needed. Choosing the right SC type is the critical step in realizing the potential of this therapeutic approach. Four possible sources of SCs are envisioned for the development of this type of treatment: (i) bone-marrow-derived SCs (BMSCs), (ii) renal adult SCs, (iii) embryonic SCs and (iv) fetal renal SCs. We suggest that resident SCs recently identified in the Bowman's capsule of adult human kidneys might prospectively be the ideal cell type for treatment of both acute and chronic renal injury because they display the potential to differentiate into multiple types of renal cells. However, BMSCs also represent an attractive alternative, especially for the treatment of patients affected by acute renal failure.


Subject(s)
Kidney Failure, Chronic/surgery , Kidney/surgery , Stem Cell Transplantation/methods , Stem Cells/cytology , Bone Marrow Cells/cytology , Humans , Kidney/pathology , Kidney Failure, Chronic/pathology , Models, Biological
11.
J Exp Med ; 205(2): 479-90, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18268039

ABSTRACT

Recently, we have identified a population of renal progenitor cells in human kidneys showing regenerative potential for injured renal tissue of SCID mice. We demonstrate here that among all known chemokine receptors, human renal progenitor cells exhibit high expression of both stromal-derived factor-1 (SDF-1) receptors, CXCR4 and CXCR7. In SCID mice with acute renal failure (ARF), SDF-1 was strongly up-regulated in resident cells surrounding necrotic areas. In the same mice, intravenously injected renal stem/progenitor cells engrafted into injured renal tissue decreased the severity of ARF and prevented renal fibrosis. These beneficial effects were abolished by blocking either CXCR4 or CXCR7, which dramatically reduced the number of engrafting renal progenitor cells. However, although SDF-1-induced migration of renal progenitor cells was only abolished by an anti-CXCR4 antibody, transendothelial migration required the activity of both CXCR4 and CXCR7, with CXCR7 being essential for renal progenitor cell adhesion to endothelial cells. Moreover, CXCR7 but not CXCR4 was responsible for the SDF-1-induced renal progenitor cell survival. Collectively, these findings suggest that CXCR4 and CXCR7 play an essential, but differential, role in the therapeutic homing of human renal progenitor cells in ARF, with important implications for the development of stem cell-based therapies.


Subject(s)
Acute Kidney Injury/metabolism , Chemokine CXCL12/metabolism , Kidney/cytology , Multipotent Stem Cells/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Cell Line , Cell Movement , Cells, Cultured , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, SCID , RNA, Messenger/metabolism , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Rhabdomyolysis/complications , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
12.
Int J Biochem Cell Biol ; 40(9): 1764-74, 2008.
Article in English | MEDLINE | ID: mdl-18291705

ABSTRACT

Chemokines binding the CXCR3 receptor have been shown to inhibit angiogenesis via the CXCR3-B isoform, but the underlying molecular mechanisms are unknown. Aim of this study was to elucidate the effects of CXCR3-B on activation of members of the mitogen-activated protein kinase family, and to explore the relevance of defined signaling pathways to the angiostatic effects of CXCR3-B ligands. Human embryonic kidney (HEK) 293 cells were transfected with expression vectors encoding for CXCR3-A or CXCR3-B. In cells expressing CXCR3-A, CXCL10 (IP-10) at nanomolar concentrations induced activation of ERK, Akt, and Src, as previously described in human vascular pericytes. In HEK-293 cells expressing CXCR3-B, exposure to CXCL10 in the micromolar concentration range led to activation of the p38(MAPK) pathway, as indicated by phosphorylation of p38(MAPK) itself, and of MKK3/6 and MAPKAPK-2, that lie upstream and downstream of p38(MAPK), respectively. Similar results were obtained in cells stimulated with CXCL4 (PF4), a specific ligand of CXCR3-B. In contrast, CXCL4 was unable to activate p38(MAPK) in mock-transfected HEK-293 cells. Only a modest induction of ERK or JNK was observed upon CXCR3-B activation. In human microvascular endothelial cells, which selectively express CXCR3-B, in a cell cycle-dependent fashion, CXCL10 and CXCL4 increased the enzymatic activity of p38(MAPK). Pharmacologic inhibition of p38(MAPK) by SB302580 resulted in a significant increase in DNA synthesis and in reversal of the inhibitory action of CXCL10. In conclusion, the p38(MAPK) pathway is a downstream effector of CXCR3-B implicated in the angiostatic action of this chemokine receptor.


Subject(s)
Angiostatic Proteins/metabolism , Receptors, CXCR3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line , Chemokine CXCL10/metabolism , Enzyme Activation , Gene Expression Regulation , Humans , Platelet Factor 4/metabolism , Protein Isoforms/metabolism , Signal Transduction
13.
J Am Soc Nephrol ; 18(12): 3128-38, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17978305

ABSTRACT

Bone marrow-and adult kidney-derived stem/progenitor cells hold promise in the development of therapies for renal failure. Here is reported the identification and characterization of renal multipotent progenitors in human embryonic kidneys that share CD24 and CD133 surface expression with adult renal progenitors and have the capacity for self-renewal and multilineage differentiation. It was found that these CD24+CD133+ cells constitute the early primordial nephron but progressively disappear during nephron development until they become selectively localized to the urinary pole of Bowman's capsule. When isolated and injected into SCID mice with acute renal failure from glycerol-induced rhabdomyolysis, these cells regenerated different portions of the nephron, reduced tissue necrosis and fibrosis, and significantly improved renal function. No tumorigenic potential was observed. It is concluded that CD24+CD133+ cells represent a subset of multipotent embryonic progenitors that persist in human kidneys from early stages of nephrogenesis. The ability of these cells to repair renal damage, together with their apparent lack of tumorigenicity, suggests their potential in the treatment of renal failure.


Subject(s)
Embryo, Mammalian/cytology , Regeneration , Renal Insufficiency/pathology , Stem Cells/cytology , AC133 Antigen , Acute Disease , Animals , Antigens, CD/biosynthesis , CD24 Antigen/biosynthesis , Glycoproteins/biosynthesis , Humans , Kidney Tubules/metabolism , Mice , Mice, SCID , Microscopy, Confocal , Nephrons/pathology , Peptides , Renal Insufficiency/metabolism , Rhabdomyolysis/pathology , Rhabdomyolysis/therapy
14.
Blood ; 109(10): 4127-34, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17218382

ABSTRACT

PF-4/CXCL4 is a member of the CXC chemokine family, which is mainly produced by platelets and known for its pleiotropic biological functions. Recently, the proteic product of a nonallelic variant gene of CXCL4 was isolated from human platelets and named as CXCL4L1. CXCL4L1 shows only 4.3% amino acid divergence in the mature protein, but exhibits a 38% amino acid divergence in the signal peptide region. We hypothesized that this may imply a difference in the cell type in which CXCL4L1 is expressed or a difference in its mode of secretion. In different types of transfected cells, CXCL4 and CXCL4L1 exhibited a distinct subcellular localization and a differential regulation of secretion, CXCL4 being stored in secretory granules and released in response to protein kinase C activation, whereas CXCL4L1 was continuously synthesized and secreted through a constitutive pathway. A protein kinase C-regulated CXCL4 secretion was observed also in lymphocytes, a cell type expressing mainly CXCL4 mRNA, whereas smooth muscle cells, which preferentially expressed CXCL4L1, exhibited a constitutive pathway of secretion. These results demonstrate that CXCL4 and CXCL4L1 exhibit a distinct subcellular localization and are secreted in a differentially regulated manner, suggesting distinct roles in inflammatory or homeostatic processes.


Subject(s)
Platelet Factor 4/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Platelet Factor 4/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , T-Lymphocytes/metabolism , Tissue Distribution , Transfection
15.
J Am Soc Nephrol ; 17(9): 2443-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16885410

ABSTRACT

Regenerative medicine represents a critical clinical goal for patients with ESRD, but the identification of renal adult multipotent progenitor cells has remained elusive. It is demonstrated that in human adult kidneys, a subset of parietal epithelial cells (PEC) in the Bowman's capsule exhibit coexpression of the stem cell markers CD24 and CD133 and of the stem cell-specific transcription factors Oct-4 and BmI-1, in the absence of lineage-specific markers. This CD24+CD133+ PEC population, which could be purified from cultured capsulated glomeruli, revealed self-renewal potential and a high cloning efficiency. Under appropriate culture conditions, individual clones of CD24+CD133+ PEC could be induced to generate mature, functional, tubular cells with phenotypic features of proximal and/or distal tubules, osteogenic cells, adipocytes, and cells that exhibited phenotypic and functional features of neuronal cells. The injection of CD24+CD133+ PEC but not of CD24-CD133- renal cells into SCID mice that had acute renal failure resulted in the regeneration of tubular structures of different portions of the nephron. More important, treatment of acute renal failure with CD24+CD133+ PEC significantly ameliorated the morphologic and functional kidney damage. This study demonstrates the existence and provides the characterization of a population of resident multipotent progenitor cells in adult human glomeruli, potentially opening new avenues for the development of regenerative medicine in patients who have renal diseases.


Subject(s)
Bowman Capsule/cytology , Cell Separation/methods , Multipotent Stem Cells/cytology , AC133 Antigen , Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Adult , Animals , Antigens, CD/biosynthesis , CD24 Antigen/biosynthesis , Clone Cells , Epithelial Cells/cytology , Female , Glycoproteins/biosynthesis , Humans , Mice , Mice, SCID , Nuclear Proteins/biosynthesis , Octamer Transcription Factor-3/biosynthesis , Peptides , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis
16.
J Exp Med ; 197(11): 1537-49, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12782716

ABSTRACT

The chemokines CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC regulate lymphocyte chemotaxis, mediate vascular pericyte proliferation, and act as angiostatic agents, thus inhibiting tumor growth. These multiple activities are apparently mediated by a unique G protein-coupled receptor, termed CXCR3. The chemokine CXCL4/PF4 shares several activities with CXCL9, CXCL10, and CXCL11, including a powerful angiostatic effect, but its specific receptor is still unknown. Here, we describe a distinct, previously unrecognized receptor named CXCR3-B, derived from an alternative splicing of the CXCR3 gene that mediates the angiostatic activity of CXCR3 ligands and also acts as functional receptor for CXCL4. Human microvascular endothelial cell line-1 (HMEC-1), transfected with either the known CXCR3 (renamed CXCR3-A) or CXCR3-B, bound CXCL9, CXCL10, and CXCL11, whereas CXCL4 showed high affinity only for CXCR3-B. Overexpression of CXCR3-A induced an increase of survival, whereas overexpression of CXCR3-B dramatically reduced DNA synthesis and up-regulated apoptotic HMEC-1 death through activation of distinct signal transduction pathways. Remarkably, primary cultures of human microvascular endothelial cells, whose growth is inhibited by CXCL9, CXCL10, CXCL11, and CXCL4, expressed CXCR3-B, but not CXCR3-A. Finally, monoclonal antibodies raised to selectively recognize CXCR3-B reacted with endothelial cells from neoplastic tissues, providing evidence that CXCR3-B is also expressed in vivo and may account for the angiostatic effects of CXC chemokines.


Subject(s)
Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Alternative Splicing , Amino Acid Sequence , Base Sequence , Cell Division , Cell Line , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/metabolism , DNA/genetics , Endothelium, Vascular/cytology , Humans , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/immunology , Platelet Factor 4/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR3 , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL