Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Am Soc Nephrol ; 27(11): 3394-3404, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26984884

ABSTRACT

Erythropoietin (EPO) may be a beneficial tissue-protective cytokine. However, high doses of EPO are associate with adverse effects, including thrombosis, tumor growth, and hypertension. Carbamylated erythropoietin (CEPO) lacks both erythropoietic and vasoconstrictive actions. In this study, we compared the renoprotective, hemodynamic, and hematologic activities and survival effects of identical EPO and CEPO doses in rat models of clinically relevant AKI presentations, including ischemia-reperfusion-induced AKI superimposed on CKD (5000 U/kg EPO or CEPO; three subcutaneous injections) and ischemia-reperfusion-induced AKI in old versus young animals and male versus female animals (1000 U/kg EPO or CEPO; three subcutaneous injections). Compared with EPO therapy, CEPO therapy induced greater improvements in renal function and body weight in AKI on CKD animals, with smaller increases in hematocrit levels and similarly improved survival. Compared with EPO therapy in the other AKI groups, CEPO therapy induced greater improvements in protection and recovery of renal function and survival, with smaller increases in systolic BP and hematocrit levels. Overall, old or male animals had more severe loss in kidney function and higher mortality rates than young or female animals, respectively. Notably, mRNA and protein expression analyses confirmed the renal expression of the heterodimeric EPO receptor/CD131 complex, which is required for the tissue-protective effects of CEPO signaling. In conclusion, CEPO improves renal function, body and kidney weight, and survival in AKI models without raising hematocrit levels and BP as substantially as EPO. Thus, CEPO therapy may be superior to EPO in improving outcomes in common forms of clinical AKI.


Subject(s)
Acute Kidney Injury/drug therapy , Erythropoietin/analogs & derivatives , Erythropoietin/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Age Factors , Animals , Female , Male , Rats , Rats, Inbred F344 , Sex Factors
3.
Am J Kidney Dis ; 60(6): 1012-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23036928

ABSTRACT

Acute kidney injury (AKI) is a common clinical entity with high morbidity and mortality rates and ever increasing medical costs. A large number of patients who are hospitalized with morbidities such as diabetes, vascular disease, or chronic kidney disease are at high risk to develop AKI due to ischemic and nephrotoxic insults. The pathophysiology of ischemic and toxic forms of AKI is complex and includes tubular and vascular cell damage and inflammation. Given the seriousness of this essentially therapy-resistant complication, treatment beyond supportive measures and renal replacement therapy is urgently needed. Recent stem cell research has shown promising results, and cell therapy-based interventions are advancing into clinical trials. An example is our phase 1 clinical trial (NCT00733876) in which cardiac surgery patients at high risk of postoperative AKI were treated safely with allogeneic mesenchymal stem cells. Together with the introduction of biomarkers for an earlier and specific AKI diagnosis, currently tested stem cell-based therapies are expected to provide an entirely new class of diagnostic and therapeutic tools.


Subject(s)
Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Kidney/physiology , Mesenchymal Stem Cell Transplantation/methods , Regeneration/physiology , Acute Kidney Injury/diagnosis , Aged , Clinical Trials, Phase I as Topic/trends , Humans , Male , Renal Dialysis/methods
4.
Kidney Int Suppl (2011) ; 1(3): 87-89, 2011 Sep.
Article in English | MEDLINE | ID: mdl-25018907

ABSTRACT

Both the homing of hematopoietic stem cells (HSCs) to the bone marrow and their engraftment in recipients of bone marrow transplants are primarily mediated by the chemokine stromal-derived factor-1 (SDF-1) or CXCL12, which activates CXCR4, its cognate receptor on HSCs. We showed that the recruitment and temporary attachment of CXCR4-expressing cells, such as HSCs and a fraction of mesenchymal stem cells (MSCs), to the kidney, following ischemia/reperfusion acute kidney injury, are similarly mediated by robustly upregulated SDF-1 in the kidney, indicating that such organ injury appears to lead to the transient expression of a facultative stem cell niche. This SDF-1 response of the injured kidney facilitates both the mobilization from the bone marrow and homing of precursor cells, and other CXCR4-expressing cells such as administered MSCs, to the kidney, where they aid in its protection and repair. Similar responses have been observed subsequent to the injury of other solid organs such as the heart, liver, and brain.

5.
Kidney Int Suppl (2011) ; 1(3): 103-106, 2011 Sep.
Article in English | MEDLINE | ID: mdl-25018910

ABSTRACT

Current therapies for acute kidney injury remain primarily supportive and have failed to reduce morbidity, mortality (>50%), and associated costs. This prompted our studies in which rats with bilateral ischemia/reperfusion-induced acute kidney injury were treated with bone marrow-derived, culture-expanded allogeneic mesenchymal stem cells. Their administration into the suprarenal aorta after reflow significantly protected renal function and hastened repair, mediated by complex antiapoptotic, mitogenic, anti-inflammatory, and immune modulating actions that were not elicited by isogeneic fibroblasts. Infused mesenchymal stem cells, recruited to renal sites of injury, did not significantly differentiate into target cells but rather disappeared from kidneys and other organs within 72 h. Furthermore, at 3 months, compared with vehicle-treated controls, renal function was well preserved and interstitial fibrosis was absent. These preclinical data served as the scientific basis for a recently completed Phase I Clinical Trial (http://www.clinicaltrials.gov; # NCT00733876), in which patients at high risk for cardiac surgery-associated AKI were treated with allogeneic mesenchymal stem cells. Until now, MSC therapy in the study subjects has been safe, and none of the patients has developed postoperative AKI or subsequent loss of renal function, suggesting that this novel form of therapy may have promise in this group of high-risk patients, which will be further investigated in a Phase II Trial.

6.
Nat Rev Nephrol ; 6(3): 179-83, 2010 03.
Article in English | MEDLINE | ID: mdl-20186233

ABSTRACT

Acute kidney injury (AKI) is a common clinical complication, associated with poor outcomes and the development of chronic kidney disease. Despite major advances in the understanding of its pathophysiology, available therapies for AKI are only supportive; therefore, adequate functional recovery from AKI must predominantly rely on the kidney's own reparative ability. An extensive body of preclinical data from our own and from other laboratories has shown that administration of adult multipotent marrow stromal cells (commonly referred to as mesenchymal stem cells [MSCs]), effectively ameliorates experimental AKI by exerting paracrine renoprotective effects and by stimulating tissue repair. Based on these findings, a clinical trial has been conducted to investigate the safety and efficacy of MSCs administered to open-heart surgery patients who are at high risk of postoperative AKI. In this Perspectives article, we discuss some of the early data from this trial and describe potential applications for stem cell therapies in other fields of nephrology.


Subject(s)
Acute Kidney Injury/therapy , Coronary Artery Bypass/adverse effects , Kidney Failure, Chronic/prevention & control , Mesenchymal Stem Cell Transplantation/methods , Acute Kidney Injury/etiology , Acute Kidney Injury/mortality , Animals , Clinical Trials, Phase I as Topic , Coronary Artery Bypass/methods , Coronary Disease/diagnosis , Coronary Disease/therapy , Disease Models, Animal , Female , Follow-Up Studies , Graft Rejection , Graft Survival , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/physiopathology , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Mice , Rats , Risk Assessment , Survival Rate , Transplantation, Autologous , Treatment Outcome
7.
Nephron Exp Nephrol ; 105(1): e33-40, 2007.
Article in English | MEDLINE | ID: mdl-17108708

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) secondary to ischemia continues to be a major clinical problem due to its high morbidity and mortality, and limited treatment options. Animal models are critical to both the study of the pathophysiology of AKI and the development of new interventions. Although histological changes at the glomerulotubular junction have been described in AKI, we examined here whether the extent of glomerular tubularization correlates with the degree of renal insufficiency in this condition. METHODS: Groups of mice with ischemia/reperfusion AKI were utilized in which the severity of renal insufficiency was defined. The resulting level of glomerular tubularization was analyzed, and the involved cell type was identified by immunohistochemistry and electron microscopy. RESULTS: The extent of glomerular tubularization increased significantly with the degree of renal insufficiency. Low level glomerular tubularization was present in normal mouse kidneys, while it was more common and increasingly circumferential in mice with more severe loss of kidney function. The parietal monolayer of cuboidal cells in glomeruli was contiguous with proximal tubular cells, showing a well-developed luminal brush border and positive staining for proliferating cell nuclear antigen and Lotus tetragonolobus, a proximal tubular cell-specific lectin. CONCLUSION: Increased levels of glomerular tubularization represent a poorly understood response to ischemic AKI in mice. As such, this glomerular 'tubularization score' may be useful to complement standard injury scores in experimental and, if detected, in clinical AKI.


Subject(s)
Ischemia/pathology , Kidney Glomerulus/pathology , Kidney/blood supply , Acute Disease , Animals , Male , Mice , Proliferating Cell Nuclear Antigen/analysis
8.
Proc Natl Acad Sci U S A ; 103(15): 5965-70, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16585502

ABSTRACT

Recombinant human erythropoietin (rhEPO) is receiving increasing attention as a potential therapy for prevention of injury and restoration of function in nonhematopoietic tissues. However, the minimum effective dose required to mimic and augment these normal paracrine functions of erythropoietin (EPO) in some organs (e.g., the brain) is higher than for treatment of anemia. Notably, a dose-dependent risk of adverse effects has been associated with rhEPO administration, especially in high-risk groups, including polycythemia-hyperviscosity syndrome, hypertension, and vascular thrombosis. Of note, several clinical trials employing relatively high dosages of rhEPO in oncology patients were recently halted after an increase in mortality and morbidity, primarily because of thrombotic events. We recently identified a heteromeric EPO receptor complex that mediates tissue protection and is distinct from the homodimeric receptor responsible for the support of erythropoiesis. Moreover, we developed receptor-selective ligands that provide tools to assess which receptor isoform mediates which biological consequence of rhEPO therapy. Here, we demonstrate that rhEPO administration in the rat increases systemic blood pressure, reduces regional renal blood flow, and increases platelet counts and procoagulant activities. In contrast, carbamylated rhEPO, a heteromeric receptor-specific ligand that is fully tissue protective, increases renal blood flow, promotes sodium excretion, reduces injury-induced elevation in procoagulant activity, and does not effect platelet production. These preclinical findings suggest that nonerythropoietic tissue-protective ligands, which appear to elicit fewer adverse effects, may be especially useful in clinical settings for tissue protection.


Subject(s)
Blood Coagulation/drug effects , Coagulants/pharmacology , Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Animals , Blood Pressure/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Line , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Erythropoietin/therapeutic use , Hematopoiesis/drug effects , Hemodynamics/drug effects , Humans , Mice , Renal Circulation/drug effects , Renal Circulation/physiology , Stem Cells/drug effects , Stem Cells/physiology , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...