Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Stem Cell Res Ther ; 15(1): 23, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38281991

RESUMEN

Over the past decade, there had been progress in the development of cell therapy for insulin-dependent diabetes. Nevertheless, important hurdles that need to be overcome still remain. Protocols for the differentiation of pluripotent stem cells into pancreatic progenitors or fully differentiated ß-cells have been developed. The resulting insulin-producing cells can control chemically induced diabetes in rodents and were the subject of several clinical trials. However, these cells are immunogenic and possibly teratogenic for their transplantation, and an immunoisolation device and/or immunosuppression is needed. A growing number of studies have utilized genetic manipulations to produce immune evasive cells. Evidence must be provided that in addition to the expected benefit, gene manipulations should not lead to any unforeseen complications. Mesenchymal stem/stromal cells (MSCs) can provide a viable alternative. MSCs are widely available from many tissues. They can form insulin-producing cells by directed differentiation. Experimentally, evidence has shown that the transplantation of allogenic insulin-producing cells derived from MSCs is associated with a muted allogeneic response that does not interfere with their functionality. This can be explained by the immunomodulatory functions of the MSC subpopulation that did not differentiate into insulin-producing cells. Recently, exosomes derived from naive MSCs have been used in the experimental domain to treat diabetes in rodents with varying degrees of success. Several mechanisms for their beneficial functions were proposed including a reduction in insulin resistance, the promotion of autophagy, and an increase in the T regulatory population. However, euglycemia was not achieved in any of these experiments. We suggest that exosomes derived from ß-cells or insulin-producing cells (educated) can provide a better therapeutic effect than those derived from undifferentiated cells.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Trasplante de Células Madre Mesenquimatosas , Humanos , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 1/metabolismo , Estudios Prospectivos , Trasplante de Células Madre Mesenquimatosas/métodos , Diferenciación Celular , Insulina/metabolismo
2.
Drugs Context ; 122023.
Artículo en Inglés | MEDLINE | ID: mdl-37077768

RESUMEN

Background: Secondary hyperparathyroidism is a common consequence of end-stage renal disease. Despite the efficacy of kidney transplantation in treating renal failure, many transplant recipients still suffer from persistent or tertiary hyperparathyroidism. Furthermore, the impact of secondary hyperparathyroidism therapy choices on other renal transplant outcomes is poorly understood. Methods: We retrieved the clinical data of 334 patients who received a kidney allograft between January 2007 and December 2014 at the Sheffield Teaching Hospitals, NHS Foundation Trust, United Kingdom. We identified three groups: parathyroidectomy group (34 patients), including patients who had parathyroidectomy before transplantation; cinacalcet group (31 patients), including patients who received cinacalcet before transplantation; and control group (269 patients), including patients who receive a transplant in the same period but did not have any evidence of hyperparathyroidism. We reviewed the demographic data, biochemical parameters and graft survival of all groups. Results: Patients who underwent parathyroidectomy before transplantation had significantly better post-transplant calcium and parathyroid hormone levels than patients in the cinacalcet group (p=0.003). In addition, a significantly lower number of patients had tertiary hyperparathyroidism in the parathyroidectomy group than in the cinacalcet group at 1 year of follow-up (p=0.001). However, short-term and long-term graft survival was comparable in all groups. Conclusions: Renal allograft survival was comparable in all groups. However, tertiary hyperparathyroidism was less likely to occur in patients who underwent parathyroidectomy than in those who were administered cinacalcet.

3.
Stem Cell Res Ther ; 13(1): 350, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35883190

RESUMEN

BACKGROUND: The purpose of this study was to investigate allogenic immune responses following the transplantation of insulin-producing cells (IPCs) differentiated from human adipose tissue-derived stem cells (hAT-MSCs) into humanized mice. METHODS: hAT-MSCs were isolated from liposuction aspirates obtained from HLA-A2-negative healthy donors. These cells were expanded and differentiated into IPCs. HLA-A2-positive humanized mice (NOG-EXL) were divided into 4 groups: diabetic mice transplanted with IPCs, diabetic but nontransplanted mice, nondiabetic mice transplanted with IPCs and normal untreated mice. Three million differentiated cells were transplanted under the renal capsule. Animals were followed-up to determine their weight, glucose levels (2-h postprandial), and human and mouse insulin levels. The mice were euthanized 6-8 weeks posttransplant. The kidneys were explanted for immunohistochemical studies. Blood, spleen and bone marrow samples were obtained to determine the proportion of immune cell subsets (CD4+, CD8+, CD16+, CD19+ and CD69+), and the expression levels of HLA-ABC and HLA-DR. RESULTS: Following STZ induction, blood glucose levels increased sharply and were then normalized within 2 weeks after cell transplantation. In these animals, human insulin levels were measurable while mouse insulin levels were negligible throughout the observation period. Immunostaining of cell-bearing kidneys revealed sparse CD45+ cells. Immunolabeling and flow cytometry of blood, bone marrow and splenic samples obtained from the 3 groups of animals did not reveal a significant difference in the proportions of immune cell subsets or in the expression levels of HLA-ABC and HLA-DR. CONCLUSION: Transplantation of IPCs derived from allogenic hAT-MSCs into humanized mice was followed by a muted allogenic immune response that did not interfere with the functionality of the engrafted cells. Our findings suggest that such allogenic cells could offer an opportunity for cell therapy for insulin-dependent diabetes without immunosuppression, encapsulation or gene manipulations.


Asunto(s)
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Antígeno HLA-A2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre/metabolismo
4.
Exp Clin Transplant ; 20(2): 136-142, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35282810

RESUMEN

OBJECTIVES: Death with graft function is one of the most catastrophic events after kidney transplant. Various pre and posttransplant risk factors have been linked to death with graft function. Characterization of this event is crucial to set successful preventive measures. Here, we reported on death with graft function among living donor kidney transplant recipients seen at the Urology and Nephrology Centre at Mansoura University (Mansoura, Egypt) throughout a period of >4 decades. MATERIALS AND METHODS: This single-center study included 2953 patients who received living donor kidney transplant between March 1976 and December 2018. Patient data were retrospectively analyzed. Patients who had death with graft function were compared with other patients with regard to pre- and posttransplant data. Causes of death with graft function were also studied. RESULTS: Among our patients (1654 male [56%] and 1299 female [44%] patients), death with graft function was reported in 9.9% of patients and responsible for 58.3% of deaths and 24.6% of graft losses. Male sex, pretransplant dialysis and blood transfusion, pre- and posttransplant diabetes and hypertension, high HLA mismatches, antithymocyte globulin induction, steroid and cyclosporine use, steroid dose, acute rejection episodes, and posttransplant infections and malignancy were significantly higher among the death with graft function group. However, multivariate analyses showed that only pretransplant diabetes, steroid dose, and posttransplant infections were risk factors for death with graft function. The most common causes of death with graft function were cardiovascular disease, infections, and malignancy. CONCLUSIONS: Death with graft function remains a significant hindrance to competent kidney transplant outcomes. We found that the most common contributors to this major event were cardiovascular disease, infections, and malignancy. More attention is needed to modify risk factors of cardiovascular disease, to update implementation policies for posttransplant vaccinations, and to conduct increased malignancy surveillance, as well to adopt less aggressive immunosuppression regimens.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus , Enfermedades Cardiovasculares/complicaciones , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/etiología , Femenino , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Humanos , Inmunosupresores/efectos adversos , Riñón , Donadores Vivos , Masculino , Estudios Retrospectivos , Esteroides , Resultado del Tratamiento
5.
Cureus ; 13(8): e16955, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34513521

RESUMEN

Introduction Systemic lupus erythematosus (SLE) is a systemic disease with clinically heterogeneous outcomes. Lupus nephritis (LN) is a common complication of SLE. LN impacts clinical SLE outcomes both directly, in the form of target organ damage, and indirectly, through the adverse effects of immunosuppressive therapy. Patients & methods The study included 402 SLE cases with biopsy-proven lupus nephritis who were under follow-up for the past 13 years at Mansoura Urology and Nephrology Center, Egypt. We studied the differences in outcome among various LN classes and between 275 proliferative cases and 102 non-proliferative cases. Results Class IV was the main LN class in our series with renal survival of 60% at 10 years. The major induction regimen after the first biopsy was cyclophosphamide. Mycophenolate mofetil was the main induction and adjunctive regimen after the second biopsy. The mean follow-up period was 6.7 + 5.2 years. Higher serum creatinine, proteinuria, activity, and chronicity indices were noted in proliferative LN. Patients suffering from proliferative lesions received higher immunosuppression and demonstrated higher morbidity than those with non-proliferative lesions. Remission was higher among the non-proliferative compared to the proliferative group. Conclusions Serum creatinine, proteinuria, and LN class were the most relevant prognostic factors for renal survival among Egyptian LN patients.

6.
Front Immunol ; 12: 690623, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248981

RESUMEN

Mesenchymal stem cell (MSC)-based therapy for type 1 diabetes mellitus (T1DM) has been the subject matter of many studies over the past few decades. The wide availability, negligible teratogenic risks and differentiation potential of MSCs promise a therapeutic alternative to traditional exogenous insulin injections or pancreatic transplantation. However, conflicting arguments have been reported regarding the immunological profile of MSCs. While some studies support their immune-privileged, immunomodulatory status and successful use in the treatment of several immune-mediated diseases, others maintain that allogeneic MSCs trigger immune responses, especially following differentiation or in vivo transplantation. In this review, the intricate mechanisms by which MSCs exert their immunomodulatory functions and the influencing variables are critically addressed. Furthermore, proposed avenues to enhance these effects, including cytokine pretreatment, coadministration of mTOR inhibitors, the use of Tregs and gene manipulation, are presented. As an alternative, the selection of high-benefit, low-risk donors based on HLA matching, PD-L1 expression and the absence of donor-specific antibodies (DSAs) are also discussed. Finally, the necessity for the transplantation of human MSC (hMSC)-derived insulin-producing cells (IPCs) into humanized mice is highlighted since this strategy may provide further insights into future clinical applications.


Asunto(s)
Glucemia/metabolismo , Diferenciación Celular , Diabetes Mellitus Tipo 1/cirugía , Células Secretoras de Insulina/trasplante , Insulina/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Animales , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Humanos , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Células Madre Mesenquimatosas/inmunología , Fenotipo
7.
Stem Cell Rev Rep ; 16(6): 1156-1172, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32880857

RESUMEN

Mesenchymal stromal cells (MSCs) are an attractive option for cell therapy for type 1 diabetes mellitus (DM). These cells can be obtained from many sources, but bone marrow and adipose tissue are the most studied. MSCs have distinct advantages since they are nonteratogenic, nonimmunogenic and have immunomodulatory functions. Insulin-producing cells (IPCs) can be generated from MSCs by gene transfection, gene editing or directed differentiation. For directed differentiation, MSCs are usually cultured in a glucose-rich medium with various growth and activation factors. The resulting IPCs can control chemically-induced diabetes in immune-deficient mice. These findings are comparable to those obtained from pluripotent cells. PD-L1 and PD-L2 expression by MSCs is upregulated under inflammatory conditions. Immunomodulation occurs due to the interaction between these ligands and PD-1 receptors on T lymphocytes. If this function is maintained after differentiation, life-long immunosuppression or encapsulation could be avoided. In the clinical setting, two sites can be used for transplantation of IPCs: the subcutaneous tissue and the omentum. A 2-stage procedure is required for the former and a laparoscopic procedure for the latter. For either site, cells should be transplanted within a scaffold, preferably one from fibrin. Several questions remain unanswered. Will the transplanted cells be affected by the antibodies involved in the pathogenesis of type 1 DM? What is the functional longevity of these cells following their transplantation? These issues have to be addressed before clinical translation is attempted. Graphical Abstract Bone marrow MSCs are isolated from the long bone of SD rats. Then they are expanded and through directed differentiation insulin-producing cells are formed. The differentiated cells are loaded onto a collagen scaffold. If one-stage transplantation is planned, a drug delivery system must be incorporated to ensure immediate oxygenation, promote vascularization and provide some growth factors. Some mechanisms involved in the immunomodulatory function of MSCs. These are implemented either by cell to cell contact or by the release of soluble factors. Collectively, these pathways results in an increase in T-regulatory cells.


Asunto(s)
Células Secretoras de Insulina/citología , Células Madre Mesenquimatosas/citología , Animales , Células Inmovilizadas/citología , Edición Génica , Humanos , Inmunidad , Trasplante de Células Madre Mesenquimatosas
9.
Heliyon ; 6(5): e03914, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32395661

RESUMEN

BACKGROUND/AIM: Diabetes mellitus (DM) is a serious, chronic and epidemic disease. Its effective therapy with exogenous insulin places an overwhelming burden on the patient's lifestyle. Moreover, pancreatic islet transplantation is limited by the scarceness of donors and the need for chronic immunosuppression. Cell-based therapy is considered an alternative source of insulin-producing cells (IPCs); encapsulating such cellular grafts in immunoisolating devices would protect the graft from immune attack without the need for immunosuppression. Herein, we investigate the ability of TheraCyte capsule as an immunoisolating device to promote the maturation of differentiated rat bone marrow derived mesenchymal stem cells (BM-MSCs), transplanted subcutaneously to treat diabetic rats in comparison with intratesticular transplantation. MAIN METHODS: Rat BM-MSC were differentiated into IPCs, and either encapsulated in TheraCyte capsules for subcutaneous transplantation or transplanted intratesticular into diabetic rats. Serum insulin, C-peptide & blood glucose levels of transplanted animals were monitored. Retrieved cells were further characterized by immunofluorescence staining and gene expression analysis. KEY FINDINGS: Differentiated rat BM-MSC were able to produce insulin in vitro, ameliorate hyperglycemia in vivo and survive for 6 months post transplantation. Transplanted cells induced higher levels of insulin and C-peptide, lower levels of blood glucose in the cured animals of both experimental groups. Gene expression revealed a further in vivo maturation of the implanted cells. SIGNIFICANCE: These data suggest that TheraCyte encapsulation of allogeneic differentiated stem cells are capable of reversing hyperglycemia, which holds a great promise as a new cell based, clinically applicable therapies for diabetes.

10.
Biomed Res Int ; 2020: 7103053, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32051828

RESUMEN

Mesenchymal stem cells (MSCs) can be differentiated in vitro to form insulin-producing cells (IPCs). However, the proportion of induced cells is modest. Extracts from injured pancreata of rodents promoted this differentiation, and three upregulated proteins were identified in these extracts. The aim of this study was to evaluate the potential benefits of adding these proteins to the differentiation medium alone or in combination. Our results indicate that the proportion of IPCs among the protein(s)-supplemented samples was significantly higher than that in the samples with no added proteins. The yield from samples supplemented with PRDX6 alone was 4-fold higher than that from samples without added protein. These findings were also supported by the results of fluorophotometry. Gene expression profiles revealed higher levels among protein-supplemented samples. Significantly higher levels of GGT, SST, Glut-2, and MafB expression were noted among PRDX6-treated samples. There was a stepwise increase in the release of insulin and c-peptide, as a function of increasing glucose concentrations, indicating that the differentiated cells were glucose sensitive and insulin responsive. PRDX6 exerts its beneficial effects as a result of its biological antioxidant properties. Considering its ease of use as a single protein, PRDX6 is now routinely used in our differentiation protocols.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Insulina/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Peroxiredoxina VI/metabolismo , Peroxiredoxina VI/farmacología , Péptido C/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Factor de Transcripción MafB/metabolismo , Peroxiredoxina VI/genética , Somatostatina/metabolismo , Transcriptoma , gamma-Glutamiltransferasa/metabolismo
11.
Exp Clin Transplant ; 17(6): 823-827, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-29534659

RESUMEN

Transplant is the optimal therapy for patients with end-stage renal disease. Acute cellular rejection refractory to treatment remains a major risk factor for graft loss and poor outcomes. In this study, we describe a 39-year-old man who received a living-related kidney transplant. Two days after transplant, the patient displayed acute deterioration of graft function. Conventional anti-rejection therapy was initiated, but graft function did not improve. Biopsy revealed acute cellular rejection (grade IIA), and C4d and HLA antibodies were negative. Immunohistochemistry phenotyping revealed clusters of CD20-positive lymphocytes, with 80% being CD3 positive. Rituximab was prescribed, and graft function improved dramatically. After 1 week, a second graft biopsy was done due to lagging of graft function, shown by serum creatinine of 2.1 mg/dL. Biopsy revealed regenerating acute tubular necrosis with disappearance of the CD20-positive lymphocyte cluster infiltrates. Two year, after transplant, the patient's graft function maintained stable. Phenotyping of the cellular infiltrate is important as it may lead to a proper selection of immunosuppression and consequent improvement of graft outcome.


Asunto(s)
Antígenos CD20/inmunología , Rechazo de Injerto/prevención & control , Inmunidad Celular/efectos de los fármacos , Inmunosupresores/uso terapéutico , Trasplante de Riñón/efectos adversos , Riñón/efectos de los fármacos , Linfocitos/efectos de los fármacos , Rituximab/uso terapéutico , Adulto , Toma de Decisiones Clínicas , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Humanos , Inmunofenotipificación , Riñón/diagnóstico por imagen , Riñón/inmunología , Riñón/patología , Donadores Vivos , Linfocitos/inmunología , Linfocitos/patología , Masculino , Resultado del Tratamiento
12.
Cell Transplant ; 27(6): 937-947, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29860900

RESUMEN

Ten mongrel dogs were used in this study. Diabetes was chemically induced in 7 dogs, and 3 dogs served as normal controls. For each diabetic dog, 5 million human bone marrow-derived mesenchymal stem cells/kg were differentiated to form insulin-producing cells using a trichostatin-based protocol. Cells were then loaded in 2 TheraCyte capsules which were transplanted under the rectus sheath. One dog died 4 d postoperatively from pneumonia. Six dogs were followed up with for 6 to 18 mo. Euglycemia was achieved in 4 dogs. Their glucose tolerance curves exhibited a normal pattern demonstrating that the encapsulated cells were glucose sensitive and insulin responsive. In the remaining 2 dogs, the fasting blood sugar levels were reduced but did not reach normal values. The sera of all transplanted dogs contained human insulin and C-peptide with a negligible amount of canine insulin. Removal of the transplanted capsules was followed by prompt return of diabetes. Intracytoplasmic insulin granules were seen by immunofluorescence in cells from the harvested capsules. Furthermore, all pancreatic endocrine genes were expressed. This study demonstrated that the TheraCyte capsule or a similar device can provide adequate immunoisolation, an important issue when stem cells are considered for the treatment of type 1 diabetes mellitus.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/trasplante , Células Madre Mesenquimatosas/citología , Adulto , Animales , Células de la Médula Ósea/citología , Separación Celular , Células Cultivadas , Células Inmovilizadas/citología , Células Inmovilizadas/trasplante , Diabetes Mellitus Tipo 1/patología , Perros , Humanos , Masculino , Adulto Joven
13.
J Genet Eng Biotechnol ; 16(2): 433-440, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30733757

RESUMEN

Mesenchymal stem cells (MSCs) is a heterogeneous population. Muse cells is a rare pluripotent subpopulation within MSCs. This study aims to evaluate the pulirpotency and the ability of Muse cells to generate insulin producing cells (IPCs) after in vitro differentiation protocol compared to the non-Muse cells. Muse cells were isolated by FACSAria III cell sorter from adipose-derived MSCs and were evaluated for its pluripotency. Following in vitro differentiation, IPCs derived from Muse and non-Muse cells were evaluated for insulin production. Muse cells comprised 3.2 ±â€¯0.7% of MSCs, approximately 82% of Muse cells were positive for anti stage-specific embryonic antigen-3 (SSEA-3). Pluripotent markers were highly expressed in Muse versus non-Muse cells. The percentage of generated IPCs by flow cytometric analysis was higher in Muse cells. Under confocal microscopy, Muse cells expressed insulin and c-peptide while it was undetected in non-Muse cells. Our results introduced Muse cells as a new adult pluripotent subpopulation, which is capable to produce higher number of functional IPCs.

14.
Biomed Res Int ; 2017: 3854232, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28584815

RESUMEN

The aim of this study is to compare human bone marrow-derived mesenchymal stem cells (BM-MSCs) and adipose tissue-derived mesenchymal stem cells (AT-MSCs), for their differentiation potentials to form insulin-producing cells. BM-MSCs were obtained during elective orthotopic surgery and AT-MSCs from fatty aspirates during elective cosmetics procedures. Following their expansion, cells were characterized by phenotyping, trilineage differentiation ability, and basal gene expression of pluripotency genes and for their metabolic characteristics. Cells were differentiated according to a Trichostatin-A based protocol. The differentiated cells were evaluated by immunocytochemistry staining for insulin and c-peptide. In addition the expression of relevant pancreatic endocrine genes was determined. The release of insulin and c-peptide in response to a glucose challenge was also quantitated. There were some differences in basal gene expression and metabolic characteristics. After differentiation the proportion of the resulting insulin-producing cells (IPCs), was comparable among both cell sources. Again, there were no differences neither in the levels of gene expression nor in the amounts of insulin and c-peptide release as a function of glucose challenge. The properties, availability, and abundance of AT-MSCs render them well-suited for applications in regenerative medicine. Conclusion. BM-MSCs and AT-MSCs are comparable regarding their differential potential to form IPCs. The availability and properties of AT-MSCs render them well-suited for applications in regenerative medicine.


Asunto(s)
Tejido Adiposo/metabolismo , Células de la Médula Ósea/metabolismo , Péptido C/metabolismo , Diferenciación Celular , Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Células de la Médula Ósea/citología , Humanos , Secreción de Insulina , Células Madre Mesenquimatosas/citología
15.
Nephrourol Mon ; 8(3): e34770, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27570751

RESUMEN

BACKGROUND: Renal transplantation is the ideal method for management of end-stage renal disease. The use of living donors for renal transplantation was critical for early development in the field and preceded the use of cadaveric donors. Most donors are related genetically to the recipients, like a parent, a child, or a sibling of the recipient, but there are an increasing percentage of cases where donors are genetically unrelated like spouses, friends, or altruistic individuals. Donor shortages constitute the major barrier for kidney transplantation, and much effort has been made to increase the supply of living donors. The impact of donor source on the outcome of renal transplantation is not adequately studied in our country. OBJECTIVES: The aim of the study was to evaluate the impact of donor source on the outcome of live donor kidney transplantation. PATIENTS AND METHODS: From March 1976 to December 2013, the number of patients that underwent living renal transplantation sharing at least one HLA haplotype with their donors was 2,485. We divided these patients into two groups: (1) 2,075 kidney transplant recipients (1,554 or 74.9% male and 521 or 25.1% female) for whom the donors were living related, (2) 410 kidney transplant recipients (297 or 72.4% male and 113 or 27.6% female) for whom the donors were living unrelated. All patients received immunosuppressive therapy, consisting of a calcineurin inhibitor, mycophenolate mofetil, or azathioprine and prednisolone. We compared acute rejection and complication rates, as well as long-term graft and patient survival of both groups. Demographic characteristics were compared using the chi-square test. Graft survival and patient survival were calculated using the Kaplan-Meier method. RESULTS: The percentages of patients with acute vascular rejection were significantly higher in the unrelated group, while percentages of patients with no rejection were significantly higher in the related group, but there were no significant differences regarding patient and graft survivals between both groups. CONCLUSIONS: Kidney transplant recipients who received their grafts either from live related donors or live unrelated donors had comparable patient and graft survival outcomes.

16.
Biomed Res Int ; 2015: 575837, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26064925

RESUMEN

The aim of this study was to provide evidence for further in vivo maturation of insulin-producing cells (IPCs) derived from human bone marrow-derived mesenchymal stem cells (HBM-MSCs). HBM-MSCs were obtained from three insulin-dependent type 2 diabetic volunteers. Following expansion, cells were differentiated according to a trichostatin-A/GLP protocol. One million cells were transplanted under the renal capsule of 29 diabetic nude mice. Blood glucose, serum human insulin and c-peptide levels, and glucose tolerance curves were determined. Mice were euthanized 1, 2, 4, or 12 weeks after transplantation. IPC-bearing kidneys were immunolabeled, number of IPCs was counted, and expression of relevant genes was determined. At the end of in vitro differentiation, all pancreatic endocrine genes were expressed, albeit at very low values. The percentage of IPCs among transplanted cells was small (≤3%). Diabetic animals became euglycemic 8 ± 3 days after transplantation. Thereafter, the percentage of IPCs reached a mean of ~18% at 4 weeks. Relative gene expression of insulin, glucagon, and somatostatin showed a parallel increase. The ability of the transplanted cells to induce euglycemia was due to their further maturation in the favorable in vivo microenvironment. Elucidation of the exact mechanism(s) involved requires further investigation.


Asunto(s)
Diferenciación Celular/genética , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Trasplante de Células Madre Mesenquimatosas , Animales , Glucemia , Células de la Médula Ósea/citología , Diabetes Mellitus Experimental/patología , Glucagón/metabolismo , Humanos , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD
17.
Exp Clin Transplant ; 13 Suppl 1: 111-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25894138

RESUMEN

OBJECTIVES: Kidney donors, similar to the general population, are at risk for development of type 2 diabetes mellitus. The course of donors who develop type 2 diabetes mellitus has not been well studied. This work is aimed at estimating the incidence of diabetes after kidney donation, and study some risk factors and some complications of diabetes mellitus after donation. MATERIALS AND METHODS: The material of this record based work comprised the records 2267 donors who donated 1 of their kidneys between 1976 and 2014 in the Urology and Nephrology Center, Mansoura University, Egypt, and regularly followed-up at its outpatient clinic. There were 388 donors included in the study and their medical records were revised. RESULTS: Postdonation weight gain and family history of diabetes mellitus were statistically significant on both the development of diabetes mellitus, high, very high albuminuria, and/or decreased creatinine clearance. Metformin and insulin use seemed to significantly reduce the protein excretion, and creatinine clearance decline in the studied group. CONCLUSIONS: There is a significant effect of the family history of diabetes mellitus on the development of high, very high albuminuria, and/or decreased creatinine clearance.


Asunto(s)
Albuminuria/epidemiología , Diabetes Mellitus Tipo 2/epidemiología , Nefropatías Diabéticas/epidemiología , Trasplante de Riñón/efectos adversos , Donadores Vivos , Nefrectomía/efectos adversos , Adulto , Anciano , Albuminuria/diagnóstico , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/fisiopatología , Egipto/epidemiología , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Incidencia , Pruebas de Función Renal , Trasplante de Riñón/métodos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Factores de Tiempo , Aumento de Peso
18.
Exp Clin Transplant ; 13(1): 26-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25654411

RESUMEN

OBJECTIVES: New-onset diabetes mellitus after transplant is a common complication in renal allograft recipients. Recently, a high prevalence of diabetes mellitus has been reported in patients with chronic hepatitis C virus. The association between hepatitis C and diabetes mellitus is well demonstrated in the general population, but some controversy still exists. This work aimed to study the effect of pretransplant hepatitis C virus on the development of new-onset diabetes mellitus after transplant in Egyptian living-donor renal allotransplant recipients. MATERIALS AND METHODS: This retrospective single center study included 913 kidney transplant recipients who were transplanted at Mansoura Urology and Nephrology Center between 2000 and 2010. The patients were divided into 4 groups according to their hepatitis C virus serology and diabetic status. RESULTS: Pretransplant dialysis duration and number of blood transfusion units were statistically significant among both viremic and nonviremic groups. With respect to induction therapy, a highly statistical significance was observed between the 4 groups regarding presence and type of adjuvant therapy (P < .001). With respect to maintenance immunosuppression, high statistically significant results were observed regarding steroid and rapamycin between the 4 groups (P < .001) with lower significance regarding mycophenolate mofetil (P = .04) but no significance regarding azathioprine, cyclosporine, or tacrolimus therapy. Incidence of new-onset diabetes mellitus after transplant was statistically higher in the viremic than nonviremic group (P < .001). CONCLUSIONS: There was a positive correlation between incidence of new-onset diabetes mellitus after transplant and positive pretransplant hepatitis C virus status.


Asunto(s)
Diabetes Mellitus/etiología , Hepacivirus/patogenicidad , Hepatitis C/complicaciones , Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Donadores Vivos , Adolescente , Adulto , Biomarcadores/sangre , Diabetes Mellitus/sangre , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/virología , Egipto , Femenino , Supervivencia de Injerto , Hepatitis C/sangre , Hepatitis C/diagnóstico , Hepatitis C/virología , Humanos , Inmunosupresores/uso terapéutico , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/diagnóstico , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
19.
Biomed Res Int ; 2014: 832736, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818157

RESUMEN

INTRODUCTION: Many protocols were utilized for directed differentiation of mesenchymal stem cells (MSCs) to form insulin-producing cells (IPCs). We compared the relative efficiency of three differentiation protocols. METHODS: Human bone marrow-derived MSCs (HBM-MSCs) were obtained from three insulin-dependent type 2 diabetic patients. Differentiation into IPCs was carried out by three protocols: conophylline-based (one-step protocol), trichostatin-A-based (two-step protocol), and ß -mercaptoethanol-based (three-step protocol). At the end of differentiation, cells were evaluated by immunolabeling for insulin production, expression of pancreatic endocrine genes, and release of insulin and c-peptide in response to increasing glucose concentrations. RESULTS: By immunolabeling, the proportion of generated IPCs was modest ( ≃ 3%) in all the three protocols. All relevant pancreatic endocrine genes, insulin, glucagon, and somatostatin, were expressed. There was a stepwise increase in insulin and c-peptide release in response to glucose challenge, but the released amounts were low when compared with those of pancreatic islets. CONCLUSION: The yield of functional IPCs following directed differentiation of HBM-MSCs was modest and was comparable among the three tested protocols. Protocols for directed differentiation of MSCs need further optimization in order to be clinically meaningful. To this end, addition of an extracellular matrix and/or a suitable template should be attempted.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Insulina/biosíntesis , Células Madre Mesenquimatosas/citología , Adulto , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Péptido C/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Fenotipo
20.
Biomed Res Int ; 2013: 912413, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23878820

RESUMEN

INTRODUCTION: The aim of this report is to study the graft and patient survival in a large cohort of recipients with an analysis of factors that may affect the final outcomes. METHODS: Between March 1976 and March 2008, 1967 consecutive live-donor renal transplants were carried out. Various variables that may have an impact on patients and/or graft survival were studied in two steps. Initially, a univariate analysis was carried out. Thereafter, significant variables were embedded in a stepwise regression analysis. RESULTS: The overall graft survival was 86.7% and 65.5%, at 5 and 10 years, respectively. The projected half-life for grafts was 17.5 years and for patients was 22 years. Five factors had an independent negative impact on graft survival: donor's age, genetic considerations, the type of primary immunosuppression, number of acute rejection episodes, and total steroid dose during the first 3 months after transplantation. CONCLUSIONS: Despite refinements in tissue matching techniques and improvements in immunosuppression protocols, an important proportion of grafts is still lost following living donor kidney transplantation, presumably due to chronic allograft nephropathy.


Asunto(s)
Rechazo de Injerto/mortalidad , Supervivencia de Injerto , Trasplante de Riñón/mortalidad , Donadores Vivos/estadística & datos numéricos , Complicaciones Posoperatorias/mortalidad , Insuficiencia Renal Crónica/mortalidad , Insuficiencia Renal Crónica/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Comorbilidad , Egipto/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Pronóstico , Medición de Riesgo , Factores de Riesgo , Tasa de Supervivencia , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...