Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
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.
Exp Clin Transplant ; 21(5): 428-433, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37334690

RESUMEN

OBJECTIVES: The main risk factor for poor graft outcomes is refractory acute rejection and its consequences. In this study, we compared the efficacy of antithymocyte globulins versus other antirejection strategies in reversing refractory acute graft rejection after living donor renal transplant. MATERIALS AND METHODS: We retrospectively reviewed the records of 745 patients who received living-donor kidney transplants and experienced acute rejection episodes at Mansoura Urology and Nephrology Center in Egypt over the past 20 years. Based on the type of antirejection medication that they received, we divided patients into 2 groups, with 80 patients in the antithymocyte globulin group and 665 patients who had other antirejection strategies. By using event-based sequential graft biopsy histopathology analysis, we compared the efficacy of antithymocyte globulins in reversing refractory rejection in terms of graft and patient complications and survival. RESULTS: Patient survival was comparable in both groups; however, graft survival was better in the antithymocyte globulin group than in the other group; in addition, event-based sequential graft biopsies revealed a lower incidence of acute and chronic rejection episodes after treatment of severe acute rejection in the antithymocyte globulin group compared with the other group. Incidence of posttreatment complications, particularly infection and malignancy, was comparable in both groups. CONCLUSIONS: Our retrospective analysis of event-based sequential graft biopsy allowed us to track graft rejection resolution or worsening. Antithymocyte globulins are highly effective in reversing acute graft rejection when compared with other approaches, with no increased risk of infection or malignancy.


Asunto(s)
Suero Antilinfocítico , Trasplante de Riñón , Humanos , Suero Antilinfocítico/efectos adversos , Inmunosupresores/efectos adversos , Trasplante de Riñón/efectos adversos , Estudios Retrospectivos , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/prevención & control , Rechazo de Injerto/patología , Supervivencia de Injerto , Biopsia , Resultado del Tratamiento
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.
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
5.
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
7.
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.

8.
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
9.
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
10.
Stem Cell Res Ther ; 8(1): 254, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29115987

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) play different roles in modulating tumor progression, growth, and metastasis. MSCs are recruited to the tumor site in large numbers and subsequently have an important microenvironmental role in modulating tumor progression and drug sensitivity. However, the effect of the tumor microenvironment on MSC plasticity remains poorly understood. Herein, we report a paracrine effect of cancer cells, in which they secrete soluble factors that promote a more stem-like state in bone marrow mesenchymal stem cells (BM-MSCs). METHODS: The effect of soluble factors secreted from MCF7, Hela, and HepG2 cancer cell lines on BM-MSCs was assessed using a Transwell indirect coculture system. After 5 days of coculture, BM-MSCs were characterized by flow cytometry for surface marker expression, by qPCR for gene expression profile, and by confocal immunofluorescence for marker expression. We then measured the sensitivity of cocultured BM-MSCs to chemotherapeutic agents, their cell cycle profile, and their response to DNA damage. The sphere formation, invasive properties, and in-vivo performance of BM-MSCs after coculture with cancer cells were also measured. RESULTS: Indirect coculture of cancer cells and BM-MSCs, without direct cell contact, generated slow cycling, chemoresistant spheroid stem cells that highly expressed markers of pluripotency, cancer cells, and cancer stem cells (CSCs). They also displayed properties of a side population and enhanced sphere formation in culture. Accordingly, these cells were termed cancer-induced stem cells (CiSCs). CiSCs showed a more mesenchymal phenotype that was further augmented upon TGF-ß stimulation and demonstrated a high expression of the ß-catenin pathway and ALDH1A1. CONCLUSIONS: These findings demonstrate that MSCs, recruited to the tumor microenvironment in large numbers, may display cellular plasticity, acquire a more stem-like state, and acquire some properties of CSCs upon exposure to cancer cell-secreted factors. These acquired characteristics may contribute to tumor progression, survival, and metastasis. Our findings provide new insights into the interactions between MSCs and cancer cells, with the potential to identify novel molecular targets for cancer therapy.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Citometría de Flujo , Humanos , Microambiente Tumoral
11.
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
12.
Appl Radiat Isot ; 118: 131-135, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27640173

RESUMEN

A point source is used to investigate the effect of water phantom thickness and source-to-detector distance (SDD) on the sensitivity and counting efficiency of a rectangular detector gamma camera. The increase in water thickness resulted in an increase in scatter fraction, a decrease in sensitivity, and counting efficiency. The increase in SDD resulted in a decrease in sensitivity and an increase in counting efficiency. An SDD of 0.79±0.02m is found to provide a good compromise for acceptable sensitivity and reasonable counting efficiency.

13.
Neurourol Urodyn ; 35(4): 497-502, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25663249

RESUMEN

AIMS: Post-exstrophy incontinence is a challenge because continence is difficult to achieve and more difficult to maintain. Feasibility and outcomes of a bulbourethral sling to treat post-exstrophy incontinence is shown in this report. METHODS: A retropubic bulbourethral sling was applied to male patients with incontinence post-exstrophy-epispadius repair. The study included children with total (continuous) incontinence who underwent multiple previous anti-incontinence procedures, ranging from bladder neck injection to bladder neck reconstruction. Preoperative assessment includes urinalysis, renal US, VCUG, 1-hr pad test and urodynamics. The bulbourethral sling applied is made of polypropylene and is suspended by 4 pairs of nylon sutures, to support the bulbar urethra within its covering muscles with the sutures tied on the rectus muscles. Continence was evaluated as well as adverse events. RESULTS: Seventeen children, (median age 8.7 years) completed 24-month of follow up. All had CPRE. Five children (29.27%) were dry. Four micturated through the urethra and one by catheterizing his cutaneous stoma every 3-4 hr. In none, PVR exceeded 10% of expected capacity. Four children underwent re-tightening 1-4 weeks after removal of urethral catheter. Perineal wound dehiscence occurred in one, perineal/suprapubic pain in seven and epididymo-orchitis in one child. CONCLUSION: The current technique is promising for difficult cases of incontinence after CPRE. It is safe, as no serious adverse events occurred during follow up period. It is economic and re-tightening is easy to perform. Neurourol. Urodynam. 35:497-502, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Procedimientos de Cirugía Plástica/efectos adversos , Complicaciones Posoperatorias/cirugía , Cabestrillo Suburetral , Uretra/cirugía , Incontinencia Urinaria/cirugía , Adolescente , Niño , Preescolar , Estudios de Seguimiento , Humanos , Masculino , Complicaciones Posoperatorias/etiología , Estudios Prospectivos , Vejiga Urinaria/cirugía , Incontinencia Urinaria/etiología , Urodinámica
14.
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
15.
Saudi J Kidney Dis Transpl ; 25(4): 733-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24969181

RESUMEN

To evaluate the role of power Doppler in the identification and differentiation between acute renal transplant rejection and acute tubular necrosis (ATN), we studied 67 live donor renal transplant recipients. All patients were examined by spectral and power Doppler sonography. Assessment of cortical perfusion (CP) by power Doppler was subjective, using our grading score system: P0 (normal CP); homogenous cortical blush extending to the capsule, P1 (reduced CP); cortical vascular cut-off at interlobular level, P2 (markedly reduced CP); scattered cortical color flow at the interlobar level. Renal biopsies were performed during acute graft dysfunction. Pathological diagnoses were based on Banff classification 1997. The Mann- Whitney test was used to test the difference between CP grades with respect to serum creatinine (SCr), and resistive index (RI). For 38 episodes of acute graft rejection grade I, power Doppler showed that CP was P1 and RI ranging from 0.78 to 0.89. For 21 episodes of acute graft rejection grade II, power Doppler showed that CP was P1, with RI ranging from 0.88 to >1. Only one case of grade III rejection had a CP of P2. Twelve biopsies of ATN had CP of P0 and RI ranging from 0.80 to 0.89 There was a statistically significant correlation between CP grading and SCr (P <0.01) as well as between CP grading and RI (P <0.05). CP grading had a higher sensitivity in the detection of early acute rejection compared with RI and cross-sectional area measurements. We conclude that power Doppler is a non-invasive sensitive technique that may help in the detection and differentiation between acute renal transplant rejection and ATN, particularly in the early post-transplantation period.


Asunto(s)
Rechazo de Injerto/diagnóstico por imagen , Trasplante de Riñón/efectos adversos , Necrosis Tubular Aguda/diagnóstico por imagen , Riñón/irrigación sanguínea , Riñón/diagnóstico por imagen , Imagen de Perfusión/métodos , Circulación Renal , Ultrasonografía Doppler , Adolescente , Adulto , Biopsia , Diagnóstico Diferencial , Femenino , Rechazo de Injerto/etiología , Rechazo de Injerto/fisiopatología , Humanos , Necrosis Tubular Aguda/etiología , Necrosis Tubular Aguda/fisiopatología , Donadores Vivos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
16.
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
17.
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
18.
J Nucleic Acids ; 2013: 194858, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762531

RESUMEN

Advanced type 2 diabetes mellitus is associated with significant morbidity and mortality due to cardiovascular, nervous, and renal complications. Attempts to cure diabetes mellitus using islet transplantation have been successful in providing a source for insulin secreting cells. However, limited donors, graft rejection, the need for continued immune suppression, and exhaustion of the donor cell pool prompted the search for a more sustained source of insulin secreting cells. Stem cell therapy is a promising alternative for islet transplantation in type 2 diabetic patients who fail to control hyperglycemia even with insulin injection. Autologous stem cell transplantation may provide the best outcome for those patients, since autologous cells are readily available and do not entail prolonged hospital stays or sustained immunotoxic therapy. Among autologous adult stem cells, mesenchymal stem cells (MSCs) therapy has been applied with varying degrees of success in both animal models and in clinical trials. This review will focus on the advantages of MSCs over other types of stem cells and the possible mechanisms by which MSCs transplant restores normoglycemia in type 2 diabetic patients. Sources of MSCs including autologous cells from diabetic patients and the use of various differentiation protocols in relation to best transplant outcome will be discussed.

19.
Appl Radiat Isot ; 77: 18-22, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23500653

RESUMEN

An evaluation of the effect of scattered radiation on the performance of a gamma camera is carried out using a specially designed home-made homogeneous circular planar flood source filled with 0.2±0.01 GBq of (99m)Tc solution as a scattering medium. The scatter effects are assessed by analyzing the energy spectrum of (99m)Tc for the scatter fraction and calculating the camera's extrinsic counting efficiency and sensitivity, for five flood source thicknesses (12, 50, 100, 150 and 200 mm ) and three source-to-detector distances (0.7, 0.9 and 1.1 m). Results showed an increase in the scatter fraction from 0.29 to 22.96 as the source thickness increased. This increase was associated with a decrease in the extrinsic sensitivity from 121.36 to 49.58 counts/s GBq, and a decrease in counting efficiency as from 3.78 to 1.55%. With the increase in source-to-detector distance, the extrinsic sensitivity decreases from 121.36 to 118.77 counts/s GBq, while the counting efficiency increases from 3.78 to 11.66%. It was found that a source-to-detector distance of 0.96±9×10(-3) m is a good compromise for an acceptable extrinsic sensitivity and a reasonable counting efficiency.


Asunto(s)
Artefactos , Cámaras gamma , Tecnecio/análisis , Tecnecio/química , Diseño de Equipo , Análisis de Falla de Equipo , Rayos gamma , Reproducibilidad de los Resultados , Dispersión de Radiación , Sensibilidad y Especificidad
20.
Cell Transplant ; 22(1): 133-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22710060

RESUMEN

Harvesting, expansion, and directed differentiation of human bone marrow-derived mesenchymal stem cells (BM-MSCs) could provide an autologous source of surrogate ß-cells that would alleviate the limitations of availability and/or allogenic rejection following pancreatic or islet transplantation. Bone marrow cells were obtained from three adult type 2 diabetic volunteers and three nondiabetic donors. After 3 days in culture, adherent MSCs were expanded for two passages. At passage 3, differentiation was carried out in a three-staged procedure. Cells were cultured in a glucose-rich medium containing several activation and growth factors. Cells were evaluated in vitro by flow cytometry, immunolabeling, RT-PCR, and human insulin and c-peptide release in responses to increasing glucose concentrations. One thousand cell clusters were inserted under the renal capsule of diabetic nude mice followed by monitoring of their diabetic status. At the end of differentiation, ∼5-10% of cells were immunofluorescent for insulin, c-peptide or glucagon; insulin, and c-peptide were coexpressed. Nanogold immunolabeling for electron microscopy demonstrated the presence of c-peptide in the rough endoplasmic reticulum. Insulin-producing cells (IPCs) expressed transcription factors and genes of pancreatic hormones similar to those expressed by pancreatic islets. There was a stepwise increase in human insulin and c-peptide release by IPCs in response to increasing glucose concentrations. Transplantation of IPCs into nude diabetic mice resulted in control of their diabetic status for 3 months. The sera of IPC-transplanted mice contained human insulin and c-peptide but negligible levels of mouse insulin. When the IPC-bearing kidneys were removed, rapid return of diabetic state was noted. BM-MSCs from diabetic and nondiabetic human subjects could be differentiated without genetic manipulation to form IPCs that, when transplanted, could maintain euglycemia in diabetic mice for 3 months. Optimization of the culture conditions are required to improve the yield of IPCs and their functional performance.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diabetes Mellitus Experimental/cirugía , Células Secretoras de Insulina/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Adulto , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Femenino , Expresión Génica , Humanos , Insulina/biosíntesis , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...