Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Intervalo de año de publicación
1.
CEN Case Rep ; 13(1): 26-31, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37120491

RESUMEN

Critical illness polyneuropathy (CIP) is a very rare complication of sepsis and multi-organ failure. Herein, we report the first case of CIP reported in a patient on maintenance hemodialysis, who improved with rehabilitation. A 55-year-old male patient was emergently admitted with fever and altered consciousness and diagnosed with bacterial meningitis based on cerebral spinal fluid and cranial magnetic resonance imaging findings. Methicillin-susceptible Staphylococcus aureus was detected in blood and cerebral spinal fluid cultures. Despite treatment with appropriate antibiotics, blood cultures were positive for 9 days and serum C-reactive protein (CRP) levels were persistently elevated. Magnetic resonance imaging of hands and feet to determine infection origin revealed osteomyelitis in several fingers and toes, which required the amputation of 14 necrotic fingers and toes. Thereafter, blood cultures became negative and CRP levels declined. However, flaccid paralysis was noted in both upper and lower extremities during sepsis treatment. Nerve conduction studies showed peripheral axonal disorder in motor and sensory nerves, and CIP was determined as the cause of paralysis based on the fulfillment of all four CIP diagnostic criteria. The patient's muscle strength improved with early and appropriate medical treatment and physical therapy, and he was discharged home 147 days after admission. Prolonged high-level inflammation is a cause of CIP. Patients on hemodialysis, who are potentially immunosuppressed and vulnerable to infection, are at high risk for CIP. In patients on maintenance hemodialysis who develop flaccid paralysis during treatment for severe infection, CIP should be considered for early diagnosis and intervention.


Asunto(s)
Polineuropatías , Sepsis , Masculino , Humanos , Persona de Mediana Edad , Staphylococcus aureus , Polineuropatías/diagnóstico , Polineuropatías/etiología , Polineuropatías/terapia , Sepsis/complicaciones , Diálisis Renal/efectos adversos , Parálisis/complicaciones , Diagnóstico Precoz
2.
Commun Biol ; 6(1): 1097, 2023 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-37898693

RESUMEN

Kidney organoids have shown promise as evaluation tools, but their in vitro maturity remains limited. Transplantation into adult mice has aided in maturation; however, their lack of urinary tract connection limits long-term viability. Thus, long-term viable generated nephrons have not been demonstrated. In this study, we present an approachable method in which mouse and rat renal progenitor cells are injected into the developing kidneys of neonatal mice, resulting in the generation of chimeric nephrons integrated with the host urinary tracts. These chimeric nephrons exhibit similar maturation to the host nephrons, long-term viability with excretion and reabsorption functions, and cisplatin-induced renal injury in both acute and chronic phases, as confirmed by single-cell RNA-sequencing. Additionally, induced human nephron progenitor cells differentiate into nephrons within the neonatal kidneys. Collectively, neonatal injection represents a promising approach for in vivo nephron generation, with potential applications in kidney regeneration, drug screening, and pathological analysis.


Asunto(s)
Cisplatino , Riñón , Ratones , Ratas , Animales , Humanos , Cisplatino/toxicidad , Regeneración , Nefronas , Células Madre
3.
Biochem Biophys Res Commun ; 662: 18-25, 2023 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-37094429

RESUMEN

The number of patients with end-stage renal failure is increasing annually worldwide and the problem is compounded by a shortage of renal transplantation donors. In our previous research, we have shown that transplantation of renal progenitor cells into the nephrogenic region of heterologous fetuses can induce the development of nephrons. We have also developed transgenic mice in which specific renal progenitor cells can be removed by drugs. By combining these two technologies, we have succeeded in generating human-mouse chimeric kidneys in fetal mice. We hope to apply these technologies to regenerative medicine. The quality of nephron progenitor cells (NPCs) derived from human pluripotent stem cells is important for the generation of chimeric kidneys, but there is currently no simple evaluation system for the chimerogenic potential of human NPCs. In this study, we focused on the fact that the re-aggregation of mouse renal progenitor cells can be used for nephron formation, even when merged into single cells. First, we examined the conditions under which nephron formation is likely to occur in mice during re-aggregation. Next, to improve the differentiation potential of human NPCs derived from pluripotent stem cells, NPCs were sorted using Integrin subunit alpha 8 (ITGA8). Finally, we demonstrated chimera formation between different species by mixing mouse cells with purified, selectively-induced human NPCs under optimum conditions. We observed these chimeric organoids at different time points to learn about these human-mouse chimeric structures at various stages of renal development. We found that the rate of chimera formation was affected by the purity of the human NPCs and the cell ratios used. We demonstrated that chimeric nephrons can be generated using a simple model, even between distant species. We believe that this admixture of human and mouse renal progenitor cells is a promising technology with potential application for the evaluation of the chimera formation abilities of NPCs.


Asunto(s)
Riñón , Nefronas , Humanos , Ratones , Animales , Células Madre Embrionarias , Diferenciación Celular , Ratones Transgénicos , Organoides
4.
Intern Med ; 62(18): 2725-2730, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36754409

RESUMEN

Exercise-induced acute kidney injury (EIAKI) is frequently complicated with renal hypouricemia (RHUC). In patients with RHUC, limiting anaerobic exercise can prevent EIAKI. However, it is challenging to reduce exercise intensity in athletes. We herein report a 16-year-old Japanese football player with familial RHUC with compound heterozygous mutations in urate transporter 1 (URAT1) who presented with recurrent EIAKI. As prophylaxis (hydration during exercise) could not prevent EIAKI, febuxostat was initiated. EIAKI was not observed for 16 months despite exercising intensively. Hence, non-purine-selective xanthine oxidoreductase inhibitors may decrease the incidence of EIAKI in athletes with RHUC.


Asunto(s)
Lesión Renal Aguda , Transportadores de Anión Orgánico , Humanos , Adolescente , Xantina Deshidrogenasa , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Inhibidores Enzimáticos
5.
J Clin Med ; 11(23)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36498811

RESUMEN

To align the xeno-metanephros and renal progenitor cell timing for transplantation treatments, cryopreservation techniques and an efficient transportation of regenerated renal products such as xeno-metanephroi and renal progenitor cells should be established. Therefore, we propose a novel method of xenogeneic regenerative medicine for patients with chronic kidney disease by grafting porcine fetal kidneys injected with human renal progenitor cells. To develop a useful cryopreserve system of porcine fetal kidney and human renal progenitor cells, we examined the cryopreservation of a fetal kidney implanted with renal progenitor cells in a mouse model. First, we developed a new method for direct cell injection under the capsule of the metanephros using gelatin as a support for unzipped fetal kidneys. Then, we confirmed in vitro that the nephrons derived from the transplanted cells were regenerated even after cryopreservation before and after cell transplantation. Furthermore, the cryopreserved chimeric metanephroi grew, and regenerated nephrons were observed in NOD. We confirmed that even in cryopreserved chimeric metanephroi, transplanted cell-derived nephrons as well as fresh transplants grew.

6.
Cell Rep ; 39(11): 110933, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705028

RESUMEN

Generation of new kidneys can be useful in various research fields, including organ transplantation. However, generating renal stroma, an important component tissue for structural support, endocrine function, and kidney development, remains difficult. Organ generation using an animal developmental niche can provide an appropriate in vivo environment for renal stroma differentiation. Here, we generate rat renal stroma with endocrine capacity by removing mouse stromal progenitor cells (SPCs) from the host developmental niche and transplanting rat SPCs. Furthermore, we develop a method to replace both nephron progenitor cells (NPCs) and SPCs, called the interspecies dual replacement of the progenitor (i-DROP) system, and successfully generate functional chimeric kidneys containing rat nephrons and stroma. This method can generate renal tissue from progenitors and reduce xenotransplant rejection. Moreover, it is a safe method, as donor cells do not stray into nontarget organs, thus accelerating research on stem cells, chimeras, and xenotransplantation.


Asunto(s)
Riñón , Nefronas , Nicho de Células Madre , Células Madre , Animales , Diferenciación Celular , Quimera , Riñón/citología , Ratones , Nefronas/citología , Ratas , Células Madre/citología
8.
Acta Cir Bras ; 36(5): e360503, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34161432

RESUMEN

PURPOSE: As a classical xenotransplantation model, porcine kidneys have been transplanted into the lower abdomen of non-human primates. However, we have improved upon this model by using size-matched grafting in the orthotopic position. The beneficial aspects and surgical details of our method are reported herein. METHODS: Donors were two newborn pigs (weighting 5 to 6 kg) and recipients were two cynomolgus monkeys (weighting, approximately, 7 kg). After bilateral nephrectomy, kidneys were cold-transported in Euro-Collins solution. The porcine kidney was transplanted to the site of a left nephrectomy and fixed to the peritoneum. RESULTS: Kidneys transplanted to the lower abdomen by the conventional method were more susceptible to torsion of the renal vein (two cases). In contrast, early-stage blood flow insufficiency did not occur in orthotopic transplants of theleft kidney. CONCLUSIONS: Size-matched porcine-primate renal grafting using our method of transplanting tothe natural position of the kidneys contributes to stable post-transplant blood flow to the kidney.


Asunto(s)
Trasplante de Riñón , Trasplantes , Animales , Supervivencia de Injerto , Riñón/cirugía , Macaca fascicularis , Nefrectomía , Porcinos
9.
STAR Protoc ; 2(1): 100314, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33554147

RESUMEN

Renal progenitor cells induced from pluripotent stem cells have attracted attention as a cell source for organ regeneration. Here, we report an in vivo protocol for the regeneration of urine-producing nephrons, i.e., neo-nephrons, in mice. We outline steps to transplant exogenous renal progenitor cells into the nephrogenic zone of transgenic mice and subsequently analyze these neo-nephrons. For complete details on the use and execution of this protocol, please refer to Fujimoto et al. (2020).


Asunto(s)
Separación Celular/métodos , Nefronas/crecimiento & desarrollo , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular/fisiología , Humanos , Riñón/citología , Ratones , Ratones Transgénicos , Organogénesis/fisiología , Células Madre Pluripotentes/fisiología , Ratas , Regeneración/fisiología , Células Madre/metabolismo
10.
Acta cir. bras ; 36(5): e360503, 2021. tab, graf
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1278103

RESUMEN

ABSTRACT Purpose As a classical xenotransplantation model, porcine kidneys have been transplanted into the lower abdomen of non-human primates. However, we have improved upon this model by using size-matched grafting in the orthotopic position. The beneficial aspects and surgical details of our method are reported herein. Methods Donors were two newborn pigs (weighting 5 to 6 kg) and recipients were two cynomolgus monkeys (weighting, approximately, 7 kg). After bilateral nephrectomy, kidneys were cold-transported in Euro-Collins solution. The porcine kidney was transplanted to the site of a left nephrectomy and fixed to the peritoneum. Results Kidneys transplanted to the lower abdomen by the conventional method were more susceptible to torsion of the renal vein (two cases). In contrast, early-stage blood flow insufficiency did not occur in orthotopic transplants of theleft kidney. Conclusions Size-matched porcine-primate renal grafting using our method of transplanting tothe natural position of the kidneys contributes to stable post-transplant blood flow to the kidney.


Asunto(s)
Animales , Trasplante de Riñón , Trasplantes , Porcinos , Supervivencia de Injerto , Riñón/cirugía , Macaca fascicularis , Nefrectomía
11.
Cell Rep ; 32(11): 108130, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937125

RESUMEN

Animal fetuses may be used for the regeneration of human organs. We have previously generated a transgenic mouse model that allows diphtheria toxin (DT)-induced ablation of Six2-positive nephron progenitor cells (NPCs). Elimination of existing native host NPCs enables their replacement with donor NPCs, which can generate neo-nephrons. However, this system cannot be applied to human NPCs, because DT induces apoptosis in human cells. Therefore, the present study presents a transgenic mouse model for the ablation of NPCs using tamoxifen, which does not affect human cells. Using this system, we successfully regenerate interspecies neo-nephrons, which exhibit urine-producing abilities, from transplanted rat NPCs in a mouse host. Transplantation of human induced pluripotent stem cell (iPSC)-derived NPCs results in differentiation into renal vesicles, which connect to the ureteric bud of the host. Thus, we demonstrate the possibility of the regeneration of human kidneys derived from human iPSC-derived NPCs via NPC replacement.


Asunto(s)
Nefronas/citología , Regeneración , Células Madre/citología , Animales , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones Endogámicos C57BL , Nefronas/efectos de los fármacos , Nefronas/ultraestructura , Especificidad de Órganos , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Especificidad de la Especie , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo , Vejiga Urinaria/embriología , Micción/efectos de los fármacos
12.
Biochem Biophys Res Commun ; 520(3): 627-633, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31623827

RESUMEN

Kidney regenerative medicine is expected to be the solution to the shortage of organs for transplantation. In a previous report, we transplanted exogenous renal progenitor cells (RPCs) including nephron progenitor cells (NPCs), stromal progenitor cells (SPCs), and the ureteric bud (UB) into the nephrogenic zone of animal embryos and succeeded in regenerating new nephrons from exogenous NPCs through a fetal developmental program. However, it was unknown whether the renal stromal lineage cells were regenerated from SPCs. The present study aimed to verify the differentiation of SPCs into mesangial cells and renal stromal lineage cells. Here, we found that simply transplanting RPCs, including SPCs, into the nephrogenic zone of wild-type fetal mice was insufficient for differentiation of SPCs. Therefore, to enrich the purity of SPCs, we sorted cells from RPCs by targeting platelet-derived growth factor receptor alpha (PDGFRa) which is a cell surface marker for immature stromal cells and transplanted the PDGFRa-positive sorted cells. As a result, we succeeded in regenerating a large number of mesangial cells and other renal stromal lineage cells including interstitial fibroblasts, vascular pericytes, and juxtaglomerular cells. We have established the method for regeneration of stromal cells from exogenous SPCs that may contribute to various fields, such as regenerative medicine and kidney embryology, and the creation of disease models for renal stromal disorders.


Asunto(s)
Riñón/embriología , Células Mesangiales/fisiología , Regeneración/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Riñón/citología , Riñón/fisiología , Masculino , Células Mesangiales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Embarazo , Medicina Regenerativa , Trasplante de Células Madre , Células del Estroma/citología , Células del Estroma/fisiología , Células del Estroma/trasplante
13.
J Am Soc Nephrol ; 30(12): 2293-2305, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548350

RESUMEN

BACKGROUND: The limited availability of donor kidneys for transplantation has spurred interest in investigating alternative strategies, such as regenerating organs from stem cells transplanted into animal embryos. However, there is no known method for transplanting cells into later-stage embryos, which may be the most suitable host stages for organogenesis, particularly into regions useful for kidney regeneration. METHODS: We demonstrated accurate transplantation of renal progenitor cells expressing green fluorescent protein to the fetal kidney development area by incising the opaque uterine muscle layer but not the transparent amniotic membrane. We allowed renal progenitor cell-transplanted fetuses to develop for 6 days postoperatively before removal for analysis. We also transplanted renal progenitor cells into conditional kidney-deficient mouse embryos. We determined growth and differentiation of transplanted cells in all cases. RESULTS: Renal progenitor cell transplantation into the retroperitoneal cavity of fetuses at E13-E14 produced transplant-derived, vascularized glomeruli with filtration function and did not affect fetal growth or survival. Cells transplanted to the nephrogenic zone produced a chimera in the cap mesenchyme of donor and host nephron progenitor cells. Renal progenitor cells transplanted to conditional kidney-deficient fetuses induced the formation of a new nephron in the fetus that is connected to the host ureteric bud. CONCLUSIONS: We developed a cell transplantation method for midstage to late-stage fetuses. In vivo kidney regeneration from renal progenitor cells using the renal developmental environment of the fetus shows promise. Our findings suggest that fetal transplantation methods may contribute to organ regeneration and developmental research.


Asunto(s)
Células Madre Embrionarias/trasplante , Riñón/fisiología , Regeneración/fisiología , Animales , Transferencia de Embrión , Femenino , Genes Reporteros , Edad Gestacional , Riñón/citología , Riñón/embriología , Masculino , Mesodermo/citología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Nefronas/embriología , Espacio Retroperitoneal , Quimera por Trasplante
14.
Sci Rep ; 9(1): 6965, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061458

RESUMEN

Kidney regeneration is expected to be a new alternative treatment to the currently limited treatments for chronic kidney disease. By transplanting exogeneous nephron progenitor cells (NPCs) into the metanephric mesenchyme of a xenogeneic foetus, we aimed to regenerate neo-kidneys that originate from transplanted NPCs. Previously, we generated a transgenic mouse model enabling drug-induced ablation of NPCs (the Six2-iDTR mouse). We demonstrated that eliminating existing native host NPCs allowed their 100% replacement with donor mouse or rat NPCs, which could generate neo-nephrons on a culture dish. To apply this method to humans in the future, we examined the possibility of the in vivo regeneration of nephrons between different species via NPC replacement. We injected NPCs-containing rat renal progenitor cells and diphtheria toxin below the renal capsule of E13.5 metanephroi (MNs) of Six2-iDTR mice; the injected MNs were then transplanted into recipient rats treated with immunosuppressants. Consequently, we successfully regenerated rat/mouse chimeric kidneys in recipient rats receiving the optimal immunosuppressive therapy. We revealed a functional connection between the neo-glomeruli and host vessels and proper neo-glomeruli filtration. In conclusion, we successfully regenerated interspecies kidneys in vivo that acquired a vascular system. This novel strategy may represent an effective method for human kidney regeneration.


Asunto(s)
Riñón/citología , Mesodermo/citología , Nefronas/citología , Organogénesis , Regeneración , Trasplante de Células Madre/métodos , Células Madre/citología , Animales , Diferenciación Celular , Femenino , Riñón/fisiología , Masculino , Mesodermo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Nefronas/fisiología , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología , Quimera por Trasplante
15.
Sci Rep ; 8(1): 14919, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297790

RESUMEN

Kidney regeneration from pluripotent stem cells is receiving a lot of attention because limited treatments are currently available for chronic kidney disease (CKD). It has been shown that uremic state in CKD is toxic to somatic stem/progenitor cells, such as endothelial progenitor and mesenchymal stem cells, affecting their differentiation and angiogenic potential. Recent studies reported that specific abnormalities caused by the non-inherited disease are often retained in induced pluripotent stem cell (iPSC)-derived products obtained from patients. Thus, it is indispensable to first assess whether iPSCs derived from patients with CKD due to non-inherited disease (CKD-iPSCs) have the ability to generate kidneys. In this study, we generated iPSCs from patients undergoing haemodialysis due to diabetes nephropathy and glomerulonephritis (HD-iPSCs) as representatives of CKD-iPSCs or from healthy controls (HC-iPSCs). HD-iPSCs differentiated into nephron progenitor cells (NPCs) with similar efficiency to HC-iPSCs. Additionally, HD-iPSC-derived NPCs expressed comparable levels of NPC markers and differentiated into vascularised glomeruli upon transplantation into mice, as HC-iPSC-derived NPCs. Our results indicate the potential of HD-iPSCs as a feasible cell source for kidney regeneration. This is the first study paving the way for CKD patient-stem cell-derived kidney regeneration, emphasising the potential of CKD-iPSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Riñón/citología , Riñón/fisiología , Regeneración , Insuficiencia Renal Crónica/terapia , Animales , Diferenciación Celular , Células Cultivadas , Nefropatías Diabéticas/fisiopatología , Nefropatías Diabéticas/terapia , Glomerulonefritis/fisiopatología , Glomerulonefritis/terapia , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Riñón/fisiopatología , Ratones , Nefronas/citología , Nefronas/fisiología , Nefronas/fisiopatología , Diálisis Renal , Insuficiencia Renal Crónica/fisiopatología
16.
Biochem Biophys Res Commun ; 496(4): 1176-1182, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29408475

RESUMEN

To address the lack of organs for transplantation, we previously developed a method for organ regeneration in which nephron progenitor cell (NPC) replacement is performed via the diphtheria toxin receptor (DTR) system. In transgenic mice with NPC-specific expression of DTR, NPCs were eliminated by DT and replaced with NPCs lacking the DTR with the ability to differentiate into nephrons. However, this method has only been verified in vitro. For applications to natural models, such as animal fetuses, it is necessary to determine the optimal administration route and dose of DT. In this study, two DT administration routes (intra-peritoneal and intra-amniotic injection) were evaluated in fetal mice. The fetus was delivered by caesarean section at E18.5, and the fetal mouse kidney and RNA expression were evaluated. Additionally, the effect of the DT dose (25, 5, 0.5, and 0.05 ng/fetus-body) was studied. Intra-amniotic injection of DT led to a reduction in kidney volume, loss of glomeruli, and decreased differentiation marker expression. The intra-peritoneal route was not sufficient for NPC elimination. By establishing that intra-amniotic injection is the optimal administration route for DT, these results will facilitate studies of kidney regeneration in vivo. In addition, this method might be useful for analysis of kidney development at various time points by deleting NPCs during development.


Asunto(s)
Toxina Diftérica/administración & dosificación , Riñón/efectos de los fármacos , Riñón/crecimiento & desarrollo , Nefronas/citología , Regeneración/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Amnios , Animales , Relación Dosis-Respuesta a Droga , Femenino , Inyecciones Intraperitoneales , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nefronas/efectos de los fármacos , Regeneración/fisiología , Células Madre/fisiología , Resultado del Tratamiento
17.
Nephrology (Carlton) ; 20 Suppl 2: 81-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26031594

RESUMEN

We herein report a refractory case of subclinical antibody-mediated rejection (AMR) due to anti-HLA-DQ antibody in a kidney transplant patient. A 45-year-old man was admitted for a protocol biopsy; he had a serum creatinine (S-Cr) level of 1.8 mg/dL 3 years following primary kidney transplantation. Histological examination revealed moderate to severe inflammatory cell infiltration in the peritubular capillaries. Thorough laboratory examination showed that the patient had donor-specific antibodies (DSAbs) to DR9 and DQ9. Considering both the histological and laboratory findings, we diagnosed acute antibody-mediated rejection. The patient underwent 3 days of consecutive steroid pulse therapy, intravenous immunoglobulin (IVIG), and plasma exchange. We also administered rituximab (200 mg/body). Six months after the treatment, a second allograft biopsy revealed the progression of interstitial fibrosis and tubular atrophy and persistence of mild peritubular capillaritis. Further analysis showed that the anti-DR9 antibodies had disappeared, but that the mean fluorescence intensity value of the anti-DQ9 antibodies had increased. Therefore, we repeated the plasma exchange and IVIG. Allograft function was stable throughout the course of treatment, and the S-Cr level remained at 1.8 mg/dL. This case report demonstrates the difficulty of treating AMR due to the presence of anti-DQ DSAbs and the necessity for subsequent therapies in refractory cases.


Asunto(s)
Rechazo de Injerto/inmunología , Antígenos HLA-DQ/inmunología , Histocompatibilidad , Inmunidad Humoral , Isoanticuerpos/sangre , Trasplante de Riñón/efectos adversos , Aloinjertos , Enfermedades Asintomáticas , Biomarcadores/sangre , Biopsia , Creatinina/sangre , Rechazo de Injerto/patología , Rechazo de Injerto/terapia , Prueba de Histocompatibilidad , Humanos , Inmunoglobulinas Intravenosas/administración & dosificación , Inmunosupresores/administración & dosificación , Trasplante de Riñón/métodos , Donadores Vivos , Masculino , Persona de Mediana Edad , Intercambio Plasmático , Quimioterapia por Pulso , Esteroides/administración & dosificación , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...