Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Adv Mater ; : e2400306, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38762768

RESUMEN

To date, strategies aiming to modulate cell to extracellular matrix (ECM) interactions during organoid derivation remain largely unexplored. Here renal decellularized ECM (dECM) hydrogels are fabricated from porcine and human renal cortex as biomaterials to enrich cell-to-ECM crosstalk during the onset of kidney organoid differentiation from human pluripotent stem cells (hPSCs). Renal dECM-derived hydrogels are used in combination with hPSC-derived renal progenitor cells to define new approaches for 2D and 3D kidney organoid differentiation, demonstrating that in the presence of these biomaterials the resulting kidney organoids exhibit renal differentiation features and the formation of an endogenous vascular component. Based on these observations, a new method to produce kidney organoids with vascular-like structures is achieved through the assembly of hPSC-derived endothelial-like organoids with kidney organoids in 3D. Major readouts of kidney differentiation and renal cell morphology are assessed exploiting these culture platforms as new models of nephrogenesis. Overall, this work shows that exploiting cell-to-ECM interactions during the onset of kidney differentiation from hPSCs facilitates and optimizes current approaches for kidney organoid derivation thereby increasing the utility of these unique cell culture platforms for personalized medicine.

2.
Cell Stem Cell ; 31(1): 52-70.e8, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38181751

RESUMEN

Human pluripotent stem cell-derived kidney organoids offer unprecedented opportunities for studying polycystic kidney disease (PKD), which still has no effective cure. Here, we developed both in vitro and in vivo organoid models of PKD that manifested tubular injury and aberrant upregulation of renin-angiotensin aldosterone system. Single-cell analysis revealed that a myriad of metabolic changes occurred during cystogenesis, including defective autophagy. Experimental activation of autophagy via ATG5 overexpression or primary cilia ablation significantly inhibited cystogenesis in PKD kidney organoids. Employing the organoid xenograft model of PKD, which spontaneously developed tubular cysts, we demonstrate that minoxidil, a potent autophagy activator and an FDA-approved drug, effectively attenuated cyst formation in vivo. This in vivo organoid model of PKD will enhance our capability to discover novel disease mechanisms and validate candidate drugs for clinical translation.


Asunto(s)
Cilios , Enfermedades Renales Poliquísticas , Humanos , Riñón , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Autofagia , Organoides
3.
Curr Opin Cell Biol ; 86: 102306, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38194750

RESUMEN

During embryogenesis, the mammalian kidney arises because of reciprocal interactions between the ureteric bud (UB) and the metanephric mesenchyme (MM), driving UB branching and nephron induction. These morphogenetic processes involve a series of cellular rearrangements that are tightly controlled by gene regulatory networks and signaling cascades. Here, we discuss how kidney developmental studies have informed the definition of procedures to obtain kidney organoids from human pluripotent stem cells (hPSCs). Moreover, bioengineering techniques have emerged as potential solutions to externally impose controlled microenvironments for organoid generation from hPSCs. Next, we summarize some of these advances with major focus On recent works merging hPSC-derived kidney organoids (hPSC-kidney organoids) with organ-on-chip to develop robust models for drug discovery and disease modeling applications. We foresee that, in the near future, coupling of different organoid models through bioengineering approaches will help advancing to recreate organ-to-organ crosstalk to increase our understanding on kidney disease progression in the human context and search for new therapeutics.


Asunto(s)
Estructuras Embrionarias , Riñón , Nefronas , Células Madre Pluripotentes , Humanos , Diferenciación Celular/fisiología , Riñón/fisiología , Riñón/embriología , Nefronas/embriología , Organoides
4.
Cancer Res ; 83(10): 1699-1710, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37129948

RESUMEN

Despite negative results of clinical trials conducted on the overall population of patients with gastric cancer, PARP inhibitor (PARPi) therapeutic strategy still might represent a window of opportunity for a subpopulation of patients with gastric cancer. An estimated 7% to 12% of gastric cancers exhibit a mutational signature associated with homologous recombination (HR) failure, suggesting that these patients could potentially benefit from PARPis. To analyze responsiveness of gastric cancer to PARPi, we exploited a gastroesophageal adenocarcinoma (GEA) platform of patient-derived xenografts (PDX) and PDX-derived primary cells and selected 10 PDXs with loss-of-function mutations in HR pathway genes. Cell viability assays and preclinical trials showed that olaparib treatment was effective in PDXs harboring BRCA2 germline mutations and somatic inactivation of the second allele. Olaparib responsive tumors were sensitive to oxaliplatin as well. Evaluation of HR deficiency (HRD) and mutational signatures efficiently stratified responder and nonresponder PDXs. A retrospective analysis on 57 patients with GEA showed that BRCA2 inactivating variants were associated with longer progression-free survival upon platinum-based regimens. Five of 7 patients with BRCA2 germline mutations carried the p.K3326* variant, classified as "benign." However, familial history of cancer, the absence of RAD51 foci in tumor cells, and a high HRD score suggest a deleterious effect of this mutation in gastric cancer. In conclusion, PARPis could represent an effective therapeutic option for BRCA2-mutated and/or high HRD score patients with GEA, including patients with familial intestinal gastric cancer. SIGNIFICANCE: PARP inhibition is a potential strategy for treating patients with gastric cancer with mutated BRCA2 or homologous repair deficiency, including patients with familial intestinal gastric cancer, for whom BRCA2 germline testing should be recommended.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Neoplasias Gástricas , Humanos , Femenino , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Mutación de Línea Germinal , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Estudios Retrospectivos , Proteína BRCA1/genética , Proteína BRCA2/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico
5.
Cell Metab ; 34(6): 857-873.e9, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35561674

RESUMEN

It is not well understood why diabetic individuals are more prone to develop severe COVID-19. To this, we here established a human kidney organoid model promoting early hallmarks of diabetic kidney disease development. Upon SARS-CoV-2 infection, diabetic-like kidney organoids exhibited higher viral loads compared with their control counterparts. Genetic deletion of the angiotensin-converting enzyme 2 (ACE2) in kidney organoids under control or diabetic-like conditions prevented viral detection. Moreover, cells isolated from kidney biopsies from diabetic patients exhibited altered mitochondrial respiration and enhanced glycolysis, resulting in higher SARS-CoV-2 infections compared with non-diabetic cells. Conversely, the exposure of patient cells to dichloroacetate (DCA), an inhibitor of aerobic glycolysis, resulted in reduced SARS-CoV-2 infections. Our results provide insights into the identification of diabetic-induced metabolic programming in the kidney as a critical event increasing SARS-CoV-2 infection susceptibility, opening the door to the identification of new interventions in COVID-19 pathogenesis targeting energy metabolism.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19 , Diabetes Mellitus , Nefropatías Diabéticas , Humanos , Riñón/metabolismo , Organoides , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2
6.
Nat Commun ; 13(1): 1503, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35314684

RESUMEN

Although reprogramming of cellular metabolism is a hallmark of cancer, little is known about how metabolic reprogramming contributes to early stages of transformation. Here, we show that the histone deacetylase SIRT6 regulates tumor initiation during intestinal cancer by controlling glucose metabolism. Loss of SIRT6 results in an increase in the number of intestinal stem cells (ISCs), which translates into enhanced tumor initiating potential in APCmin mice. By tracking down the connection between glucose metabolism and tumor initiation, we find a metabolic compartmentalization within the intestinal epithelium and adenomas, where a rare population of cells exhibit features of Warburg-like metabolism characterized by high pyruvate dehydrogenase kinase (PDK) activity. Our results show that these cells are quiescent cells expressing +4 ISCs and enteroendocrine markers. Active glycolysis in these cells suppresses ROS accumulation and enhances their stem cell and tumorigenic potential. Our studies reveal that aerobic glycolysis represents a heterogeneous feature of cancer, and indicate that this metabolic adaptation can occur in non-dividing cells, suggesting a role for the Warburg effect beyond biomass production in tumors.


Asunto(s)
Neoplasias , Sirtuinas , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Glucólisis/fisiología , Intestinos/patología , Ratones , Neoplasias/patología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Sirtuinas/metabolismo
7.
STAR Protoc ; 3(4): 101872, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595951

RESUMEN

This protocol presents the use of SARS-CoV-2 isolates to infect human kidney organoids, enabling exploration of the impact of SARS-CoV-2 infection in a human multicellular in vitro system. We detail steps to generate kidney organoids from human pluripotent stem cells (hPSCs) and emulate a diabetic milieu via organoids exposure to diabetogenic-like cell culture conditions. We further describe preparation and titration steps of SARS-CoV-2 virus stocks, their subsequent use to infect the kidney organoids, and assessment of the infection via immunofluorescence. For complete details on the use and execution of this protocol, please refer to Garreta et al. (2022).1.


Asunto(s)
COVID-19 , Células Madre Pluripotentes , Humanos , SARS-CoV-2 , Diferenciación Celular , Riñón , Organoides
8.
Gastric Cancer ; 24(4): 897-912, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33755862

RESUMEN

BACKGROUND: Trastuzumab is the only approved targeted therapy in patients with HER2-amplified metastatic gastric cancer (GC). Regrettably, in clinical practice, only a fraction of them achieves long-term benefit from trastuzumab-based upfront strategy. To advance precision oncology, we investigated the therapeutic efficacy of different HER2-targeted strategies, in HER2 "hyper"-amplified (≥ 8 copies) tumors. METHODS: We undertook a prospective evaluation of HER2 targeting with monoclonal antibodies, tyrosine kinase inhibitors and antibody-drug conjugates, in a selected subgroup of HER2 "hyper"-amplified gastric patient-derived xenografts (PDXs), through the design of ad hoc preclinical trials. RESULTS: Despite the high level of HER2 amplification, trastuzumab elicited a partial response only in 2 out of 8 PDX models. The dual-HER2 blockade with trastuzumab plus either pertuzumab or lapatinib led to complete and durable responses in 5 (62.5%) out of 8 models, including one tumor bearing a concomitant HER2 mutation. In a resistant PDX harboring KRAS amplification, the novel antibody-drug conjugate trastuzumab deruxtecan (but not trastuzumab emtansine) overcame KRAS-mediated resistance. We also identified a HGF-mediated non-cell-autonomous mechanism of secondary resistance to anti-HER2 drugs, responsive to MET co-targeting. CONCLUSION: These preclinical randomized trials clearly indicate that in HER2-driven gastric tumors, a boosted HER2 therapeutic blockade is required for optimal efficacy, leading to complete and durable responses in most of the cases. Our results suggest that a selected subpopulation of HER2-"hyper"-amplified GC patients could strongly benefit from this strategy. Despite the negative results of clinical trials, the dual blockade should be reconsidered for patients with clearly HER2-addicted cancers.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Medicina de Precisión/métodos , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Humanos , Inmunoconjugados/uso terapéutico , Estudios Prospectivos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Neoplasias Gástricas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 27(11): 3126-3140, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33542076

RESUMEN

PURPOSE: Gastric and gastroesophageal adenocarcinomas represent the third leading cause of cancer mortality worldwide. Despite significant therapeutic improvement, the outcome of patients with advanced gastroesophageal adenocarcinoma is poor. Randomized clinical trials failed to show a significant survival benefit in molecularly unselected patients with advanced gastroesophageal adenocarcinoma treated with anti-EGFR agents. EXPERIMENTAL DESIGN: We performed analyses on four cohorts: IRCC (570 patients), Foundation Medicine, Inc. (9,397 patients), COG (214 patients), and the Fondazione IRCCS Istituto Nazionale dei Tumori (206 patients). Preclinical trials were conducted in patient-derived xenografts (PDX). RESULTS: The analysis of different gastroesophageal adenocarcinoma patient cohorts suggests that EGFR amplification drives aggressive behavior and poor prognosis. We also observed that EGFR inhibitors are active in patients with EGFR copy-number gain and that coamplification of other receptor tyrosine kinases or KRAS is associated with worse response. Preclinical trials performed on EGFR-amplified gastroesophageal adenocarcinoma PDX models revealed that the combination of an EGFR mAb and an EGFR tyrosine kinase inhibitor (TKI) was more effective than each monotherapy and resulted in a deeper and durable response. In a highly EGFR-amplified nonresponding PDX, where resistance to EGFR drugs was due to inactivation of the TSC2 tumor suppressor, cotreatment with the mTOR inhibitor everolimus restored sensitivity to EGFR inhibition. CONCLUSIONS: This study underscores EGFR as a potential therapeutic target in gastric cancer and identifies the combination of an EGFR TKI and a mAb as an effective therapeutic approach. Finally, it recognizes mTOR pathway activation as a novel mechanism of primary resistance that can be overcome by the combination of EGFR and mTOR inhibitors.See related commentary by Openshaw et al., p. 2964.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Esofágicas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animales , Estudios de Cohortes , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Ratones , Terapia Molecular Dirigida , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
10.
Front Cell Dev Biol ; 8: 296, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32432111

RESUMEN

BACKGROUND: Cell therapies and derived products have a high potential in aiding tissue and organ repairing and have therefore been considered as potential therapies for treating renal diseases. However, few studies have evaluated the impact of these therapies according to the stage of chronic kidney disease. The aim of this study was to evaluate the renoprotective effect of murine bone marrow mesenchymal stromal cells (BM-MSCs), their extracellular vesicles (EVs) and EVs-depleted conditioned medium (dCM) in an aggressive mouse model of chronic cyclosporine (CsA) nephrotoxicity in a preventive and curative manner. METHODS: After 4 weeks of CsA-treatment (75 mg/kg daily) mice developed severe nephrotoxicity associated with a poor survival rate of 25%, and characterized by tubular vacuolization, casts, and cysts in renal histology. BM-MSC, EVs and dCM groups were administered as prophylaxis or as treatment of CsA nephrotoxicity. The effect of the cell therapies was analyzed by assessing renal function, histological damage, apoptotic cell death, and gene expression of fibrotic mediators. RESULTS: Combined administration of CsA and BM-MSCs ameliorated the mice survival rates (6-15%), but significantly renal function, and histological parameters, translating into a reduction of apoptosis and fibrotic markers. On the other hand, EVs and dCM administration were only associated with a partial recovery of renal function or histological damage. Better results were obtained when used as treatment rather than as prophylactic regimen i.e., cell therapy was more effective once the damage was established. CONCLUSION: In this study, we showed that BM-MSCs induce an improvement in renal outcomes in an animal model of CsA nephrotoxicity, particularly if the inflammatory microenvironment is already established. EVs and dCM treatment induce a partial recovery, indicating that further experiments are required to adjust timing and dose for better long-term outcomes.

11.
Updates Surg ; 72(4): 951-966, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32253687

RESUMEN

Patient-Derived Xenografts (PDXs) are, so far, the best preclinical model to validate targets and predictors of response to therapy. While subcutaneous implantation very rarely allows metastatic dissemination, orthotopic implantation (Patient-Derived Orthotopic Xenograft-PDOX) increases metastatic capability. Using a modified tool to analyze model validity, we performed a systematic review of Embase, PubMed, and Web of Science up to December 2018 to identify all original publications describing gastric cancer (GC) PDOXs. We identified ten studies of PDOX model validation from January 1981 to December 2018 that fulfilled the inclusion and exclusion criteria. Most models (70%) were derived from human GC cell lines rather than tissue fragments. In 90% of studies, the implantation was performed in the subserosal layer. Tumour engraftment rate ranged from 0 to 100%, despite the technique. Metastases were observed in 40% of PDOX models implanted into the subserosal layer, employing either cell suspension or cell line-derived tumour fragments. According to our modified model validity tool, half of the studies were defined as unclear because one or more validation criteria were not reported. Available GC PDOX models are not adequate according to our model validity tool. There is no demonstration that the submucosal site is more effective than the subserosal layer, and that tissue fragments are better than cell suspensions for successful engraftment and metastatic spread. Further studies should strictly employ model validity tools and large samples with orthotopic implant sites mirroring as much as possible the donor tumour characteristics.


Asunto(s)
Modelos Animales de Enfermedad , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias/métodos , Neoplasias Gástricas , Animales , Línea Celular Tumoral , Humanos , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Neoplasias Gástricas/patología
12.
Front Cell Dev Biol ; 8: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32064259

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) from different sources possess great therapeutic potential due to their immunomodulatory properties associated with allograft tolerance. However, a crucial role in this activity resides in extracellular vesicles (EVs) and signaling molecules secreted by cells. This study aimed to evaluate the immunomodulatory properties of donor and recipient MSCs isolated from adipose tissue (AD) or bone marrow (BM) and their EVs on kidney outcome in a rat kidney transplant model. METHODS: The heterotopic-kidney-transplant Fisher-to-Lewis rat model (F-L) was performed to study mixed cellular and humoral rejection. After kidney transplantation, Lewis recipients were assigned to 10 groups; two control groups; four groups received autologous MSCs (either AD- or BM- MSC) or EVs (derived from both cell types); and four groups received donor-derived MSCs or EVs. AD and BM-EVs were purified by ultracentrifugation. Autologous cell therapies were administered three times intravenously; immediately after kidney transplantation, 4 and 8 weeks, whereas donor-derived cell therapies were administered once intravenously immediately after transplantation. Survival and renal function were monitored. Twelve weeks after kidney transplantation grafts were harvested, infiltrating lymphocytes were analyzed by flow cytometry and histological lesions were characterized. RESULTS: Autologous AD- and BM-MSCs, but not their EVs, prolonged graft and recipient survival in a rat model of kidney rejection. Autologous AD- and BM-MSCs significantly improved renal function during the first 4 weeks after transplantation. The amelioration of graft function could be associated with an improvement in tubular damage, as well as in T, and NK cell infiltration. On the other side, the application of donor-derived AD-MSC was harmful, and all rats died before the end of the protocol. AD-EVs did not accelerate the rejection. Contrary to autologous MSCs results, the single dose of donor-derived BM-MSCs is not enough to ameliorate kidney graft damage. CONCLUSION: EVs treatments did not exert any benefit in our experimental settings. In the autologous setting, BM-MSCs prompted as a potentially promising therapy to improve kidney graft outcomes in rats with chronic mixed rejection. In the donor-derived setting, AD-MSC accelerated progression to end-stage kidney disease. Further experiments are required to adjust timing and dose for better long-term outcomes.

13.
Cancer Res ; 79(22): 5884-5896, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31585941

RESUMEN

Gastric cancer is the world's third leading cause of cancer mortality. In spite of significant therapeutic improvements, the clinical outcome for patients with advanced gastric cancer is poor; thus, the identification and validation of novel targets is extremely important from a clinical point of view. We generated a wide, multilevel platform of gastric cancer models, comprising 100 patient-derived xenografts (PDX), primary cell lines, and organoids. Samples were classified according to their histology, microsatellite stability, Epstein-Barr virus status, and molecular profile. This PDX platform is the widest in an academic institution, and it includes all the gastric cancer histologic and molecular types identified by The Cancer Genome Atlas. PDX histopathologic features were consistent with those of patients' primary tumors and were maintained throughout passages in mice. Factors modulating grafting rate were histology, TNM stage, copy number gain of tyrosine kinases/KRAS genes, and microsatellite stability status. PDX and PDX-derived cells/organoids demonstrated potential usefulness to study targeted therapy response. Finally, PDX transcriptomic analysis identified a cancer cell-intrinsic microsatellite instability (MSI) signature, which was efficiently exported to gastric cancer, allowing the identification, among microsatellite stable (MSS) patients, of a subset of MSI-like tumors with common molecular aspects and significant better prognosis. In conclusion, we generated a wide gastric cancer PDX platform, whose exploitation will help identify and validate novel "druggable" targets and optimize therapeutic strategies. Moreover, transcriptomic analysis of gastric cancer PDXs allowed the identification of a cancer cell-intrinsic MSI signature, recognizing a subset of MSS patients with MSI transcriptional traits, endowed with better prognosis. SIGNIFICANCE: This study reports a multilevel platform of gastric cancer PDXs and identifies a MSI gastric signature that could contribute to the advancement of precision medicine in gastric cancer.


Asunto(s)
Neoplasias Gástricas/genética , Transcripción Genética/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor/genética , Femenino , Perfilación de la Expresión Génica/métodos , Genes ras/genética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Inestabilidad de Microsatélites , Persona de Mediana Edad , Estadificación de Neoplasias/métodos , Fenotipo , Pronóstico , Neoplasias Gástricas/patología
14.
Sci Rep ; 9(1): 11177, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371743

RESUMEN

Colorectal cancer (CRC) occurs with more aggressiveness in kidney transplant recipients compared to the general population. Immunosuppressive therapy plays a crucial role in the development of post-transplant malignancy. Concretely, cyclosporine A (CsA) has intrinsic pro-oncologic properties, while several studies report a regression of cancer after the introduction of rapamycin (RAPA). However, their effect on the extracellular vesicle (EV) content from CRC cell lines and their relevance in the pre-metastatic niche have not yet been studied. Here, we investigated the effect of RAPA and CsA in EV-miRNAs from metastatic and non-metastatic CRC cell lines and the role of relevant miRNAs transferred into a pre-metastatic niche model. EV-miRNA profiles showed a significant upregulation of miR-6127, miR-6746-5p, and miR-6787-5p under RAPA treatment compared to CsA and untreated conditions in metastatic cell lines that were not observed in non-metastatic cells. From gene expression analysis of transfected lung fibroblasts, we identified 22 shared downregulated genes mostly represented by the histone family involved in chromatin organization, DNA packaging, and cell cycle. These results suggest that EV-miR-6127, miR-6746-5p and miR-6787-5p could be a potential epigenetic mechanism induced by RAPA therapy in the regulation of the pre-metastatic niche of post-transplant colorectal cancer.


Asunto(s)
Neoplasias del Colon/patología , Vesículas Extracelulares/patología , Terapia de Inmunosupresión/efectos adversos , MicroARNs/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Ciclosporina/farmacología , Epigénesis Genética , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Perfilación de la Expresión Génica , Humanos , Sirolimus/farmacología , Transcripción Genética/efectos de los fármacos
15.
Transplantation ; 102(7): 1075-1084, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29620612

RESUMEN

BACKGROUND: The progression from acute to chronic antibody-mediated rejection in kidney transplant recipients is usually not prevented by current therapeutic options. Here, we investigated whether the use of tofacitinib (TOFA), a Janus kinase 3 inhibitor, was capable of preventing the progression of allograft dysfunction in a Fisher-to-Lewis rat model of kidney transplantation. METHODS: Rats were treated from the third week after transplantation to allow the development of rejection. Treatment was based on cyclosporin A, rapamycin or TOFA. Renal function was assessed at 1, 4, 8, and 12 weeks after transplantation, whereas rat survival, histological lesions, and infiltrating lymphocytes were analyzed at 12 weeks. RESULTS: Tofacitinib prolonged graft survival, preserved tubular and glomerular structures and reduced humoral damage characterized by C4d deposition. Tofacitinib was able to reduce donor-specific antibodies. In addition, T and natural killer cell graft infiltration was reduced in TOFA-treated rats. Although rapamycin-treated rats also showed prolonged graft survival, glomerular structures were more affected. Moreover, only TOFA treatment reduced the presence of T, B and natural killer cells in splenic parenchyma. CONCLUSIONS: Tofacitinib is able to reduce the immune response generated in a rat model of kidney graft rejection, providing prolonged graft and recipient survival, better graft function, and less histological lesions.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Riñón/efectos adversos , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Animales , Enfermedad Crónica/prevención & control , Complemento C4b/inmunología , Complemento C4b/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Supervivencia de Injerto/efectos de los fármacos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunosupresores/uso terapéutico , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/metabolismo , Riñón/inmunología , Riñón/patología , Masculino , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Resultado del Tratamiento
16.
Transplant Direct ; 2(2): e65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27500257

RESUMEN

BACKGROUND: Sirolimus (SRL) has been associated with new-onset diabetes mellitus after transplantation. The aim was to determine the effect of SRL on development of insulin resistance and ß -cell toxicity. METHODS: Lean Zucker rat (LZR) and obese Zucker rat (OZR) were distributed into groups: vehicle and SRL (0.25, 0.5, or 1.0 mg/kg) during 12 or 28 days. Intraperitoneal glucose tolerance test (IPGTT) was evaluated at days 0, 12, 28, and 45. Islet morphometry, ß-cell proliferation, and apoptosis were analyzed at 12 days. Islets were isolated to analyze insulin content, insulin secretion, and gene expression. RESULTS: After 12 days, SRL treatment only impaired IPGTT in a dose-dependent manner in OZR. Treatment prolongation induced increase of area under the curve of IPGTT in LZR and OZR; however, in contrast to OZR, LZR normalized glucose levels after 2 hours. The SRL reduced pancreas weight and islet proliferation in LZR and OZR as well as insulin content. Insulin secretion was only affected in OZR. Islets from OZR + SRL rats presented a downregulation of Neurod1, Pax4, and Ins2 gene. Genes related with insulin secretion remained unchanged or upregulated. CONCLUSIONS: In conditions that require adaptive ß -cell proliferation, SRL might reveal harmful effects by blocking ß -cell proliferation, insulin production and secretion. These effects disappeared when removing the therapy.

17.
Transpl Int ; 29(9): 1008-16, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27208691

RESUMEN

Kaposi's sarcoma (KS) is one of the most frequent transplant related tumors. Several pathways are involved; however, the impact of the molecular phenotype associated to the tumor stage and the behavior-depending resultant therapy is still unknown. The aim of our study was to analyze the role of HHV-8 and mTOR pathway in tumor stages of skin KS after renal transplantation. Twelve renal transplant recipients with cutaneous KS from five transplant centers (1980-2007) under reduction of immunosuppression or conversion to mTOR inhibitor were included. The expression of HHV-8, PTEN, TGFß, VEGF, phospho-mTOR, and phospho-P70S6K in tumoral tissue was analyzed. KS lesions were classified as patch, plaque, and nodule state. HHV-8 infection was found in all tissue samples. KS lesions showed high activation of VEGF, p-mTOR and p-P70S6K, low PTEN, and null TGFß expression. The only pathway activated in a staging-dependent manner was mTOR with higher p-mTOR and p-P70S6K expression in nodule versus patch stage. KS lesions disappeared after 5.24 months in all converted patients without any recurrence in 14.05 years of mean follow-up. The activation of mTOR pathway according to KS stages supports the rational of the mTOR inhibitor in post-transplant Kaposi.


Asunto(s)
Trasplante de Riñón , Estadificación de Neoplasias/métodos , Sarcoma de Kaposi/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Anciano , Femenino , Supervivencia de Injerto , Herpesvirus Humano 8 , Humanos , Terapia de Inmunosupresión , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia , Fosfohidrolasa PTEN/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sarcoma de Kaposi/virología , España , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Transpl Int ; 28(10): 1240-4, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25939687

RESUMEN

Cancer in transplant recipients represents a therapeutic challenge even when the patient is already under mTOR inhibitors. A 78-year-old man received a deceased donor kidney transplant in 1993. After 6 months, he developed a multifocal cutaneous and nonvisceral Kaposi's Sarcoma while on cyclosporine immunosuppressant therapy. The patient was converted to sirolimus monotherapy in 2001 with subsequent complete recovery within 2 years. In 2007, the patient was diagnosed with an esophageal adenocarcinoma stage IIA. An esophagectomy was performed without requirement of further treatment. He has continued on sirolimus monotherapy ever since, with no other incidents and no recurrences of either tumor. In this report, we describe an interesting case of a second cancer while on immunosuppressive therapy with anticancer activity. Moreover, the present knowledge of the matter is discussed.


Asunto(s)
Adenocarcinoma , Neoplasias Esofágicas , Inmunosupresores/uso terapéutico , Trasplante de Riñón , Neoplasias Primarias Secundarias , Complicaciones Posoperatorias , Sirolimus/uso terapéutico , Adenocarcinoma/cirugía , Anciano , Azatioprina/uso terapéutico , Biomarcadores , Ciclosporina/efectos adversos , Ciclosporina/uso terapéutico , Sustitución de Medicamentos , Neoplasias Esofágicas/cirugía , Esofagectomía , Humanos , Huésped Inmunocomprometido , Inmunosupresores/efectos adversos , Masculino , Proteínas de Neoplasias/metabolismo , Neoplasias Primarias Secundarias/cirugía , Fosforilación , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/cirugía , Prednisona/efectos adversos , Prednisona/uso terapéutico , Procesamiento Proteico-Postraduccional , Sarcoma de Kaposi/etiología , Transducción de Señal , Sirolimus/efectos adversos , Neoplasias Cutáneas/etiología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
19.
J Biol Chem ; 287(29): 24131-8, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22613719

RESUMEN

The tubular epithelium of the kidney is susceptible to injury from a number of different causes, including inflammatory and immune disorders, oxidative stress, and nephrotoxins, among others. Primary renal epithelial cells remain one of the few tools for studying the biochemical and physiological characteristics of the renal tubular system. Nevertheless, differentiated primary cells are not suitable for recapitulation of disease properties that might arise during embryonic kidney formation and further maturation. Thus, cellular systems resembling kidney characteristics are in urgent need to model disease as well as to establish reliable drug-testing platforms. Induced pluripotent stem cells (iPSCs) bear the capacity to differentiate into every cell lineage comprising the adult organism. Thus, iPSCs bring the possibility for recapitulating embryonic development by directed differentiation into specific lineages. iPSC differentiation ultimately allows for both disease modeling in vitro and the production of cellular products with potential for regenerative medicine. Here, we describe the rapid, reproducible, and highly efficient generation of iPSCs derived from endogenous kidney tubular renal epithelial cells with only two transcriptional factors, OCT4 and SOX2. Kidney-derived iPSCs may provide a reliable cellular platform for the development of kidney differentiation protocols allowing drug discovery studies and the study of kidney pathology.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Persona de Mediana Edad , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética
20.
Transplantation ; 93(9): 874-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357177

RESUMEN

BACKGROUND: Mammalian target of rapamycin (mTOR) inhibition has been associated with gonadal dysfunction. The aim of this study was to characterize the effect of sirolimus (SRL) on male gonadal function in an experimental model. METHODS: Male Wistar rats were treated with intraperitoneal administration of vehicle or SRL. Vehicle group was treated for 12 weeks. Rats treated with SRL were killed at 4, 8, and 12 weeks. A group of rats was treated with SRL for 4 weeks and then observed during 8 weeks to analyze the possible reversibility of the effect of mTOR inhibition. Body and testicular weight, testosterone, follicle-stimulating hormone level, and luteinizing hormone level were measured and testicular histology was analyzed including proliferation and apoptosis analysis. RESULTS: Testicular weight was significantly lower in all SRL groups. After SRL withdrawal testicular weight had partially recovered. The expression of steroidogenic acute regulatory protein decreased during SRL treatment, which could explain the reduction of testosterone levels, because steroidogenic acute regulatory protein is crucial for testosterone synthesis. Spermatogenesis was blocked on the spermatogonial level by SRL treatment. Withdrawal of SRL treatment led to complete recovery. CONCLUSIONS: mTOR inhibition in healthy animals produces sexual hormone dysfunction, seminiferous tubule dystrophy and spermatogenesis blockade. Furthermore, the spermatogenesis blockade produced by SRL is reversible.


Asunto(s)
Sirolimus/toxicidad , Espermatogénesis/efectos de los fármacos , Enfermedades Testiculares/inducido químicamente , Testículo/efectos de los fármacos , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Estudios de Seguimiento , Inmunosupresores/administración & dosificación , Inmunosupresores/toxicidad , Inyecciones Intraperitoneales , Masculino , Tamaño de los Órganos , Fosfoproteínas/biosíntesis , Fosfoproteínas/efectos de los fármacos , Ratas , Ratas Wistar , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Enfermedades Testiculares/metabolismo , Enfermedades Testiculares/patología , Testículo/metabolismo , Testículo/patología , Testosterona/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA