Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Methods ; 99: 112-9, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26134528

RESUMEN

Kidney disease is a worldwide public health problem. Renal failure follows several disease stages including acute and chronic kidney symptoms. Acute kidney injury (AKI) may lead to chronic kidney disease (CKD), which can progress to end-stage renal disease (ESRD) with a mortality rate. Current treatment options are limited to dialysis and kidney transplantation; however, problems such as donor organ shortage, graft failure and numerous complications remain a concern. To address this issue, cell-based approaches using tissue engineering (TE) and regenerative medicine (RM) may provide attractive approaches to replace the damaged kidney cells with functional renal specific cells, leading to restoration of normal kidney functions. While development of renal tissue engineering is in a steady state due to the complex composition and highly regulated functionality of the kidney, cell therapy using stem cells and primary kidney cells has demonstrated promising therapeutic outcomes in terms of restoration of renal functions in AKI and CKD. In this review, basic components needed for successful renal kidney engineering are discussed, and recent TE and RM approaches to treatment of specific kidney diseases will be presented.


Asunto(s)
Enfermedades Renales/terapia , Ingeniería de Tejidos , Animales , Diferenciación Celular , Humanos , Riñón/patología , Riñón/fisiopatología , Técnicas de Cultivo de Órganos , Regeneración , Medicina Regenerativa , Trasplante de Células Madre , Células Madre/fisiología , Andamios del Tejido
2.
Curr Opin Organ Transplant ; 20(2): 165-70, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25856178

RESUMEN

PURPOSE OF REVIEW: Renal transplantation is currently the only definitive treatment for end-stage renal disease; however, this treatment is severely limited by the shortage of implantable kidneys. To address this shortcoming, development of an engineered, transplantable kidney has been proposed. Although current advances in engineering kidneys based on decellularization and recellularization techniques have offered great promises for the generation of functional kidney constructs, most studies have been conducted using rodent kidney constructs and short-term in-vivo evaluation. Toward clinical translations of this technique, several limitations need to be addressed. RECENT FINDINGS: Human-sized renal scaffolds are desirable for clinical application, and the fabrication is currently feasible using native porcine and discarded human kidneys. Current progress in stem cell biology and cell culture methods have demonstrated feasibility of the use of embryonic stem cells, induced pluripotent stem cells, and primary renal cells as clinically relevant cell sources for the recellularization of renal scaffolds. Finally, approaches to long-term implantation of engineered kidneys are under investigation using antithrombogenic strategies such as functional reendothelialization of acellular kidney matrices. SUMMARY: In the field of bioengineering, whole kidneys have taken a number of important initial steps toward clinical translations, but many challenges must be addressed to achieve a successful treatment for the patient with end-stage renal disease.


Asunto(s)
Trasplante de Riñón , Riñón/fisiología , Animales , Humanos , Riñón/cirugía , Fallo Renal Crónico/cirugía , Ingeniería de Tejidos/métodos , Andamios del Tejido
3.
FASEB J ; 26(1): 158-68, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21965595

RESUMEN

Whereas the conventional tissue engineering strategy involves the use of scaffolds combined with appropriate cell types to restore normal functions, the concept of in situ tissue regeneration uses host responses to a target-specific scaffold to mobilize host cells to a site of injury without the need for cell seeding. For this purpose, local delivery of bioactive molecules from scaffolds has been generally used. However, this approach has limited stem cell recruitment into the implants. Thus, we developed a combination of systemic delivery of substance P (SP) and local release of stromal-derived factor-1α (SDF-1α) from an implant. In this study, we examined whether this combined system would significantly enhance recruitment of host stem cells into the implants. Flow cytometry and immunohistochemistry for CD29/CD45, CD146/α-smooth muscle actin, and c-kit demonstrated that this system significantly increased the number of stem cell-like cells within the implants when compared with other systems. In vitro culture of the cells that had infiltrated into the scaffolds from the combined system confirmed that host stem cells were recruited into these implants and indicated that they were capable of differentiation into multiple lineages. These results indicate that this combined system may lead to more efficient tissue regeneration.


Asunto(s)
Quimiocina CXCL12/farmacocinética , Regeneración/fisiología , Células Madre/citología , Sustancia P/farmacocinética , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Sistemas de Liberación de Medicamentos/métodos , Citometría de Flujo , Gelatina , Ácido Láctico , Masculino , Ratones , Ratones Endogámicos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Neurotransmisores/farmacocinética , Poliésteres , Polímeros , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/fisiología
4.
Front Bioeng Biotechnol ; 10: 848679, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35646873

RESUMEN

Recent advances in cell therapy have shown the potential to treat kidney diseases. As the treatment effects of the cell therapies are mainly attributed to secretomes released from the transplanted cells, the delivery of secretomes or conditioned medium (CM) has emerged as a promising treatment option for kidney disease. We previously demonstrated that the controlled delivery of human placental stem cells (hPSC)-derived CM using platelet-rich plasma (PRP) ameliorated renal damages and restored kidney function in an acute kidney injury (AKI) model in rats. The proteomics study of the hPSC-CM revealed that hPSC secrets several proteins that contribute to kidney tissue repair. Based on our results, this study proposed that the proteins expressed in the hPSC-CM and effective for kidney repair could be used as a recombinant protein cocktail to treat kidney diseases as an alternative to CM. In this study, we analyzed the secretome profile of hPSC-CM and identified five proteins (follistatin, uPAR, ANGPLT4, HGF, VEGF) that promote kidney repair. We investigated the feasibility of delivering the recombinant protein cocktail to improve structural and functional recovery after AKI. The pro-proliferative and anti-apoptotic effects of the protein cocktail on renal cells are demonstrated in vitro and in vivo. The intrarenal delivery of these proteins with PRP ameliorates the renal tubular damage and improved renal function in the AKI-induced rats, yielding similar therapeutic effects compared to the CM delivery. These results indicate that our strategy may provide a therapeutic solution to many challenges associated with kidney repair resulting from the lack of suitable off-the-shelf regenerative medicine products.

5.
Mol Ther ; 18(7): 1365-72, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20389289

RESUMEN

Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is an inflammatory autoimmune disease characterized by T-cell infiltration to the colon. Mesenchymal stem cells (MSCs) have the potential to rescue IBD owing to their immunosuppressive capabilities and clinical studies have shown positive influence on intestinal graft versus host disease. We demonstrate here a new method to coat MSCs with antibodies against addressins to enhance their delivery to the colon and thereby increase the therapeutic effectiveness. Bioluminescence imaging (BLI) demonstrated that vascular cell adhesion molecule antibody (Ab)-coated MSCs (Ab(VCAM-1)- MSCs) had the highest delivery efficiency to inflamed mesenteric lymph node (MLN) and colon compared to untreated MSCs, Ab(isotype)-MSCs, and Ab(MAdCAM)-MSCs. Therapeutically, when mice with IBD were injected with addressin Ab-coated MSCs, they showed dramatically improved survival rates, higher IBD therapeutic scores, and significantly improved body weight gain compared to mice injected with MSCs only, isotype Ab, free Ab plus MSCs, or vehicle-only controls. These data demonstrate that anti-addressin Ab coating on MSC increased cell delivery to inflamed colon and increased the efficacy of MSC treatment of IBD. This is the first study showing an increased therapeutic efficacy when stem cells are first coated with antibodies specifically target them to inflamed sites.


Asunto(s)
Enfermedades Inflamatorias del Intestino/terapia , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología , Animales , Anticuerpos/química , Anticuerpos/inmunología , Línea Celular , Proliferación Celular , Células Cultivadas , Femenino , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/mortalidad , Mediciones Luminiscentes , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Ratones , Bazo/citología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología
6.
Acta Biomater ; 124: 233-243, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33524561

RESUMEN

Establishing an adequate vascularization of three-dimensional (3D) bioengineered tissues remains a critical challenge. We previously fabricated a vascular scaffold using the vascular corrosion casting technique, which provides a similar 3D geometry of native kidney vasculature. In this study, we functionalized the collagen vascular scaffold with a controlled release of vascular endothelial growth factor (VEGF vascular scaffold) to further promote vascularization. The VEGF vascular scaffold showed improved angiogenic capability in 2-dimensional (2D) and 3D in vitro settings. Implantation of the VEGF vascular scaffold seeded with human renal cells into a rat kidney demonstrated enhanced implant vascularization and reduced apoptosis of implanted human renal cells. Hybrid renal tubule-like structures composed of implanted human and migrated host renal cells were formed. This work highlights the critical role of early vascularization of the geometrically mimetic vascular scaffold using the VEGF incorporated vascular scaffold in reducing apoptosis of implanted cells as well as the formation of renal tissue structures.


Asunto(s)
Andamios del Tejido , Factor A de Crecimiento Endotelial Vascular , Colágeno , Molde por Corrosión , Riñón , Neovascularización Fisiológica , Ingeniería de Tejidos
7.
Acta Biomater ; 121: 237-249, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33321220

RESUMEN

Damages in pelvic floor muscles often cause dysfunction of the entire pelvic urogenital system, which is clinically challenging. A bioengineered skeletal muscle construct that mimics structural and functional characteristics of native skeletal muscle could provide a therapeutic option to restore normal muscle function. However, most of the current bioengineered muscle constructs are unable to provide timely innervation necessary for successful grafting and functional recovery. We previously have demonstrated that post-synaptic acetylcholine receptors (AChR) clusters can be pre-formed on cultured skeletal muscle myofibers with agrin treatment and suggested that implantation of AChR clusters containing myofibers could accelerate innervation and recovery of muscle function. In this study, we develop a 3-dimensional (3D) bioprinted human skeletal muscle construct, consisting of multi-layers bundles with aligned and AChR clusters pre-formed human myofibers, and investigate the effect of pre-formed AChR clusters in bioprinted skeletal muscle constructs and innervation efficiency in vivo. Agrin treatment successfully pre-formed functional AChR clusters on the bioprinted muscle constructs in vitro that increased neuromuscular junction (NMJ) formation in vivo in a transposed nerve implantation model in rats. In a rat model of pelvic floor muscle injury, implantation of skeletal muscle constructs containing the pre-formed AChR clusters resulted in functional muscle reconstruction with accelerated construct innervation. This approach may provide a therapeutic solution to the many challenges associated with pelvic floor reconstruction resulting from the lack of suitable bioengineered tissue for efficient innervation and muscle function restoration.


Asunto(s)
Fibras Musculares Esqueléticas , Diafragma Pélvico , Agrina , Animales , Músculo Esquelético , Unión Neuromuscular , Ratas , Recuperación de la Función
8.
Nat Commun ; 11(1): 1025, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094341

RESUMEN

A bioengineered skeletal muscle construct that mimics structural and functional characteristics of native skeletal muscle is a promising therapeutic option to treat extensive muscle defect injuries. We previously showed that bioprinted human skeletal muscle constructs were able to form multi-layered bundles with aligned myofibers. In this study, we investigate the effects of neural cell integration into the bioprinted skeletal muscle construct to accelerate functional muscle regeneration in vivo. Neural input into this bioprinted skeletal muscle construct shows the improvement of myofiber formation, long-term survival, and neuromuscular junction formation in vitro. More importantly, the bioprinted constructs with neural cell integration facilitate rapid innervation and mature into organized muscle tissue that restores normal muscle weight and function in a rodent model of muscle defect injury. These results suggest that the 3D bioprinted human neural-skeletal muscle constructs can be rapidly integrated with the host neural network, resulting in accelerated muscle function restoration.


Asunto(s)
Bioimpresión/métodos , Regeneración Tisular Dirigida/métodos , Enfermedades Musculares/terapia , Mioblastos Esqueléticos/fisiología , Neuronas/fisiología , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/uso terapéutico , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Estudios de Factibilidad , Humanos , Hidrogeles/química , Masculino , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología , Red Nerviosa/fisiología , Unión Neuromuscular/citología , Unión Neuromuscular/fisiología , Impresión Tridimensional , Ratas , Factores de Tiempo
9.
Int J Biol Sci ; 16(3): 435-446, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32015680

RESUMEN

Purpose: Drug-induced nephrotoxicity can occur in patients with pre-existing renal dysfunction or renal ischemia, potentially leading to chronic kidney disease (CKD) and end-stage renal disease (ESRD). Prompt treatment of CKD and the related side effects is critical in preventing progression to ESRD. The goal of this study was to demonstrate the therapeutic potential of urine-derived stem cells (USC) to treat chronic kidney disease-induced by nephrotoxic drugs and renal ischemia. Materials and methods: Human USC were collected, expanded and characterized by flow cytometry. A CKD model was induced by creating an ischemia-reperfusion injury and gentamicin administration. Twenty-eight adult immunodeficient rats were divided into three groups: PBS-treated group (n=9), USC-treated group (n=9), and sham group with age-matched control animals (n=10). Cell suspension of USC (5 x 106 / 100µl / kidney) or PBS was injected bilaterally into the renal parenchyma 9 weeks after CKD model creation. Renal function was evaluated by collection blood and urine samples to measure serum creatinine and glomerulus filtration rate. The kidneys were harvested 12 weeks after cell injection. Histologically, the extent of glomerulosclerosis and tubular atrophy, the amount of collagen deposition, interstitial fibrosis, inflammatory monocyte infiltration, and expression of transforming growth factor beta 1 (TGF-ß1), and superoxide dismutase 1 (SOD-1) were examined. Results: USC expressed renal parietal epithelial cells (CD24, CD29 and CD44). Renal function, measured by GFR and serum Cr in USC-treated group were significantly improved compared to PBS-treated animals (p<0.05). The degree of glomerular sclerosis and atrophic renal tubules, the amount of fibrosis, and monocyte infiltration significantly decreased in USC-treated group compared to the PBS group (p<0.05). The level of TGF-ß1 expression in renal tissues was also significantly lower in the PBS group, while the level of SOD-1 expression was significantly elevated in the USC group, compared to PBS group (p<0.05). Conclusions: The present study demonstrates the nephron-protective effect of USC on renal function via anti-inflammatory, anti-oxidative stress, and anti-fibrotic activity in a dual-injury CKD rat model. This provides an alternative treatment for CKD in certain clinical situations, such as instances where CKD is due to drug-induced nephrotoxicity and renal ischemia.


Asunto(s)
Diferenciación Celular/fisiología , Insuficiencia Renal Crónica/terapia , Daño por Reperfusión/terapia , Adipogénesis/fisiología , Animales , Fibrosis/metabolismo , Fibrosis/terapia , Humanos , Isquemia/metabolismo , Isquemia/terapia , Riñón/metabolismo , Riñón/patología , Masculino , Osteogénesis/fisiología , Ratas , Ratas Desnudas , Insuficiencia Renal Crónica/metabolismo , Daño por Reperfusión/metabolismo
10.
Vasc Specialist Int ; 35(2): 77-89, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31297357

RESUMEN

Engineering three-dimensional (3D) implantable tissue constructs is a promising strategy for replacing damaged or diseased tissues and organs with functional replacements. However, the efficient vascularization of new 3D organs is a major scientific and technical challenge since large tissue constructs or organs require a constant blood supply to survive in vivo. Current approaches to solving this problem generally fall into the following three major categories: (a) cell-based, (b) angiogenic factor-based, and (c) scaffold-based. In this review, we summarize state-of-the-art technologies that are used to develop complex, stable, and functional vasculature for engineered 3D tissue constructs and organs; additionally, we have suggested directions for future research.

11.
Acta Biomater ; 95: 328-336, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30953799

RESUMEN

We have developed a biomimetic renal vascular scaffold based on a vascular corrosion casting technique. This study evaluated the feasibility of using this novel biomimetic scaffold for kidney regeneration in a rat kidney cortical defect model. Vascular corrosion casts were prepared from normal rat kidneys by perfusion with 10% polycaprolactone (PCL) solution, followed by tissue digestion. The corrosion PCL cast was coated with collagen, and PCL was removed from within the collagen coating, leaving only a hollow collagen-based biomimetic vascular scaffold. The fabricated scaffolds were pre-vascularized with MS1 endothelial cell coating, incorporated into 3D renal constructs, and subsequently implanted either with or without human renal cells in the renal cortex of nude rats. The implanted collagen-based vascular scaffold was easily identified and integrated into native kidney tissue. The biomimetic vascular scaffold coated with endothelial cells (MS1) showed significantly enhanced vascularization, as compared to the uncoated scaffold and hydrogel only groups (P < 0.001). Along with the improved vascularization effects, the MS1-coated scaffolds showed a significant renal cell infiltration from the neighboring host tissue, as compared to the other groups (P < 0.05). Moreover, addition of human renal cells to the MS1-coated scaffold resulted in further enhancement of vascularization and tubular structure regeneration within the implanted constructs. The biomimetic collagen vascular scaffolds coated with endothelial cells are able to enhance vascularization and facilitate the formation of renal tubules after 14 days when combined with human renal cells. This study shows the feasibility of bioengineering vascularized functional renal tissues for kidney regeneration. STATEMENT OF SIGNIFICANCE: Vascularization is one of the major hurdles affecting the survival and integration of implanted three-dimensional tissue constructs in vivo. A novel, biomimetic, collagen-based vascular scaffold that is structurally identical to native kidney tissue was developed and tested. This biomimetic vascularized scaffold system facilitates the development of new vessels and renal cell viability in vivo when implanted in a partial renal defect. The use of this scaffold system could address the challenges associated with vascularization, and may be an ideal treatment strategy for partial augmentation of renal function in patients with chronic kidney disease.


Asunto(s)
Materiales Biomiméticos/farmacología , Molde por Corrosión , Riñón/fisiología , Regeneración/fisiología , Andamios del Tejido/química , Animales , Supervivencia Celular/efectos de los fármacos , Implantes Experimentales , Riñón/efectos de los fármacos , Riñón/cirugía , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Ratas Desnudas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
12.
J Tissue Eng Regen Med ; 13(7): 1230-1240, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31050866

RESUMEN

Treatment of extensive muscle loss due to traumatic injury, congenital defects, or tumor ablations is clinically challenging. The current treatment standard is grafting of autologous muscle flaps; however, significant donor site morbidity and graft tissue availability remain a problem. Alternatively, muscle fiber therapy has been attempted to treat muscle injury by transplanting single fibers into the defect site. However, irregularly organized long fibers resulted in low survivability due to delay in vascular and neural integration, thus limiting the therapeutic efficacy. Therefore, no effective method is available to permanently restore extensive muscle injuries. To address the current limitations, we developed a novel method that produces uniformly sized native muscle fiber fragments (MFFs) for muscle transplantation. We hypothesized that fragmentation of muscle fibers into small and uniformly sized fragments would allow for rapid reassembly and efficient engraftment within the defect site, resulting in accelerated recovery of muscle function. Our results demonstrate that the processed MFFs have a dimension of approximately 100 µm and contain living muscle cells on extracellular matrices. In preclinical animal studies using volumetric defect and urinary incontinence models, histological and functional analyses confirmed that the transplanted MFFs into the injury sites were able to effectively integrate with host muscle tissue, vascular, and neural systems, which resulted in significant improvement of muscle function and mass. These results indicate that the MFF technology platform is a promising therapeutic option for the restoration of muscle function and can be applied to various muscle defect and injury cases.


Asunto(s)
Fibras Musculares Esqueléticas , Recuperación de la Función , Regeneración , Trasplante de Tejidos , Heridas y Lesiones , Animales , Masculino , Ratones , Fibras Musculares Esqueléticas/fisiología , Fibras Musculares Esqueléticas/trasplante , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología , Heridas y Lesiones/fisiopatología , Heridas y Lesiones/terapia
13.
Tissue Eng Part A ; 25(21-22): 1493-1503, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30829146

RESUMEN

Kidney disease is a major medical problem globally. Chronic kidney disease (CKD) is a progressive loss of kidney function. It causes accumulation of waste and fluid in the body, eventually resulting in kidney failure as well as damaging other organs. Although dialysis and kidney transplantation have been used as primary treatments for renal disease, dialysis does not restore full renal function, and there is a shortage of donor kidneys for transplantation. Recent advances in cell-based therapies have offered a means to augment and restore renal function. Various types of cells have been tested to evaluate their therapeutic effects on injured kidneys. Among various types of cells, amniotic fluid stem cells (AFSCs) share advantages of both embryonic and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects, which may allow their future therapeutic use as an "off-the-shelf" cell source. AFSC presents advantages of both conventional pluripotent and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects. This study demonstrates that administration of human-derived AFSC facilitates functional and structural improvement in a rat model of CKD, and suggests that cell therapy with AFSC has potential as a therapeutic strategy to recover renal function in patients with CKD. Impact Statement Patients with chronic kidney disease (CKD) have limited treatment options, and renal transplantation is the only definitive treatment method that restores kidney function. However, challenges associated with transplantation, including donor organ shortage, rejection, and life-long immunosuppression, remain a problem. Recently, stem cell-based therapies have been proposed as an alternative approach to augment and restore renal function. In this study, we used human-derived amniotic fluid stem cells (AFSCs) to treat CKD in a rat model and demonstrated that AFSC treatment facilitated positive effects in terms of improvements of renal function.


Asunto(s)
Líquido Amniótico/citología , Pruebas de Función Renal , Riñón/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Humanos , Riñón/patología , Masculino , Podocitos/ultraestructura , Ratas Desnudas
14.
Stem Cells Transl Med ; 8(9): 959-970, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31144785

RESUMEN

Renal disease is a worldwide health issue. Besides transplantation, current therapies revolve around dialysis, which only delays disease progression but cannot replace other renal functions, such as synthesizing erythropoietin. To address these limitations, cell-based approaches have been proposed to restore damaged kidneys as an alternative to current therapies. Recent studies have shown that stem cell-derived secretomes can enhance tissue regeneration. However, many growth factors undergo rapid degradation when they are injected into the body in a soluble form. Efficient delivery and controlled release of secreting factors at the sites of injury would improve the efficacy in tissue regeneration. Herein, we developed a gel-based delivery system for controlled delivery of trophic factors in the conditioned medium (CM) secreted from human placental stem cells (HPSCs) and evaluated the effect of trophic factors on renal regeneration. CM treatment significantly enhanced cell proliferation and survival in vitro. Platelet-rich plasma (PRP) was used as a delivery vehicle for CM. Analysis of the release kinetics demonstrated that CM delivery through the PRP gel resulted in a controlled release of the factors both in vitro and in vivo. In an acute kidney injury model in rats, functional and structural analysis showed that CM delivery using the PRP gel system into the injured kidney minimized renal tissue damage, leading to a more rapid functional recovery when compared with saline, CM, or vehicle only injection groups. These results suggest that controlled delivery of HPSC-derived trophic factors may provide efficient repair of renal tissue injury. Stem Cells Translational Medicine 2019;8:959&970.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Riñón/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/metabolismo , Femenino , Geles/química , Riñón/citología , Riñón/patología , Masculino , Placenta/citología , Plasma Rico en Plaquetas/química , Embarazo , Ratas , Ratas Desnudas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Células Madre/citología , Células Madre/metabolismo
15.
Biomaterials ; 29(28): 3872-81, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18585778

RESUMEN

New acid-degradable cationic nanoparticles were synthesized using a monomer-to-polymer approach, which enabled highly flexible nanoparticle fabrication to obtain controlled properties such as size and conjugation with additional functionalities. The nanoparticles were designed to cause swelling and osmotic destabilization of the endosome, while cationic branches holding anionic DNA are cleaved from the polymeric backbone of the nanoparticles and make plasmid DNA accessible for efficient gene expression. Efficient release of plasmid DNA upon hydrolysis of the nanoparticles at the endosomal pH 5.0 and transportation of the released DNA to the nucleus of a cell were shown. In vitro studies showed significantly higher transfection efficiency by the degradable nanoparticles than polyethylenimine (PEI) polyplexes at very low concentrations (i.e., ng/mL). Size-dependent selective transfection of phagocytic cells (e.g., RAW 309 macrophages) and non-phagocytic cells (e.g., NIH 3T3 fibroblasts) was also achieved by using nanoparticles of two different sizes (240 nm and 680 nm in diameter), which implies feasibility of tunable gene therapy and DNA vaccination using the nanoparticle system. Preliminary pulmonary transfection of mice using the degradable nanoparticles demonstrated a remarkably higher expression of firefly luciferase at 70% lower concentration than using naked DNA alone. Implications and further improvement of the nanoparticles to be used in gene therapy are also discussed.


Asunto(s)
Acrilamidas/química , ADN/química , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/metabolismo , Plásmidos/genética , Polímeros/química , Acrilamidas/metabolismo , Animales , Línea Celular , Vectores Genéticos/genética , Luciferasas/genética , Luciferasas/metabolismo , Ensayo de Materiales , Ratones , Estructura Molecular , Tamaño de la Partícula , Polímeros/metabolismo , Transfección
16.
Yonsei Med J ; 59(9): 1015-1025, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30328315

RESUMEN

Kidney diseases including acute kidney injury and chronic kidney disease are among the largest health issues worldwide. Dialysis and kidney transplantation can replace a significant portion of renal function, however these treatments still have limitations. To overcome these shortcomings, a variety of innovative efforts have been introduced, including cell-based therapies. During the past decades, advances have been made in the stem cell and developmental biology, and tissue engineering. As part of such efforts, studies on renal cell therapy and artificial kidney developments have been conducted, and multiple therapeutic interventions have shown promise in the pre-clinical and clinical settings. More recently, therapeutic cell-secreting secretomes have emerged as a potential alternative to cell-based approaches. This approach involves the use of renotropic factors, such as growth factors and cytokines, that are produced by cells and these factors have shown effectiveness in facilitating kidney function recovery. This review focuses on the renotropic functions of bioactive compounds that provide protective and regenerative effects for kidney tissue repair, based on the available data in the literature.


Asunto(s)
Lesión Renal Aguda/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedades Renales/terapia , Medicina Regenerativa/métodos , Ingeniería de Tejidos , Lesión Renal Aguda/patología , Animales , Trasplante de Células , Citocinas , Humanos , Péptidos y Proteínas de Señalización Intercelular , Enfermedades Renales/patología , Trasplante de Células Madre , Células Madre , Ingeniería de Tejidos/métodos , Resultado del Tratamiento
17.
Sci Rep ; 8(1): 12307, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30120282

RESUMEN

A bioengineered skeletal muscle tissue as an alternative for autologous tissue flaps, which mimics the structural and functional characteristics of the native tissue, is needed for reconstructive surgery. Rapid progress in the cell-based tissue engineering principle has enabled in vitro creation of cellularized muscle-like constructs; however, the current fabrication methods are still limited to build a three-dimensional (3D) muscle construct with a highly viable, organized cellular structure with the potential for a future human trial. Here, we applied 3D bioprinting strategy to fabricate an implantable, bioengineered skeletal muscle tissue composed of human primary muscle progenitor cells (hMPCs). The bioprinted skeletal muscle tissue showed a highly organized multi-layered muscle bundle made by viable, densely packed, and aligned myofiber-like structures. Our in vivo study presented that the bioprinted muscle constructs reached 82% of functional recovery in a rodent model of tibialis anterior (TA) muscle defect at 8 weeks of post-implantation. In addition, histological and immunohistological examinations indicated that the bioprinted muscle constructs were well integrated with host vascular and neural networks. We demonstrated the potential of the use of the 3D bioprinted skeletal muscle with a spatially organized structure that can reconstruct the extensive muscle defects.


Asunto(s)
Bioimpresión/métodos , Músculo Esquelético/fisiología , Células Cultivadas , Humanos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido
18.
Acta Biomater ; 75: 226-234, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29883813

RESUMEN

Kidney transplantation is currently the only definitive solution for the treatment of end-stage renal disease (ESRD), however transplantation is severely limited by the shortage of available donor kidneys. Recent progress in whole organ engineering based on decellularization/recellularization techniques has enabled pre-clinical in vivo studies using small animal models; however, these in vivo studies have been limited to short-term assessments. We previously developed a decellularization system that effectively removes cellular components from porcine kidneys. While functional re-endothelialization on the porcine whole kidney scaffold was able to improve vascular patency, as compared to the kidney scaffold only, the duration of patency lasted only a few hours. In this study, we hypothesized that significant damage in the microvasculatures within the kidney scaffold resulted in the cessation of blood flow, and that thorough investigation is necessary to accurately evaluate the vascular integrity of the kidney scaffolds. Two decellularization protocols [sodium dodecyl sulfate (SDS) with DNase (SDS + DNase) or Triton X-100 with SDS (TRX + SDS)] were used to evaluate and optimize the levels of vascular integrity within the kidney scaffold. Results from vascular analysis studies using vascular corrosion casting and angiograms demonstrated that the TRX + SDS method was able to better maintain intact and functional microvascular architectures such as glomeruli within the acellular matrices than that by the SDS + DNase treatment. Importantly, in vitro blood perfusion of the re-endothelialized kidney construct revealed improved vascular function of the scaffold by TRX + SDS treatment compared with the SDS + DNase. Our results suggest that the optimized TRX + SDS decellularization method preserves kidney-specific microvasculatures and may contribute to long-term vascular patency following implantation. STATEMENT OF SIGNIFICANCE: Kidney transplantation is the only curative therapy for patients with end-stage renal disease (ESRD). However, in the United States, the supply of donor kidneys meets less than one-fifth of the demand; and those patients that receive a donor kidney need life-long immunosuppressive therapy to avoid organ rejection. In the last two decades, regenerative medicine and tissue engineering have emerged as an attractive alternative to overcome these limitations. In 2013, Song et al. published the first experimental orthotopic transplantation of a bioengineering kidney in rodents. In this study, they demonstrated evidences of kidney tissue regeneration and partial function restoration. Despite these initial promising results, there are still many challenges to achieve long-term blood perfusion without graft thrombosis. In this paper, we demonstrated that perfusion of detergents through the renal artery of porcine kidneys damages the glomeruli microarchitecture as well as peritubular capillaries. Modifying dynamic parameters such as flow rate, detergent concentration, and decellularization time, we were able to establish an optimized decellularization protocol with no evidences of disruption of glomeruli microarchitecture. As a proof of concept, we recellularized the kidney scaffolds with endothelial cells and in vitro perfused whole porcine blood successfully for 24 h with no evidences of thrombosis.


Asunto(s)
Vasos Sanguíneos/química , Riñón/irrigación sanguínea , Riñón/química , Andamios del Tejido/química , Animales , Porcinos
19.
Biomaterials ; 28(6): 1048-60, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17081602

RESUMEN

Neural stem cell (NSC) has emerged as a potential source for cell replacement therapy following traumatic injuries and degenerative diseases of the central nervous system. However, clinical applications of NSC further require technological advances especially for controlling differentiation of NSC. This study aimed at developing biomaterials that serve to expand undifferentiated NSC or to induce cells with specific phenotypes. Our approach is to construct composite biomaterials that consist of extracellular matrix components and growth factors. In order to optimize matrix-growth factor combinations, we conducted the parallel and rapid screening of composite biomaterials through assays using cell-based arrays. The photo-assisted patterning of an alkanethiol self-assembled monolayer was employed to achieve site-addressable combinatorial immobilization of natural and synthetic matrices incorporated with growth factors including epidermal growth factor (EGF), ciliary neurotrophic factor (CNTF), nerve growth factor (NGF), and neurotrophin-3 (NT-3). NSC obtained from the rat embryonic striatum was cultured directly on the array to screen for cell adhesion, proliferation, and promotion of neuronal and glial specification. The results showed that the significant number of cells adhered to laminin-1, fibronectin, ProNectin, and poly(ethyleneimine). It was found that cells proliferated most extensively on a spot with immobilized EGF among the spots with different matrix-growth factor combinations. The results also showed that neuronal differentiation was promoted on the spots with immobilized NGF or NT-3, and astroglial differentiation with CNTF. Importantly, observed effects of growth factors were frequently altered depending on the type of co-immobilized matrices, suggesting synergic effects of adhesion and growth factor signals.


Asunto(s)
Separación Celular/métodos , Ensayo de Materiales/métodos , Factores de Crecimiento Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/química , Neuronas/citología , Células Madre/citología , Ingeniería de Tejidos/métodos , Adsorción , Animales , Materiales Biocompatibles/química , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Técnicas Químicas Combinatorias , Factores de Crecimiento Nervioso/química , Neuronas/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Células Madre/efectos de los fármacos
20.
Curr Stem Cell Rep ; 3(2): 68-76, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33005562

RESUMEN

PURPOSE OF REVIEW: Historically, there have been many advances in the ways in which we treat kidney diseases. In particular, hemodialysis has set the standard for treatment since the early 1960s and continues today as the most common form of treatment for acute, chronic, and end-stage conditions. However, the rising global prevalence of kidney diseases and our limited understanding of their etiologies have placed significant burdens on current clinical management regimens. This has resulted in a desperate need to improve the ways in which we treat the underlying and ensuing causes of kidney diseases for those who are unable to receive transplants. RECENT FINDINGS: One way of possibly addressing these issues is through the use of improved bioartificial kidneys. Bioartificial kidneys provide an extension to conventional artificial kidneys and dialysis systems, by incorporating aspects of living cellular and tissue function, in an attempt to better mimic normal kidneys. Recent advancements in genomic, cellular, and tissue engineering technologies are facilitating the improved design of these systems. SUMMARY: In this review, we outline various research efforts that have focused on the development of regenerated organs, implantable constructs, and whole bioengineered kidneys, as well as the transitions from conventional dialysis to these novel alternatives. As a result, we envision that these pioneering efforts can one day produce bioartificial renal technologies that can either perform or reintroduce essential function, and thus provide practical options to treat and potentially prevent kidney diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA