Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Adv Mater ; : e2403594, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38639424

RESUMEN

Automated delivery of insulin based on continuous glucose monitoring is revolutionizing the way insulin-dependent diabetes is treated. However, challenges remain for the widespread adoption of these systems, including the requirement of a separate glucose sensor, sophisticated electronics and algorithms, and the need for significant user input to operate these costly therapies. Herein, a user-centric glucose-responsive cannula is reported for electronics-free insulin delivery. The cannula-made from a tough, elastomer-hydrogel hybrid membrane formed through a one-pot solvent exchange method-changes permeability to release insulin rapidly upon physiologically relevant varying glucose levels, providing simple and automated insulin delivery with no additional hardware or software. Two prototypes of the cannula are evaluated in insulin-deficient diabetic mice. The first cannula-an ends-sealed, subcutaneously inserted prototype-normalizes blood glucose levels for 3 d and controls postprandial glucose levels. The second, more translational version-a cannula with the distal end sealed and the proximal end connected to a transcutaneous injection port-likewise demonstrates tight, 3-d regulation of blood glucose levels when refilled twice daily. This proof-of-concept study may aid in the development of "smart" cannulas and next-generation insulin therapies at a reduced burden-of-care toll and cost to end-users.

2.
Chaos ; 34(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38457847

RESUMEN

The functional networks of the human brain exhibit the structural characteristics of a scale-free topology, and these neural networks are exposed to the electromagnetic environment. In this paper, we consider the effects of magnetic induction on synchronous activity in biological neural networks, and the magnetic effect is evaluated by the four-stable discrete memristor. Based on Rulkov neurons, a scale-free neural network model is established. Using the initial value and the strength of magnetic induction as control variables, numerical simulations are carried out. The research reveals that the scale-free neural network exhibits multiple coexisting behaviors, including resting state, period-1 bursting synchronization, asynchrony, and chimera states, which are dependent on the different initial values of the multi-stable discrete memristor. In addition, we observe that the strength of magnetic induction can either enhance or weaken the synchronization in the scale-free neural network when the parameters of Rulkov neurons in the network vary. This investigation is of significant importance in understanding the adaptability of organisms to their environment.

3.
Nat Biomed Eng ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052996

RESUMEN

Cellular therapies for type-1 diabetes can leverage cell encapsulation to dispense with immunosuppression. However, encapsulated islet cells do not survive long, particularly when implanted in poorly vascularized subcutaneous sites. Here we show that the induction of neovascularization via temporary controlled inflammation through the implantation of a nylon catheter can be used to create a subcutaneous cavity that supports the transplantation and optimal function of a geometrically matching islet-encapsulation device consisting of a twisted nylon surgical thread coated with an islet-seeded alginate hydrogel. The neovascularized cavity led to the sustained reversal of diabetes, as we show in immunocompetent syngeneic, allogeneic and xenogeneic mouse models of diabetes, owing to increased oxygenation, physiological glucose responsiveness and islet survival, as indicated by a computational model of mass transport. The cavity also allowed for the in situ replacement of impaired devices, with prompt return to normoglycemia. Controlled inflammation-induced neovascularization is a scalable approach, as we show with a minipig model, and may facilitate the clinical translation of immunosuppression-free subcutaneous islet transplantation.

4.
Sci Rep ; 13(1): 15641, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37730815

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease that leads to the loss of insulin-producing beta cells. Bioartificial pancreas (BAP) or beta cell replacement strategies have shown promise in curing T1D and providing long-term insulin independence. Hypoxia (low oxygen concentration) that may occur in the BAP devices due to cell oxygen consumption at the early stages after implantation damages the cells, in addition to imposing limitations to device dimensions when translating promising results from rodents to humans. Finding ways to provide cells with sufficient oxygenation remains the major challenge in realizing BAP devices' full potential. Therefore, in vitro oxygen imaging assessment of BAP devices is crucial for predicting the devices' in vivo efficiency. Electron paramagnetic resonance oxygen imaging (EPROI, also known as electron MRI or eMRI) is a unique imaging technique that delivers absolute partial pressure of oxygen (pO2) maps and has been used for cancer hypoxia research for decades. However, its applicability for assessing BAP devices has not been explored. EPROI utilizes low magnetic fields in the mT range, static gradients, and the linear relationship between the spin-lattice relaxation rate (R1) of oxygen-sensitive spin probes such as trityl OX071 and pO2 to generate oxygen maps in tissues. With the support of the Juvenile Diabetes Research Foundation (JDRF), an academic-industry partnership consortium, the "Oxygen Measurement Core" was established at O2M to perform oxygen imaging assessment of BAP devices originated from core members' laboratories. This article aims to establish the protocols and demonstrate a few examples of in vitro oxygen imaging of BAP devices using EPROI. All pO2 measurements were performed using a recently introduced 720 MHz/25 mT preclinical oxygen imager instrument, JIVA-25™. We began by performing pO2 calibration of the biomaterials used in BAPs at 25 mT magnetic field since no such data exist. We compared the EPROI pO2 measurement with a single-point probe for a few selected materials. We also performed trityl OX071 toxicity studies with fibroblasts, as well as insulin-producing cells (beta TC6, MIN6, and human islet cells). Finally, we performed proof-of-concept in vitro pO2 imaging of five BAP devices that varied in size, shape, and biomaterials. We demonstrated that EPROI is compatible with commonly used biomaterials and that trityl OX071 is nontoxic to cells. A comparison of the EPROI with a fluorescent-based point oxygen probe in selected biomaterials showed higher accuracy of EPROI. The imaging of typically heterogenous BAP devices demonstrated the utility of obtaining oxygen maps over single-point measurements. In summary, we present EPROI as a quality control tool for developing efficient cell transplantation devices and artificial tissue grafts. Although the focus of this work is encapsulation systems for diabetes, the techniques developed in this project are easily transferable to other biomaterials, tissue grafts, and cell therapy devices used in the field of tissue engineering and regenerative medicine (TERM). In summary, EPROI is a unique noninvasive tool to experimentally study oxygen distribution in cell transplantation devices and artificial tissues, which can revolutionize the treatment of degenerative diseases like T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Insulinas , Humanos , Oxígeno , Diabetes Mellitus Tipo 1/terapia , Hipoxia , Materiales Biocompatibles
5.
Semin Pediatr Surg ; 32(3): 151311, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37276782

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating neonatal intestinal disease associated with significant morbidity and mortality. Although decades of research have been dedicated to understanding the pathogenesis of NEC and developing therapies, it remains the leading cause of death among neonatal gastrointestinal diseases. Mesenchymal stem cells (MSCs) have garnered significant interest recently as potential therapeutic agents for the treatment of NEC. They have been shown to rescue intestinal injury and reduce the incidence and severity of NEC in various preclinical animal studies. MSCs and MSC-derived organoids and tissue engineered small intestine (TESI) have shown potential for the treatment of long-term sequela of NEC such as short bowel syndrome, neurodevelopmental delay, and chronic lung disease. Although the advances made in the use of MSCs are promising, further research is needed prior to the widespread use of these cells for the treatment of NEC.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Síndrome del Intestino Corto , Animales , Recién Nacido , Humanos , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/terapia , Células Madre/patología , Intestinos , Síndrome del Intestino Corto/terapia
6.
Sci Adv ; 9(24): eade9488, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37327334

RESUMEN

Biomedical devices comprise a major component of modern medicine, however immune-mediated fibrosis and rejection can limit their function over time. Here, we describe a humanized mouse model that recapitulates fibrosis following biomaterial implantation. Cellular and cytokine responses to multiple biomaterials were evaluated across different implant sites. Human innate immune macrophages were verified as essential to biomaterial rejection in this model and were capable of cross-talk with mouse fibroblasts for collagen matrix deposition. Cytokine and cytokine receptor array analysis confirmed core signaling in the fibrotic cascade. Foreign body giant cell formation, often unobserved in mice, was also prominent. Last, high-resolution microscopy coupled with multiplexed antibody capture digital profiling analysis supplied spatial resolution of rejection responses. This model enables the study of human immune cell-mediated fibrosis and interactions with implanted biomaterials and devices.


Asunto(s)
Materiales Biocompatibles , Cuerpos Extraños , Humanos , Animales , Ratones , Reacción a Cuerpo Extraño/etiología , Modelos Animales de Enfermedad , Citocinas , Fibrosis
7.
Semin Perinatol ; 47(3): 151727, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36964032

RESUMEN

Stem cell research and the use of stem cells in therapy have seen tremendous growth in the last two decades. Neonatal intestinal disorders such as necrotizing enterocolitis, Hirschsprung disease, and gastroschisis have high morbidity and mortality and limited treatment options with varying success rates. Stem cells have been used in several pre-clinical studies to address various neonatal disorders with promising results. Stem cell and patient population selection, timing of therapy, as well as safety and quality control are some of the challenges that must be addressed prior to the widespread clinical application of stem cells. Further research and technological advances such as the use of cell delivery technology can address these challenges and allow for continued progress towards clinical translation.


Asunto(s)
Enterocolitis Necrotizante , Gastrosquisis , Enfermedades del Recién Nacido , Recién Nacido , Humanos , Intestinos , Trasplante de Células Madre/métodos , Enterocolitis Necrotizante/terapia
8.
Nat Commun ; 13(1): 6031, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36229614

RESUMEN

The delivery of encapsulated islets or stem cell-derived insulin-producing cells (i.e., bioartificial pancreas devices) may achieve a functional cure for type 1 diabetes, but their efficacy is limited by mass transport constraints. Modeling such constraints is thus desirable, but previous efforts invoke simplifications which limit the utility of their insights. Herein, we present a computational platform for investigating the therapeutic capacity of generic and user-programmable bioartificial pancreas devices, which accounts for highly influential stochastic properties including the size distribution and random localization of the cells. We first apply the platform in a study which finds that endogenous islet size distribution variance significantly influences device potency. Then we pursue optimizations, determining ideal device structures and estimates of the curative cell dose. Finally, we propose a new, device-specific islet equivalence conversion table, and develop a surrogate machine learning model, hosted on a web application, to rapidly produce these coefficients for user-defined devices.


Asunto(s)
Diabetes Mellitus Tipo 1 , Insulinas , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Diabetes Mellitus Tipo 1/terapia , Humanos , Insulina , Páncreas
9.
Nat Commun ; 13(1): 3821, 2022 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-35780226

RESUMEN

Metabolic reprogramming of the tumor microenvironment (TME) and poor immunogenicity are two of the challenges that cancer immunotherapies have to overcome for improved clinical benefits. Among various immunosuppressive metabolites that keep anti-tumor immunity in check, the tryptophan catabolite kynurenine (Kyn) is an attractive target for blockade given its role in mediating immunosuppression through multiple pathways. Here, we present a local chemo-immunometabolic therapy through injection of a supramolecular hydrogel concurrently releasing doxorubicin that induces immunogenic tumor cell death and kynureninase that disrupts Kyn-mediated immunosuppressive pathways in TME. The combination synergically enhances tumor immunogenicity and unleashes anti-tumor immunity. In mouse models of triple negative breast cancer and melanoma, a single low dose peritumoral injection of the therapeutic hydrogel promotes TME transformation toward more immunostimulatory, which leads to enhanced tumor suppression and extended mouse survival. In addition, the systemic anti-tumor surveillance induced by the local treatment exhibits an abscopal effect and prevents tumor relapse post-resection. This versatile approach for local chemo-immunometabolic therapy may serve as a general strategy for enhancing anti-tumor immunity and boosting the efficacy of cancer immunotherapies.


Asunto(s)
Hidrogeles , Recurrencia Local de Neoplasia , Animales , Doxorrubicina/farmacología , Hidrogeles/farmacología , Inmunoterapia , Quinurenina/metabolismo , Ratones , Microambiente Tumoral
10.
Adv Healthc Mater ; 11(19): e2200922, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35894816

RESUMEN

Implanted cell-containing grafts require a robust and functional vasculature to supply oxygen and nutrients, as well as clear metabolic waste products. However, it remains challenging to fabricate tunable, vascular-promoting scaffolds without incorporating additional biologics. Here, a biphasic gel consisting of a highly porous aerogel and a degradable fibrin hydrogel for inducing vascularization is presented. The highly porous (>90%) and stable aerogel is assembled from short microfibers by being dispersed in an aqueous solution that can be 3D printed into various configurations. The biphasic gel demonstrates good compression-resistance: 70.30% Young's modulus is recovered over 20 cycles of 65% compression under water. Furthermore, it is confirmed that tissue cells and blood vessels can penetrate a thick (≈3 mm) biphasic gel in the subcutaneous space of mice. Finally, the biphasic gel doubles the vascular ingrowth compared to a composite of a commercial surgical polyester felt and a fibrin hydrogel upon subcutaneous implantation in mice after 4 weeks. The design of this biphasic gel may advance the development of vascularized scaffolds.


Asunto(s)
Productos Biológicos , Hidrogeles , Neovascularización Fisiológica , Andamios del Tejido , Animales , Fibrina , Hidrogeles/farmacología , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Oxígeno , Poliésteres , Ingeniería de Tejidos , Residuos
11.
Sci Adv ; 8(29): eabn0071, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35867788

RESUMEN

Islet transplantation has been established as a viable treatment modality for type 1 diabetes. However, the side effects of the systemic immunosuppression required for patients often outweigh its benefits. Here, we engineer programmed death ligand-1 and cytotoxic T lymphocyte antigen 4 immunoglobulin fusion protein-modified mesenchymal stromal cells (MSCs) as accessory cells for islet cotransplantation. The engineered MSCs (eMSCs) improved the outcome of both syngeneic and allogeneic islet transplantation in diabetic mice and resulted in allograft survival for up to 100 days without any systemic immunosuppression. Immunophenotyping revealed reduced infiltration of CD4+ or CD8+ T effector cells and increased infiltration of T regulatory cells within the allografts cotransplanted with eMSCs compared to controls. The results suggest that the eMSCs can induce local immunomodulation and may be applicable in clinical islet transplantation to reduce or minimize the need of systemic immunosuppression and ameliorate its negative impact.


Asunto(s)
Diabetes Mellitus Experimental , Trasplante de Células Madre Hematopoyéticas , Trasplante de Islotes Pancreáticos , Animales , Diabetes Mellitus Experimental/terapia , Inmunomodulación , Terapia de Inmunosupresión , Trasplante de Islotes Pancreáticos/métodos , Ratones , Ratones Endogámicos BALB C
12.
Bioact Mater ; 14: 52-60, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35310345

RESUMEN

Adhesive hydrogels have been recently proposed as a potential option to seal and treat gastric perforation (GP) which causes high mortality despite advancements in surgical treatments. However, to be effective, the hydrogels must have sufficient tissue adhesiveness, tough mechanical property, tunable biodegradability and ideally are easy to apply and form. Herein, we report an adhesive and resilient hydrogel for the sealing and treatment of gastric perforation. The hydrogel consists of a bioactive, transglutaminase (TG)-crosslinked gelatin network and a dynamic, borate-crosslinked poly-N-[Tris(hydroxymethyl)methyl]acrylamide (PTH) network. The hydrogel can be formed in situ, facilitating easy delivery to the GP and allowing for precise sealing of the defects. In vivo experiments, using a perforated stomach mouse model, shows that the adhesive hydrogel plug effectively seals GP defects and promotes gastric mucosa regeneration. Overall, this hydrogel represents a promising biomaterial for GP treatment.

13.
Adv Healthc Mater ; 11(2): e2101714, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34755476

RESUMEN

The receptor binding domain (RBD) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein that mediates viral entry into host cells is a good candidate immunogen for vaccine development against coronavirus disease 2019 (COVID-19). Because of its small size, most preclinical and early clinical efforts have focused on multimerizing RBD on various formats of nanoparticles to increase its immunogenicity. Using an easily administered injectable hydrogel scaffold that is rationally designed for enhanced retainment of RBD, an alternative and facile approach for boosting RBD immunogenicity in mice is demonstrated. Prolonged delivery of poly (I:C) adjuvanted RBD by the hydrogel scaffold results in sustained exposure to lymphoid tissues, which elicits serum IgG titers comparable to those induced by three bolus injections, but more long-lasting and polarized toward TH 1-mediated IgG2b. The hydrogel scaffold induces potent germinal center (GC) reactions, correlating with RBD-specific antibody generation and robust type 1 T cell responses. Besides being an enduring RBD reservoir, the hydrogel scaffold becomes a local inflammatory niche for innate immune cell activation. Collectively, the injectable hydrogel scaffold provides a simple, practical, and inexpensive means to enhance the efficacy of RBD-based subunit vaccines against COVID-19 and may be applicable to other circulating and emerging pathogens.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos Antivirales , Vacunas contra la COVID-19 , Humanos , Hidrogeles , Ratones , SARS-CoV-2 , Desarrollo de Vacunas , Vacunas de Subunidad
14.
Small ; 18(8): e2104899, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34897997

RESUMEN

Encapsulation and transplantation of insulin-producing cells offer a promising curative treatment for type 1 diabetes (T1D) without immunosuppression. However, biomaterials used to encapsulate cells often elicit foreign body responses, leading to cellular overgrowth and deposition of fibrotic tissue, which in turn diminishes mass transfer to and from transplanted cells. Meanwhile, the encapsulation device must be safe, scalable, and ideally retrievable to meet clinical requirements. Here, a durable and safe nanofibrous device coated with a thin and uniform, fibrosis-mitigating, zwitterionically modified alginate hydrogel for encapsulation of islets and stem cell-derived beta (SC-ß) cells is reported. The device with a configuration that has cells encapsulated within the cylindrical wall, allowing scale-up in both radial and longitudinal directions without sacrificing mass transfer, is designed. Due to its facile mass transfer and low level of fibrotic reactions, the device supports long-term cell engraftment, correcting diabetes in C57BL6/J mice with rat islets for up to 399 days and SCID-beige mice with human SC-ß cells for up to 238 days. The scalability and retrievability in dogs are further demonstrated. These results suggest the potential of this new device for cell therapies to treat T1D and other diseases.


Asunto(s)
Diabetes Mellitus Experimental , Insulinas , Trasplante de Islotes Pancreáticos , Animales , Diabetes Mellitus Experimental/terapia , Perros , Fibrosis , Trasplante de Islotes Pancreáticos/métodos , Ratones , Ratones SCID , Ratas
15.
Nat Commun ; 12(1): 5846, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34615868

RESUMEN

Inadequate oxygenation is a major challenge in cell encapsulation, a therapy which holds potential to treat many diseases including type I diabetes. In such systems, cellular oxygen (O2) delivery is limited to slow passive diffusion from transplantation sites through the poorly O2-soluble encapsulating matrix, usually a hydrogel. This constrains the maximum permitted distance between the encapsulated cells and host site to within a few hundred micrometers to ensure cellular function. Inspired by the natural gas-phase tracheal O2 delivery system of insects, we present herein the design of a biomimetic scaffold featuring internal continuous air channels endowed with 10,000-fold higher O2 diffusivity than hydrogels. We incorporate the scaffold into a bulk hydrogel containing cells, which facilitates rapid O2 transport through the whole system to cells several millimeters away from the device-host boundary. A computational model, validated by in vitro analysis, predicts that cells and islets maintain high viability even in a thick (6.6 mm) device. Finally, the therapeutic potential of the device is demonstrated through the correction of diabetes in immunocompetent mice using rat islets for over 6 months.


Asunto(s)
Oxígeno/química , Animales , Biomimética , Encapsulación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Hidrogeles/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
16.
Adv Funct Mater ; 31(47): 2103477, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34512227

RESUMEN

SARS-CoV-2, the virus that caused the COVID-19 pandemic, can remain viable and infectious on surfaces for days, posing a potential risk for fomite transmission. Liquid-based disinfectants, such as chlorine-based ones, have played an indispensable role in decontaminating surfaces but they do not provide prolonged protection from recontamination. Here a safe, inexpensive, and scalable membrane with covalently immobilized chlorine, large surface area, and fast wetting that exhibits long-lasting, exceptional killing efficacy against a broad spectrum of bacteria and viruses is reported. The membrane achieves a more than 6 log reduction within several minutes against all five bacterial strains tested, including gram-positive, gram-negative, and drug-resistant ones as well as a clinical bacterial cocktail. The membrane also efficiently deactivated nonenveloped and enveloped viruses in minutes. In particular, a 5.17 log reduction is achieved against SARS-CoV-2 after only 10 min of contact with the membrane. This membrane may be used on high-touch surfaces in healthcare and other public facilities or in air filters and personal protective equipment to provide continuous protection and minimize transmission risks.

17.
Adv Mater ; 33(39): e2102852, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34363254

RESUMEN

Encapsulation of insulin-producing cells is a promising strategy for treatment of type 1 diabetes. However, engineering an encapsulation device that is both safe (i.e., no cell escape and no breakage) and functional (i.e., low foreign-body response (FBR) and high mass transfer) remains a challenge. Here, a family of zwitterionic polyurethanes (ZPU) with sulfobetaine groups in the polymer backbone is developed, which are fabricated into encapsulation devices with tunable nanoporous structures via electrospinning. The ZPU encapsulation device is hydrophilic and fouling-resistant, exhibits robust mechanical properties, and prevents cell escape while still allowing efficient mass transfer. The ZPU device also induces a much lower FBR or cellular overgrowth upon intraperitoneal implantation in C57BL/6 mice for up to 6 months compared to devices made of similar polyurethane without the zwitterionic modification. The therapeutic potential of the ZPU device is shown for islet encapsulation and diabetes correction in mice for ≈3 months is demonstrated. As a proof of concept, the scalability and retrievability of the ZPU device in pigs and dogs are further demonstrated. Collectively, these attributes make ZPU devices attractive candidates for cell encapsulation therapies.


Asunto(s)
Materiales Biocompatibles/química , Islotes Pancreáticos/química , Nanoporos , Poliuretanos/química , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Diabetes Mellitus Experimental/terapia , Perros , Interacciones Hidrofóbicas e Hidrofílicas , Islotes Pancreáticos/fisiología , Trasplante de Islotes Pancreáticos/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Porcinos
18.
APL Bioeng ; 5(3): 031503, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34286170

RESUMEN

Diabetic wounds often have a slow healing process and become easily infected owing to hyperglycemia in wound beds. Once planktonic bacterial cells develop into biofilms, the diabetic wound becomes more resistant to treatment. Although it remains challenging to accelerate healing in a diabetic wound due to complex pathology, including bacterial infection, high reactive oxygen species, chronic inflammation, and impaired angiogenesis, the development of multifunctional hydrogels is a promising strategy. Multiple functions, including antibacterial, pro-angiogenesis, and overall pro-healing, are high priorities. Here, design strategies, mechanisms of action, performance, and application of functional hydrogels are systematically discussed. The unique properties of hydrogels, including bactericidal and wound healing promotive effects, are reviewed. Considering the clinical need, stimuli-responsive and multifunctional hydrogels that can accelerate diabetic wound healing are likely to form an important part of future diabetic wound management.

19.
Adv Sci (Weinh) ; 8(17): e2003708, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34258870

RESUMEN

Islet transplantation has shown promise as a curative therapy for type 1 diabetes (T1D). However, the side effects of systemic immunosuppression and limited long-term viability of engrafted islets, together with the scarcity of donor organs, highlight an urgent need for the development of new, improved, and safer cell-replacement strategies. Induction of local immunotolerance to prevent allo-rejection against islets and stem cell derived ß cells has the potential to improve graft function and broaden the applicability of cellular therapy while minimizing adverse effects of systemic immunosuppression. In this mini review, recent developments in non-encapsulation, local immunomodulatory approaches for T1D cell replacement therapies, including islet/ß cell modification, immunomodulatory biomaterial platforms, and co-transplantation of immunomodulatory cells are discussed. Key advantages and remaining challenges in translating such technologies to clinical settings are identified. Although many of the studies discussed are preliminary, the growing interest in the field has led to the exploration of new combinatorial strategies involving cellular engineering, immunotherapy, and novel biomaterials. Such interdisciplinary research will undoubtedly accelerate the development of therapies that can benefit the whole T1D population.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/cirugía , Rechazo de Injerto/prevención & control , Inmunomodulación/inmunología , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos/métodos , Rechazo de Injerto/inmunología , Humanos
20.
Sci Transl Med ; 13(596)2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078744

RESUMEN

Transplantation of stem cell-derived ß (SC-ß) cells represents a promising therapy for type 1 diabetes (T1D). However, the delivery, maintenance, and retrieval of these cells remain a challenge. Here, we report the design of a safe and functional device composed of a highly porous, durable nanofibrous skin and an immunoprotective hydrogel core. The device consists of electrospun medical-grade thermoplastic silicone-polycarbonate-urethane and is soft but tough (~15 megapascal at a rupture strain of >2). Tuning the nanofiber size to less than ~500 nanometers prevented cell penetration while maintaining maximum mass transfer and decreased cellular overgrowth on blank (cell-free) devices to as low as a single-cell layer (~3 micrometers thick) when implanted in the peritoneal cavity of mice. We confirmed device safety, indicated as continuous containment of proliferative cells within the device for 5 months. Encapsulating syngeneic, allogeneic, or xenogeneic rodent islets within the device corrected chemically induced diabetes in mice and cells remained functional for up to 200 days. The function of human SC-ß cells was supported by the device, and it reversed diabetes within 1 week of implantation in immunodeficient and immunocompetent mice, for up to 120 and 60 days, respectively. We demonstrated the scalability and retrievability of the device in dogs and observed viable human SC-ß cells despite xenogeneic immune responses. The nanofibrous device design may therefore provide a translatable solution to the balance between safety and functionality in developing stem cell-based therapies for T1D.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Insulinas , Trasplante de Islotes Pancreáticos , Nanofibras , Animales , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Perros , Insulina , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...