Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Am J Obstet Gynecol ; 2024 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-39191364

RESUMEN

OBJECTIVES: Vaginal childbirth is a key risk factor for pelvic floor muscle injury and dysfunction, and subsequent pelvic floor disorders. Multiparity further exacerbates these risks. Using the rat model, validated for the studies of the human pelvic floor muscles, we have previously identified that a single simulated birth injury results in pelvic floor muscle atrophy and fibrosis. We hypothesized that multiple birth injuries would further overwhelm the muscle regenerative capacity, leading to functionally relevant pathological alterations long-term. STUDY DESIGN: Sprague-Dawley rats underwent simulated birth injury and were allowed to recover for 8 weeks before undergoing additional birth injury. Animals were sacrificed at acute (3- and 7-days post injury), subacute (21-, 28-, and 35-days post-injury), and long-term (8- and 12-weeks post-injury) time points post-second injury (N=3-8/time point), and the pubocaudalis portion of the rat levator ani complex was harvested to assess the impact of repeated birth injuries on muscle mechanical and histomorphological properties. The accompanying transcriptional changes were assessed by a customized NanoString panel. Uninjured animals were used as controls. Data with a parametric distribution were analyzed by a two-way analysis of variance followed by post hoc pairwise comparisons using Tukey's or Sidak's tests; non-parametrically distributed data were compared with Kruskal-Wallis test followed by pairwise comparisons with Dunn's test. Data, analyzed using GraphPad Prism v8.0, San Diego, CA, are presented as mean ± SEM or median (range). RESULTS: Following the 1st simulated birth injury, active muscle force decreased acutely relative to uninjured controls (12.9±0.9 vs 25.98±2.1 g/mm2, P<0.01). At 4 weeks, muscle active force production recovered to baseline and remained unchanged at 8 weeks after birth injury (P>0.99). Similarly, precipitous decrease in active force was observed immediately after repeated birth injury (18.07±1.2 vs 25.98±2.1 g/mm2, P<0.05). In contrast to the functional recovery after a single birth injury, a long-term decrease in muscle contractile function was observed up to 12 weeks after repeated birth injuries (18.3±1.6 vs 25.98±2.1 g/mm2, P<0.05). Fiber size was smaller at the long-term time points after 2nd injury compared to the uninjured group (12 weeks vs uninjured control: 1485 (60.7-5000) vs 1989 (65.6-4702) µm2, P<0.0001). The proportion of fibers with centralized nuclei, indicating active myofiber regeneration, returned to baseline at 8 weeks post-1st birth injury, (P=0.95), but remained elevated as far as 12 weeks post-2nd injury (12 weeks vs uninjured control: 7.1±1.5 vs 0.84±0.13%, P<0.0001). In contrast to the plateauing intramuscular collagen content after 4 weeks post-1st injury, fibrotic degeneration increased progressively over 12 weeks after repeated injury (12 weeks vs uninjured control: 6. 7±1.1 vs 2.03±0.2%, P<0.001). Prolonged expression of pro-inflammatory genes accompanied by a greater immune infiltrate was observed after repeated compared to a single birth injury. CONCLUSIONS: Overall, repeated birth injuries led to a greater magnitude of pathological alterations compared to a single injury, resulting in more pronounced pelvic floor muscle degeneration and muscle dysfunction in the rat model. The above provides a putative mechanistic link between multiparity and the increased risk of pelvic floor dysfunction in women.

2.
Bioact Mater ; 38: 528-539, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38803824

RESUMEN

While oropharyngeal cancer treatment regimens, including surgical resection, irradiation, and chemotherapy, are effective at removing tumors, they lead to muscle atrophy, denervation, and fibrosis, contributing to the pathogenesis of oropharyngeal dysphagia - difficulty swallowing. Current standard of care of rehabilitative tongue strengthening and swallowing exercises is ineffective. Here, we evaluate an alternative approach utilizing an acellular and injectable biomaterial to preserve muscle content and reduce fibrosis of the tongue after injury. Skeletal muscle extracellular matrix (SKM) hydrogel is fabricated from decellularized porcine skeletal muscle tissue. A partial glossectomy injury in the rat is used to induce tongue fibrosis, and SKM hydrogels along with saline controls are injected into the site of scarring two weeks after injury. Tissues are harvested at 3 and 7 days post-injection for gene expression and immunohistochemical analyses, and at 4 weeks post-injection to evaluate histomorphological properties. SKM hydrogel reduces scar formation and improves muscle regeneration at the site of injury compared to saline. SKM additionally modulates the immune response towards an anti-inflammatory phenotype. This study demonstrates the immunomodulatory and tissue-regenerative capacity of an acellular and minimally invasive ECM hydrogel in a rodent model of tongue injury.

3.
JACC Basic Transl Sci ; 9(3): 322-338, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38559631

RESUMEN

This study evaluates the effectiveness of myocardial matrix (MM) hydrogels in mitigating negative right ventricular (RV) remodeling in a rat model of RV heart failure. The goal was to assess whether a hydrogel derived from either the right or left ventricle could promote cardiac repair. Injured rat right ventricles were injected with either RV-or left ventricular-derived MM hydrogels. Both hydrogels improved RV function and morphology and reduced negative remodeling. This study supports the potential of injectable biomaterial therapies for treating RV heart failure.

4.
Urogynecology (Phila) ; 30(5): 519-527, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38683203

RESUMEN

ABSTRACT: Pelvic floor disorders (PFDs) constitute a major public health issue given their negative effect on quality of life for millions of women worldwide and the associated economic burden. As the prevalence of PFDs continues to increase, novel therapeutic approaches for the effective treatment of these disorders are urgently needed. Regenerative medicine techniques, including cellular therapies, extracellular vesicles, secretomes, platelet-rich plasma, laser therapy, and bioinductive acellular biomaterial scaffolds, are emerging as viable clinical options to counteract urinary and fecal incontinence, as well as pelvic organ prolapse. This brief expert review explores the current state-of-science regarding application of these therapies for the treatment of PFDs. Although regenerative approaches have not been widely deployed in clinical care to date, these innovative techniques show a promising safety profile and potential to positively affect the quality of life of patients with PFDs. Furthermore, investigations focused on regeneration of the main constituents of the pelvic floor and lower urinary tract improve our understanding of the underlying pathophysiology of PFDs. Regenerative medicine techniques have a high potential not only to revolutionize treatment of PFDs but also to prevent these complex conditions.


Asunto(s)
Trastornos del Suelo Pélvico , Medicina Regenerativa , Humanos , Medicina Regenerativa/métodos , Femenino , Trastornos del Suelo Pélvico/terapia , Calidad de Vida , Ginecología/métodos
5.
Biofabrication ; 16(2)2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38128127

RESUMEN

Insulin is an essential regulator of blood glucose homeostasis that is produced exclusively byßcells within the pancreatic islets of healthy individuals. In those affected by diabetes, immune inflammation, damage, and destruction of isletßcells leads to insulin deficiency and hyperglycemia. Current efforts to understand the mechanisms underlyingßcell damage in diabetes rely onin vitro-cultured cadaveric islets. However, isolation of these islets involves removal of crucial matrix and vasculature that supports islets in the intact pancreas. Unsurprisingly, these islets demonstrate reduced functionality over time in standard culture conditions, thereby limiting their value for understanding native islet biology. Leveraging a novel, vascularized micro-organ (VMO) approach, we have recapitulated elements of the native pancreas by incorporating isolated human islets within a three-dimensional matrix nourished by living, perfusable blood vessels. Importantly, these islets show long-term viability and maintain robust glucose-stimulated insulin responses. Furthermore, vessel-mediated delivery of immune cells to these tissues provides a model to assess islet-immune cell interactions and subsequent islet killing-key steps in type 1 diabetes pathogenesis. Together, these results establish the islet-VMO as a novel,ex vivoplatform for studying human islet biology in both health and disease.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Humanos , Insulina/metabolismo , Diabetes Mellitus/metabolismo , Glucosa/metabolismo
6.
NPJ Regen Med ; 8(1): 53, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37730736

RESUMEN

Bioactive immunomodulatory biomaterials have shown promise for influencing the immune response to promote tissue repair and regeneration. Macrophages and T cells have been associated with this response; however, other immune cell types have been traditionally overlooked. In this study, we investigated the role of mast cells in the regulation of the immune response to decellularized biomaterial scaffolds using a subcutaneous implant model. In mast cell-deficient mice, there was dysregulation of the expected M1 to M2 macrophage transition typically induced by the biomaterial scaffold. Polarization progression deviated in a sex-specific manner with an early transition to an M2 profile in female mice, while the male response was unable to properly transition past a pro-inflammatory M1 state. Both were reversed with adoptive mast cell transfer. Further investigation of the later-stage immune response in male mice determined a greater sustained pro-inflammatory gene expression profile, including the IL-1 cytokine family, IL-6, alarmins, and chemokines. These results highlight mast cells as another important cell type that influences the immune response to pro-regenerative biomaterials.

7.
Biomacromolecules ; 24(11): 4695-4704, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37695847

RESUMEN

Herein, we have developed a drug-loaded matrix metalloproteinase (MMP)-responsive micellar nanoparticle (NP) intended for minimally invasive intravenous injection during the acute phase of myocardial infarction (MI) and prolonged retention in the heart for small-molecule drug delivery. Peptide-polymer amphiphiles (PPAs) bearing a small-molecule MMP inhibitor (MMPi), PD166793, were synthesized via ring-opening metathesis polymerization (ROMP) and formulated into spherical micelles by transitioning to aqueous solution. The resulting micellar NPs underwent MMP-induced aggregation, demonstrating enzyme responsiveness. Using a rat MI model, we observed that these NPs were capable of successfully extravasating into the infarcted region of the heart where they were retained due to the active, enzyme-mediated targeting, remaining detectable after 1 week post administration without increasing macrophage recruitment. Furthermore, in vitro studies show that these NPs demonstrated successful drug release following MMP treatment and maintained drug bioactivity as evidenced by comparable MMP inhibition to free MMPi. This work establishes a targeted NP platform for delivering small-molecule therapeutics to the heart after MI, opening possibilities for myocardial infarction treatment.


Asunto(s)
Infarto del Miocardio , Nanopartículas , Ratas , Animales , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Péptidos/uso terapéutico , Micelas
10.
Sci Transl Med ; 15(707): eabj3138, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37531414

RESUMEN

Pelvic floor disorders, including pelvic organ prolapse and urinary and fecal incontinence, affect millions of women globally and represent a major public health concern. Pelvic floor muscle (PFM) dysfunction has been identified as one of the leading risk factors for the development of these morbid conditions. Childbirth, specifically vaginal delivery, has been recognized as the most important potentially modifiable risk factor for PFM injury; however, the precise mechanisms of PFM dysfunction after parturition remain elusive. In this study, we demonstrated that PFMs exhibit atrophy and fibrosis in parous women with symptomatic pelvic organ prolapse. These pathological alterations were recapitulated in a preclinical rat model of simulated birth injury (SBI). The transcriptional signature of PFMs after injury demonstrated an impairment in muscle anabolism, persistent expression of genes that promote extracellular matrix (ECM) deposition, and a sustained inflammatory response. We also evaluated the administration of acellular injectable skeletal muscle ECM hydrogel for the prevention of these pathological alterations. Treatment of PFMs with the ECM hydrogel either at the time of birth injury or 4 weeks after injury mitigated PFM atrophy and fibrosis. By evaluating gene expression, we demonstrated that these changes are mainly driven by the hydrogel-induced enhancement of endogenous myogenesis, ECM remodeling, and modulation of the immune response. This work furthers our understanding of PFM birth injury and demonstrates proof of concept for future investigations of proregenerative biomaterial approaches for the treatment of injured pelvic soft tissues.


Asunto(s)
Traumatismos del Nacimiento , Prolapso de Órgano Pélvico , Embarazo , Femenino , Ratas , Animales , Hidrogeles , Diafragma Pélvico/fisiología , Parto , Músculo Esquelético , Traumatismos del Nacimiento/complicaciones , Fibrosis , Prolapso de Órgano Pélvico/etiología , Matriz Extracelular
11.
Adv Healthc Mater ; 12(25): e2300782, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37390094

RESUMEN

Traumatic brain injury (TBI) affects millions of people each year and, in many cases, results in long-term disabilities. Once a TBI has occurred, there is a significant breakdown of the blood-brain barrier resulting in increased vascular permeability and progression of the injury. In this study, the use of an infusible extracellular matrix-derived biomaterial (iECM) for its ability to reduce vascular permeability and modulate gene expression in the injured brain is investigated. First, the pharmacokinetics of iECM administration in a mouse model of TBI is characterized, and the robust accumulation of iECM at the site of injury is demonstrated. Next, it is shown that iECM administration after injury can reduce the extravasation of molecules into the brain, and in vitro, iECM increases trans-endothelial electrical resistance across a monolayer of TNFα-stimulated endothelial cells. In gene expression analysis of brain tissue, iECM induces changes that are indicative of downregulation of the proinflammatory response 1-day post-injury/treatment and neuroprotection at 5 days post-injury/treatment. Therefore, iECM shows potential as a treatment for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Humanos , Ratones , Animales , Células Endoteliales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad
12.
J Am Chem Soc ; 145(20): 11185-11194, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37184379

RESUMEN

Nanoparticles that undergo a localized morphology change to target areas of inflammation have been previously developed but are limited by their lack of biodegradability. In this paper, we describe a low-ring-strain cyclic olefin monomer, 1,3-dimethyl-2-phenoxy-1,3,4,7-tetrahydro-1,3,2-diazaphosphepine 2-oxide (MePTDO), that rapidly polymerizes via ring-opening metathesis polymerization at room temperature to generate well-defined degradable polyphosphoramidates with high monomer conversion (>84%). Efficient MePTDO copolymerizations with norbornene-based monomers are demonstrated, including a norbornenyl monomer functionalized with a peptide substrate for inflammation-associated matrix metalloproteinases (MMPs). The resulting amphiphilic peptide brush copolymers self-assembled in aqueous solution to generate micellar nanoparticles (30 nm in diameter) which exhibit excellent cyto- and hemocompatibility and undergo MMP-induced assembly into micron-scale aggregates. As MMPs are upregulated in the heart postmyocardial infarction (MI), the MMP-responsive micelles were applied to target and accumulate in the infarcted heart following intravenous administration in a rat model of MI. These particles displayed a distinct biodistribution and clearance pattern in comparison to nondegradable analogues. Specifically, accumulation at the site of MI competed with elimination predominantly through the kidney rather than the liver. Together, these results suggest this as a promising new biodegradable platform for inflammation targeted delivery.


Asunto(s)
Infarto del Miocardio , Nanopartículas , Ratas , Animales , Micelas , Distribución Tisular , Péptidos , Inflamación , Metaloproteinasas de la Matriz
13.
Adv Mater ; : e2300603, 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-36989469

RESUMEN

Each year, nearly 19 million people die of cardiovascular disease with coronary heart disease and myocardial infarction (MI) as the leading cause of the progression of heart failure. Due to the high risk associated with surgical procedures, a variety of minimally invasive therapeutics aimed at tissue repair and regeneration are being developed. While biomaterials delivered via intramyocardial injection have shown promise, there are challenges associated with delivery in acute MI. In contrast, intravascularly injectable biomaterials are a desirable category of therapeutics due to their ability to be delivered immediately post-MI via less invasive methods. In addition to passive diffusion into the infarct, these biomaterials can be designed to target the molecular and cellular characteristics seen in MI pathophysiology, such as cells and proteins present in the ischemic myocardium, to reduce off-target localization. These injectable materials can also be stimuli-responsive through enzymes or chemical imbalances. This review outlines the natural and synthetic biomaterial designs that allow for retention and accumulation within the infarct via intravascular delivery, including intracoronary infusion and intravenous injection.

14.
Nat Biomed Eng ; 7(2): 94-109, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36581694

RESUMEN

Decellularized extracellular matrix in the form of patches and locally injected hydrogels has long been used as therapies in animal models of disease. Here we report the safety and feasibility of an intravascularly infused extracellular matrix as a biomaterial for the repair of tissue in animal models of acute myocardial infarction, traumatic brain injury and pulmonary arterial hypertension. The biomaterial consists of decellularized, enzymatically digested and fractionated ventricular myocardium, localizes to injured tissues by binding to leaky microvasculature, and is largely degraded in about 3 d. In rats and pigs with induced acute myocardial infarction followed by intracoronary infusion of the biomaterial, we observed substantially reduced left ventricular volumes and improved wall-motion scores, as well as differential expression of genes associated with tissue repair and inflammation. Delivering pro-healing extracellular matrix by intravascular infusion post injury may provide translational advantages for the healing of inflamed tissues 'from the inside out'.


Asunto(s)
Materiales Biocompatibles , Infarto del Miocardio , Ratas , Porcinos , Animales , Miocardio/metabolismo , Infarto del Miocardio/terapia , Hidrogeles , Matriz Extracelular/metabolismo
15.
NPJ Regen Med ; 7(1): 72, 2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36526635

RESUMEN

Pelvic floor muscle (PFM) injury during childbirth is a key risk factor for pelvic floor disorders that affect millions of women worldwide. Muscle stem cells (MuSCs), supported by the fibro-adipogenic progenitors (FAPs) and immune cells, are indispensable for the regeneration of injured appendicular skeletal muscles. However, almost nothing is known about their role in PFM regeneration following birth injury. To elucidate the role of MuSCs, FAPs, and immune infiltrate in this context, we used radiation to perturb cell function and followed PFM recovery in a validated simulated birth injury (SBI) rat model. Non-irradiated and irradiated rats were euthanized at 3,7,10, and 28 days post-SBI (dpi). Twenty-eight dpi, PFM fiber cross-sectional area (CSA) was significantly lower and the extracellular space occupied by immune infiltrate was larger in irradiated relative to nonirradiated injured animals. Following SBI in non-irradiated animals, MuSCs and FAPs expanded significantly at 7 and 3 dpi, respectively; this expansion did not occur in irradiated animals at the same time points. At 7 and 10 dpi, we observed persistent immune response in PFMs subjected to irradiation compared to non-irradiated injured PFMs. CSA of newly regenerated fibers was also significantly smaller following SBI in irradiated compared to non-irradiated injured PFMs. Our results demonstrate that the loss of function and decreased expansion of MuSCs and FAPs after birth injury lead to impaired PFM recovery. These findings form the basis for further studies focused on the identification of novel therapeutic targets to counteract postpartum PFM dysfunction and the associated pelvic floor disorders.

16.
Acta Biomater ; 152: 47-59, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36041648

RESUMEN

As the native regenerative potential of adult cardiac tissue is limited post-injury, stimulating endogenous repair mechanisms in the mammalian myocardium is a potential goal of regenerative medicine therapeutics. Injection of myocardial matrix hydrogels into the heart post-myocardial infarction (MI) has demonstrated increased cardiac muscle and promotion of pathways associated with cardiac development, suggesting potential promotion of cardiomyocyte turnover. In this study, the myocardial matrix hydrogel was shown to have native capability as an effective reactive oxygen species scavenger and protect against oxidative stress induced cell cycle inhibition in vitro. Encapsulation of cardiomyocytes demonstrated an enhanced turnover in in vitro studies, and in vivo assessments of myocardial matrix hydrogel treatment post-MI showed increased thymidine analog uptake in cardiomyocyte nuclei compared to saline controls. Overall, this study provides evidence that properties of the myocardial matrix material provide a microenvironment mitigating oxidative damage and supportive of cardiomyocytes undergoing DNA synthesis, toward possible DNA repair or cell cycle activation. STATEMENT OF SIGNIFICANCE: Loss of adult mammalian cardiomyocyte turnover is influenced by shifts in oxidative damage, which represents a potential mechanism for improving restoration of cardiac muscle after myocardial infarction (MI). Injection of a myocardial matrix hydrogel into the heart post-MI previously demonstrated increased cardiac muscle and promotion of pathways associated with cardiac development, suggesting potential in promoting proliferation of cardiomyocytes. In this study, the myocardial matrix hydrogel was shown to protect cells from oxidative stress and increase proliferation in vitro. In a rat MI model, greater presence of tissue free thiol content spared from oxidative damage, lesser mitochondrial superoxide content, and increased thymidine analog uptake in cardiomyocytes was found in matrix injected animals compared to saline controls. Overall, this study provides evidence that properties of the myocardial matrix material provide a microenvironment supportive of cardiomyocytes undergoing DNA synthesis, toward possible DNA repair or cell cycle activation.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Animales , ADN/metabolismo , Hidrogeles/metabolismo , Hidrogeles/farmacología , Mamíferos , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/farmacología , Superóxidos , Timidina/metabolismo , Timidina/farmacología
17.
J Mol Cell Cardiol ; 171: 45-55, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35780862

RESUMEN

Congenital heart defects are the leading cause of right heart failure in pediatric patients. Implantation of c-kit+ cardiac-derived progenitor cells (CPCs) is being clinically evaluated to treat the failing right ventricle (RV), but faces limitations due to reduced transplant cell survival, low engraftment rates, and low retention. These limitations have been exacerbated due to the nature of cell delivery (narrow needles) and the non-optimal recipient microenvironment (reactive oxygen species (ROS)). Extracellular matrix (ECM) hydrogels derived from porcine left ventricular (LV) myocardium have emerged as a potential therapy to treat the ischemic LV and have shown promise as a vehicle to deliver cells to injured myocardium. However, no studies have evaluated the combination of an injectable biomaterial, such as an ECM hydrogel, in combination with cell therapy for treating RV failure. In this study we characterized LV and RV myocardial matrix (MM) hydrogels and performed in vitro evaluations of their potential to enhance CPC delivery, including resistance to forces experienced during injection and exposure to ROS, as well as their potential to enhance angiogenic paracrine signaling. While physical properties of the two hydrogels are similar, the decellularized LV and RV have distinct protein signatures. Both materials were equally effective in protecting CPCs against needle forces and ROS. CPCs encapsulated in either the LV MM or RV MM exhibited similar enhanced potential for angiogenic paracrine signaling when compared to CPCs in collagen. The RV MM without cells, however, likewise improved tube formation, suggesting it should also be evaluated as a potential standalone treatment.


Asunto(s)
Insuficiencia Cardíaca , Hidrogeles , Animales , Materiales Biocompatibles/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos , Hidrogeles/metabolismo , Miocardio , Especies Reactivas de Oxígeno/metabolismo , Células Madre , Porcinos
18.
Methods Mol Biol ; 2485: 255-268, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35618911

RESUMEN

Injectable biomaterials have been developed as potential minimally invasive therapies for treating myocardial infarction (MI) and heart failure. Christman et al. first showed that the injection of a biomaterial alone into rat myocardium can improve cardiac function after MI. More recently, hydrogel forms of decellularized extracellular matrix (ECM) materials have shown substantial promise. Here, we present the methods for fabricating an injectable cardiac-specific ECM biomaterial with demonstrated positive outcomes in small and large animal models for cardiac repair as well as initial safety in a Phase I clinical trial. This chapter also covers the methods for the injection of a biomaterial into rat myocardium using a surgical approach through the diaphragm. Although the methods shown here are for injection of an acellular biomaterial, cells or other therapeutics could also be added to the injection for testing other regenerative medicine strategies.


Asunto(s)
Infarto del Miocardio , Miocardio , Animales , Materiales Biocompatibles/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Corazón , Hidrogeles , Ratas
19.
Biomater Sci ; 10(2): 444-456, 2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-34878443

RESUMEN

Pediatric patients with congenital heart defects (CHD) often present with heart failure from increased load on the right ventricle (RV) due to both surgical methods to treat CHD and the disease itself. Patients with RV failure often require transplantation, which is limited due to lack of donor availability and rejection. Previous studies investigating the development and in vitro assessment of a bioprinted cardiac patch composed of cardiac extracellular matrix (cECM) and human c-kit + progenitor cells (hCPCs) showed that the construct has promise in treating cardiac dysfunction. The current study investigates in vivo cardiac outcomes of patch implantation in a rat model of RV failure. Patch parameters including cECM-inclusion and hCPC-inclusion are investigated. Assessments include hCPC retention, RV function, and tissue remodeling (vascularization, hypertrophy, and fibrosis). Animal model evaluation shows that both cell-free and neonatal hCPC-laden cECM-gelatin methacrylate (GelMA) patches improve RV function and tissue remodeling compared to other patch groups and controls. Inclusion of cECM is the most influential parameter driving therapeutic improvements, with or without cell inclusion. This study paves the way for clinical translation in treating pediatric heart failure using bioprinted GelMA-cECM and hCPC-GelMA-cECM patches.


Asunto(s)
Insuficiencia Cardíaca , Células Madre , Animales , Niño , Matriz Extracelular , Gelatina , Corazón , Humanos , Ratas
20.
Nat Commun ; 12(1): 3764, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145265

RESUMEN

Post-surgical cardiac adhesions represent a significant problem during routine cardiothoracic procedures. This fibrous tissue can impair heart function and inhibit surgical access in reoperation procedures. Here, we propose a hydrogel barrier composed of oxime crosslinked poly(ethylene glycol) (PEG) with the inclusion of a catechol (Cat) group to improve retention on the heart for pericardial adhesion prevention. This three component system is comprised of aldehyde (Ald), aminooxy (AO), and Cat functionalized PEG mixed to form the final gel (Ald-AO-Cat). Ald-AO-Cat has favorable mechanical properties, degradation kinetics, and minimal swelling, as well as superior tissue retention compared to an initial Ald-AO gel formulation. We show that the material is cytocompatible, resists cell adhesion, and led to a reduction in the severity of adhesions in an in vivo rat model. We further show feasibility in a pilot porcine study. The Ald-AO-Cat hydrogel barrier may therefore serve as a promising solution for preventing post-surgical cardiac adhesions.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Hidrogeles/química , Hidrogeles/uso terapéutico , Adherencias Tisulares/prevención & control , Aldehídos/química , Animales , Materiales Biocompatibles/química , Catecoles/química , Línea Celular , Masculino , Ratones , Oximas/química , Oximas/uso terapéutico , Polietilenglicoles/química , Ratas , Ratas Sprague-Dawley , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA