Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Biosci Bioeng ; 135(6): 493-499, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36966053

ABSTRACT

Cardiovascular disease, primarily caused by coronary artery disease, is the leading cause of death in the United States. While standard clinical interventions have improved patient outcomes, mortality rates associated with eventual heart failure still represent a clinical challenge. Macrorevascularization techniques inadequately address the microvascular perfusion deficits that persist beyond primary and secondary interventions. In this work, we investigate a photosynthetic oxygen delivery system that rescues the myocardium following acute ischemia. Using a simple microfluidic system, we encapsulated Synechococcus elongatus into alginate hydrogel microparticles (HMPs), which photosynthetically deliver oxygen to ischemic tissue in the absence of blood flow. We demonstrate that HMPs improve the viability of S. elongatus during the injection process and allow for simple oxygen diffusion. Adult male Wistar rats (n = 45) underwent sham surgery, acute ischemia reperfusion surgery, or a chronic ischemia reperfusion surgery, followed by injection of phosphate buffered saline (PBS), S. elongatus suspended in PBS, HMPs, or S. elongatus encapsulated in HMPs. Treatment with S. elongatus-HMPs mitigated cellular apoptosis and improved left ventricular function. Thus, delivery of S. elongatus encapsulated in HMPs improves clinical translation by utilizing a minimally invasive delivery platform that improves S. elongatus viability and enhances the therapeutic benefit of a novel photosynthetic system for the treatment of myocardial ischemia.


Subject(s)
Cyanobacteria , Hydrogels , Rats , Animals , Male , Microfluidics , Rats, Wistar , Myocardium , Oxygen
2.
J Mech Behav Biomed Mater ; 126: 105074, 2022 02.
Article in English | MEDLINE | ID: mdl-35030471

ABSTRACT

After myocardial infarction (MI), adult mammals exhibit scar formation, adverse left ventricular (LV) remodeling, LV stiffening, and impaired contractility, ultimately resulting in heart failure. Neonatal mammals, however, are capable of natural heart regeneration after MI. We hypothesized that neonatal cardiac regeneration conserves native biaxial LV mechanics after MI. Wistar rat neonates (1 day old, n = 46) and adults (8-10 weeks old, n = 20) underwent sham surgery or permanent left anterior descending coronary artery ligation. At 6 weeks after neonatal MI, Masson's trichrome staining revealed negligible fibrosis. Echocardiography for the neonatal MI (n = 15) and sham rats (n = 14) revealed no differences in LV wall thickness or chamber diameter, and both groups had normal ejection fraction (72.7% vs 77.5%, respectively, p = 0.1946). Biaxial tensile testing revealed similar stress-strain curves along both the circumferential and longitudinal axes across a full range of physiologic stresses and strains. The circumferential modulus (267.9 kPa vs 274.2 kPa, p = 0.7847), longitudinal modulus (269.3 kPa vs 277.1 kPa, p = 0.7435), and maximum shear stress (3.30 kPa vs 3.95 kPa, p = 0.5418) did not differ significantly between the neonatal MI and sham groups, respectively. In contrast, transmural scars were observed at 4 weeks after adult MI. Adult MI hearts (n = 7) exhibited profound LV wall thinning (p < 0.0001), chamber dilation (p = 0.0246), and LV dysfunction (ejection fraction 45.4% vs 79.7%, p < 0.0001) compared to adult sham hearts (n = 7). Adult MI hearts were significantly stiffer than adult sham hearts in both the circumferential (321.5 kPa vs 180.0 kPa, p = 0.0111) and longitudinal axes (315.4 kPa vs 172.3 kPa, p = 0.0173), and also exhibited greater maximum shear stress (14.87 kPa vs 3.23 kPa, p = 0.0162). Our study is the first to show that native biaxial LV mechanics are conserved after neonatal heart regeneration following MI, thus adding biomechanical support for the therapeutic potential of cardiac regeneration in the treatment of ischemic heart disease.


Subject(s)
Myocardial Infarction , Animals , Animals, Newborn , Biomechanical Phenomena , Cicatrix/pathology , Disease Models, Animal , Myocardial Infarction/pathology , Myocardium/pathology , Rats , Rats, Wistar , Ventricular Remodeling
3.
Chem Rev ; 121(18): 11385-11457, 2021 09 22.
Article in English | MEDLINE | ID: mdl-33938724

ABSTRACT

Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.


Subject(s)
Hydrogels , Tissue Engineering , Prostheses and Implants
4.
Clin Transl Med ; 11(4): e387, 2021 04.
Article in English | MEDLINE | ID: mdl-33931977

ABSTRACT

Understanding how automated insulin delivery (AID) algorithm features impact glucose control under full closed loop delivery represents a critical step toward reducing patient burden by eliminating the need for carbohydrate entries at mealtimes. Here, we use a pig model of diabetes to compare AndroidAPS and Loop open-source AID systems without meal announcements. Overall time-in-range (70-180 mg/dl) for AndroidAPS was 58% ± 5%, while time-in-range for Loop was 35% ± 5%. The effect of the algorithms on time-in-range differed between meals and overnight. During the overnight monitoring period, pigs had an average time-in-range of 90% ± 7% when on AndroidAPS compared to 22% ± 8% on Loop. Time-in-hypoglycemia also differed significantly during the lunch meal, whereby pigs running AndroidAPS spent an average of 1.4% (+0.4/-0.8)% in hypoglycemia compared to 10% (+3/-6)% for those using Loop. As algorithm design for closed loop systems continues to develop, the strategies employed in the OpenAPS algorithm (known as oref1) as implemented in AndroidAPS for unannounced meals may result in a better overall control for full closed loop systems.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Insulin Infusion Systems , Algorithms , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/blood , Disease Models, Animal , Female , Glycemic Control/methods , Insulin/administration & dosage , Insulin/therapeutic use , Swine
5.
Curr Opin Biotechnol ; 66: 246-254, 2020 12.
Article in English | MEDLINE | ID: mdl-33011453

ABSTRACT

Ischemic heart disease is the most common type of heart disease, responsible for roughly 10 million deaths worldwide annually. While standard clinical interventions have resulted in improved patient outcomes, access to small diameter vessels required for cardiovascular interventions, and long-term patient mortality rates associated with eventual heart failure, remain critical challenges. In this current opinion piece we discuss novel methodologies for the advancement of vascular grafts, cardiac patches, and injectable drug delivery depot technologies as they relate to treatment of ischemic heart disease, including bilayered conduits, acellular bioactive extracellular matrix (ECM) scaffolds, and protease-responsive hydrogel delivery platforms. We address the motivation for innovation and current limitations in the field of engineered biomaterials for myocardial ischemia therapeutics and interventions.


Subject(s)
Biocompatible Materials , Heart Diseases , Extracellular Matrix , Heart Diseases/surgery , Humans , Hydrogels , Tissue Engineering , Tissue Scaffolds
6.
Sci Transl Med ; 12(550)2020 07 01.
Article in English | MEDLINE | ID: mdl-32611683

ABSTRACT

Insulin has been used to treat diabetes for almost 100 years; yet, current rapid-acting insulin formulations do not have sufficiently fast pharmacokinetics to maintain tight glycemic control at mealtimes. Dissociation of the insulin hexamer, the primary association state of insulin in rapid-acting formulations, is the rate-limiting step that leads to delayed onset and extended duration of action. A formulation of insulin monomers would more closely mimic endogenous postprandial insulin secretion, but monomeric insulin is unstable in solution using present formulation strategies and rapidly aggregates into amyloid fibrils. Here, we implement high-throughput-controlled radical polymerization techniques to generate a large library of acrylamide carrier/dopant copolymer (AC/DC) excipients designed to reduce insulin aggregation. Our top-performing AC/DC excipient candidate enabled the development of an ultrafast-absorbing insulin lispro (UFAL) formulation, which remains stable under stressed aging conditions for 25 ± 1 hours compared to 5 ± 2 hours for commercial fast-acting insulin lispro formulations (Humalog). In a porcine model of insulin-deficient diabetes, UFAL exhibited peak action at 9 ± 4 min, whereas commercial Humalog exhibited peak action at 25 ± 10 min. These ultrafast kinetics make UFAL a promising candidate for improving glucose control and reducing burden for patients with diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin , Animals , Blood Glucose , Excipients , High-Throughput Screening Assays , Humans , Hypoglycemic Agents , Insulin Lispro , Swine
7.
Microb Biotechnol ; 13(6): 1780-1792, 2020 11.
Article in English | MEDLINE | ID: mdl-32476224

ABSTRACT

The cyanobacterium Synechococcus elongatus (SE) has been shown to rescue ischaemic heart muscle after myocardial infarction by photosynthetic oxygen production. Here, we investigated SE toxicity and hypothesized that systemic SE exposure does not elicit a significant immune response in rats. Wistar rats intravenously received SE (n = 12), sterile saline (n = 12) or E. coli lipopolysaccharide (LPS, n = 4), and a subset (8 SE, 8 saline) received a repeat injection 4 weeks later. At baseline, 4 h, 24 h, 48 h, 8 days and 4 weeks after injection, clinical assessments, blood cultures, blood counts, lymphocyte phenotypes, liver function tests, proinflammatory cytokines and immunoglobulins were assessed. Across all metrics, SE rats responded comparably to saline controls, displaying no clinically significant immune response. As expected, LPS rats exhibited severe immunological responses. Systemic SE administration does not induce sepsis or toxicity in rats, thereby supporting the safety of cyanobacteria-mammalian symbiotic therapeutics using this organism.


Subject(s)
Escherichia coli , Synechococcus , Animals , Photosynthesis , Rats , Rats, Wistar
8.
Sci Rep ; 10(1): 7319, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355240

ABSTRACT

Neonatal mice exhibit natural heart regeneration after myocardial infarction (MI) on postnatal day 1 (P1), but this ability is lost by postnatal day 7 (P7). Cardiac biomechanics intricately affect long-term heart function, but whether regenerated cardiac muscle is biomechanically similar to native myocardium remains unknown. We hypothesized that neonatal heart regeneration preserves native left ventricular (LV) biomechanical properties after MI. C57BL/6J mice underwent sham surgery or left anterior descending coronary artery ligation at age P1 or P7. Echocardiography performed 4 weeks post-MI showed that P1 MI and sham mice (n = 22, each) had similar LV wall thickness, diameter, and ejection fraction (59.6% vs 60.7%, p = 0.6514). Compared to P7 shams (n = 20), P7 MI mice (n = 20) had significant LV wall thinning, chamber enlargement, and depressed ejection fraction (32.6% vs 61.8%, p < 0.0001). Afterward, the LV was explanted and pressurized ex vivo, and the multiaxial lenticular stress-strain relationship was tracked. While LV tissue modulus for P1 MI and sham mice were similar (341.9 kPa vs 363.4 kPa, p = 0.6140), the modulus for P7 MI mice was significantly greater than that for P7 shams (691.6 kPa vs 429.2 kPa, p = 0.0194). We conclude that, in neonatal mice, regenerated LV muscle has similar biomechanical properties as native LV myocardium.


Subject(s)
Heart Ventricles/physiopathology , Heart/physiology , Myocardial Infarction/pathology , Myocardium/pathology , Regeneration , Animals , Animals, Newborn , Biomechanical Phenomena , Cell Proliferation , Collagen/chemistry , Echocardiography , Female , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Stress, Mechanical , Ventricular Remodeling
9.
Nat Biomed Eng ; 4(5): 507-517, 2020 05.
Article in English | MEDLINE | ID: mdl-32393892

ABSTRACT

Treatment of patients with diabetes with insulin and pramlintide (an amylin analogue) is more effective than treatment with insulin only. However, because mixtures of insulin and pramlintide are unstable and have to be injected separately, amylin analogues are only used by 1.5% of people with diabetes needing rapid-acting insulin. Here, we show that the supramolecular modification of insulin and pramlintide with cucurbit[7]uril-conjugated polyethylene glycol improves the pharmacokinetics of the dual-hormone therapy and enhances postprandial glucagon suppression in diabetic pigs. The co-formulation is stable for over 100 h at 37 °C under continuous agitation, whereas commercial formulations of insulin analogues aggregate after 10 h under similar conditions. In diabetic rats, the administration of the stabilized co-formulation increased the area-of-overlap ratio of the pharmacokinetic curves of pramlintide and insulin from 0.4 ± 0.2 to 0.7 ± 0.1 (mean ± s.d.) for the separate administration of the hormones. The co-administration of supramolecularly stabilized insulin and pramlintide better mimics the endogenous kinetics of co-secreted insulin and amylin, and holds promise as a dual-hormone replacement therapy.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Drug Compounding , Glucagon/metabolism , Insulin/therapeutic use , Islet Amyloid Polypeptide/therapeutic use , Animals , Bridged-Ring Compounds/chemistry , Diffusion , Drug Administration Routes , Drug Stability , Hydrogen-Ion Concentration , Imidazoles/chemistry , Insulin/administration & dosage , Insulin/pharmacokinetics , Insulin/pharmacology , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/pharmacokinetics , Islet Amyloid Polypeptide/pharmacology , Male , Polyethylene Glycols/chemistry , Rats, Sprague-Dawley , Signal Transduction/drug effects , Swine
10.
Cells ; 9(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31963369

ABSTRACT

Newborn mice and piglets exhibit natural heart regeneration after myocardial infarction (MI). Discovering other mammals with this ability would provide evidence that neonatal cardiac regeneration after MI may be a conserved phenotype, which if activated in adults could open new options for treating ischemic cardiomyopathy in humans. Here, we hypothesized that newborn rats undergo natural heart regeneration after MI. Using a neonatal rat MI model, we performed left anterior descending coronary artery ligation or sham surgery in one-day-old rats under hypothermic circulatory arrest (n = 74). Operative survival was 97.3%. At 1 day post-surgery, rats in the MI group exhibited significantly reduced ejection fraction (EF) compared to shams (87.1% vs. 53.0%, p < 0.0001). At 3 weeks post-surgery, rats in the sham and MI groups demonstrated no difference in EF (71.1% vs. 69.2%, respectively, p = 0.2511), left ventricular wall thickness (p = 0.9458), or chamber diameter (p = 0.7801). Masson's trichome and picrosirius red staining revealed minimal collagen scar after MI. Increased numbers of cardiomyocytes positive for 5-ethynyl-2'-deoxyuridine (p = 0.0072), Ki-67 (p = 0.0340), and aurora B kinase (p = 0.0430) were observed within the peri-infarct region after MI, indicating ischemia-induced cardiomyocyte proliferation. Overall, we present a neonatal rat MI model and demonstrate that newborn rats are capable of endogenous neocardiomyogenesis after MI.


Subject(s)
Myocardial Infarction/physiopathology , Regeneration , Animals , Animals, Newborn , Aurora Kinase B/metabolism , Cell Proliferation , Cicatrix/pathology , Collagen/metabolism , Disease Models, Animal , Electrocardiography , Female , Fibrosis , Ki-67 Antigen/metabolism , Ligation , Male , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocytes, Cardiac/pathology , Rats, Wistar , Time Factors , Troponin/metabolism
11.
Cytokine ; 127: 154974, 2020 03.
Article in English | MEDLINE | ID: mdl-31978642

ABSTRACT

Although ischemic heart disease is the leading cause of death worldwide, mainstay treatments ultimately fail because they do not adequately address disease pathophysiology. Restoring the microvascular perfusion deficit remains a significant unmet need and may be addressed via delivery of pro-angiogenic cytokines. The therapeutic effect of cytokines can be enhanced by encapsulation within hydrogels, but current hydrogels do not offer sufficient clinical translatability due to unfavorable viscoelastic mechanical behavior which directly impacts the ability for minimally-invasive catheter delivery. In this report, we examine the therapeutic implications of dual-stage cytokine release from a novel, highly shear-thinning biocompatible catheter-deliverable hydrogel. We chose to encapsulate two protein-engineered cytokines, namely dimeric fragment of hepatocyte growth factor (HGFdf) and engineered stromal cell-derived factor 1α (ESA), which target distinct disease pathways. The controlled release of HGFdf and ESA from separate phases of the hyaluronic acid-based hydrogel allows extended and pronounced beneficial effects due to the precise timing of release. We evaluated the therapeutic efficacy of this treatment strategy in a small animal model of myocardial ischemia and observed a significant benefit in biological and functional parameters. Given the encouraging results from the small animal experiment, we translated this treatment to a large animal preclinical model and observed a reduction in scar size, indicating this strategy could serve as a potential adjunct therapy for the millions of people suffering from ischemic heart disease.


Subject(s)
Hydrogels/administration & dosage , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Catheters , Cells, Cultured , Disease Models, Animal , Hepatocyte Growth Factor/metabolism , Humans , Hyaluronic Acid/administration & dosage , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocardium/pathology , Rats
12.
Proc Natl Acad Sci U S A ; 116(42): 20820-20827, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570592

ABSTRACT

Polyphosphate fire retardants are a critical tactical resource for fighting fires in the wildland and in the wildland-urban interface. Yet, application of these retardants is limited to emergency suppression strategies because current formulations cannot retain fire retardants on target vegetation for extended periods of time through environmental exposure and weathering. New retardant formulations with persistent retention to target vegetation throughout the peak fire season would enable methodical, prophylactic treatment strategies of landscapes at high risk of wildfires through prolonged prevention of ignition and continual impediment to active flaming fronts. Here we develop a sprayable, environmentally benign viscoelastic fluid comprising biopolymers and colloidal silica to enhance adherence and retention of polyphosphate retardants on common wildfire-prone vegetation. These viscoelastic fluids exhibit appropriate wetting and rheological responses to enable robust retardant adherence to vegetation following spray application. Further, laboratory and pilot-scale burn studies establish that these materials drastically reduce ignition probability before and after simulated weathering events. Overall, these studies demonstrate how these materials actualize opportunities to shift the approach of retardant-based wildfire management from reactive suppression to proactive prevention at the source of ignitions.


Subject(s)
Conservation of Natural Resources/methods , Flame Retardants/analysis , Viscoelastic Substances/chemistry , Wildfires/prevention & control , Forests , Polyphosphates/chemistry , Seasons , Trees/chemistry
13.
Nat Biomed Eng ; 3(8): 611-620, 2019 08.
Article in English | MEDLINE | ID: mdl-31391596

ABSTRACT

Post-operative adhesions form as a result of normal wound healing processes following any type of surgery. In cardiac surgery, pericardial adhesions are particularly problematic during reoperations, as surgeons must release the adhesions from the surface of the heart before the intended procedure can begin, thereby substantially lengthening operation times and introducing risks of haemorrhage and injury to the heart and lungs during sternal re-entry and cardiac dissection. Here we show that a dynamically crosslinked supramolecular polymer-nanoparticle hydrogel, with viscoelastic and flow properties that enable spraying onto tissue as well as robust tissue adherence and local retention in vivo for two weeks, reduces the formation of pericardial adhesions. In a rat model of severe pericardial adhesions, the hydrogel markedly reduced the severity of the adhesions, whereas commercial adhesion barriers (including Seprafilm and Interceed) did not. The hydrogels also reduced the severity of cardiac adhesions (relative to untreated animals) in a clinically relevant cardiopulmonary-bypass model in sheep. This viscoelastic supramolecular polymeric hydrogel represents a promising clinical solution for the prevention of post-operative pericardial adhesions.


Subject(s)
Cardiac Surgical Procedures/methods , Hydrogels/chemistry , Pericardium/surgery , Polymers/chemistry , Tissue Adhesions , Animals , Cellulose, Oxidized , Hyaluronic Acid , Hydrogels/therapeutic use , Male , Models, Animal , Nanoparticles , Polymers/therapeutic use , Rats , Sheep
14.
J Mech Behav Biomed Mater ; 96: 165-171, 2019 08.
Article in English | MEDLINE | ID: mdl-31035067

ABSTRACT

Adverse remodeling of the left ventricle (LV) after myocardial infarction (MI) results in abnormal tissue biomechanics and impaired cardiac function, often leading to heart failure. We hypothesized that intramyocardial delivery of engineered stromal cell-derived factor 1α analog (ESA), our previously-developed supra-efficient pro-angiogenic chemokine, preserves biaxial LV mechanical properties after MI. Male Wistar rats (n = 45) underwent sham surgery (n = 15) or permanent left anterior descending coronary artery ligation. Rats sustaining MI were randomized for intramyocardial injections of either saline (100 µL, n = 15) or ESA (6 µg/kg, n = 15), delivered at four standardized borderzone sites. After 4 weeks, echocardiography was performed, and the hearts were explanted. Tensile testing of the anterolateral LV wall was performed using a displacement-controlled biaxial load frame, and modulus was determined after constitutive modeling. At 4 weeks post-MI, compared to saline controls, ESA-treated hearts had greater wall thickness (1.68 ±â€¯0.05 mm vs 1.42 ±â€¯0.08 mm, p = 0.008), smaller end-diastolic LV internal dimension (6.88 ±â€¯0.29 mm vs 7.69 ±â€¯0.22 mm, p = 0.044), and improved ejection fraction (62.8 ±â€¯3.0% vs 49.4 ±â€¯4.5%, p = 0.014). Histologic analysis revealed significantly reduced infarct size for ESA-treated hearts compared to saline controls (29.4 ±â€¯2.9% vs 41.6 ±â€¯3.1%, p = 0.021). Infarcted hearts treated with ESA exhibited decreased modulus compared to those treated with saline in both the circumferential (211.5 ±â€¯6.9 kPa vs 264.3 ±â€¯12.5 kPa, p = 0.001) and longitudinal axes (194.5 ±â€¯6.5 kPa vs 258.1 ±â€¯14.4 kPa, p < 0.001). In both principal directions, ESA-treated infarcted hearts possessed similar tissue compliance as sham non-infarcted hearts. Overall, intramyocardial ESA therapy improves post-MI ventricular remodeling and function, reduces infarct size, and preserves native LV biaxial mechanical properties.


Subject(s)
Chemokine CXCL12/genetics , Chemokine CXCL12/pharmacology , Heart/drug effects , Heart/physiopathology , Mechanical Phenomena/drug effects , Myocardial Infarction/physiopathology , Protein Engineering , Animals , Biomechanical Phenomena/drug effects , Male , Rats , Rats, Wistar , Ventricular Remodeling/drug effects
15.
J Thorac Cardiovasc Surg ; 158(2): 392-403, 2019 08.
Article in English | MEDLINE | ID: mdl-30745047

ABSTRACT

OBJECTIVE: The optimal conduit for valve-sparing aortic root replacement is still debated, with several conduit variations available, ranging from straight tubular grafts to Valsalva grafts. Benefits of neosinus reconstruction include enhanced flow profiles and improved hemodynamics. Curiously, however, some clinical data suggest that straight grafts may have greater long-term durability. In this study, we hypothesized that straight tubular grafts may help maintain the native cylindrical position of the aortic valve commissures radially, resulting in preserved leaflet coaptation, reduced stresses, and potentially improved valve performance. METHODS: Using 3D printing, a left heart simulator with a valve-sparing root replacement model and a physiologic coronary circulation was constructed. Aortic valves were dissected from fresh porcine hearts and reimplanted into either straight tubular grafts (n = 6) or Valsalva grafts (n = 6). Conduits were mounted into the heart simulator and hemodynamic, echocardiographic, and high-speed videometric data were collected. RESULTS: Hemodynamic parameters and coronary blood flow were similar between straight and Valsalva grafts, although the former were associated with lower regurgitant fractions, less peak intercommissural radial separation, preserved leaflet coaptation, decreased leaflet velocities, and lower relative leaflet forces compared with Valsalva grafts. CONCLUSIONS: Valsalva grafts and straight grafts perform equally well in terms of gross hemodyanics and coronary blood flow. Interestingly, however, the biomechanics of these 2 conduits differ considerably, with straight grafts providing increased radial commissural stability and leaflet coaptation. Further investigation into how these parameters influence clinical outcomes is warranted.


Subject(s)
Aorta/surgery , Aortic Valve/surgery , Printing, Three-Dimensional , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Valve/pathology , Aortic Valve/physiopathology , Biomechanical Phenomena , Coronary Circulation , Echocardiography, Transesophageal , Hemodynamics , Models, Anatomic , Sinus of Valsalva/pathology , Sinus of Valsalva/physiopathology , Sinus of Valsalva/surgery , Swine , Vascular Grafting/methods
16.
Adv Healthc Mater ; 8(5): e1801147, 2019 03.
Article in English | MEDLINE | ID: mdl-30714355

ABSTRACT

Hydrogels have emerged as a diverse class of biomaterials offering a broad range of biomedical applications. Specifically, injectable hydrogels are advantageous for minimally invasive delivery of various therapeutics and have great potential to treat a number of diseases. However, most current injectable hydrogels are limited by difficult and time-consuming fabrication techniques and are unable to be delivered through long, narrow catheters, preventing extensive clinical translation. Here, the development of an easily-scaled, catheter-injectable hydrogel utilizing a polymer-nanoparticle crosslinking mechanism is reported, which exhibits notable shear-thinning and self-healing behavior. Gelation of the hydrogel occurs immediately upon mixing the biochemically modified hyaluronic acid polymer with biodegradable nanoparticles and can be easily injected through a high-gauge syringe due to the dynamic nature of the strong, yet reversible crosslinks. Furthermore, the ability to deliver this novel hydrogel through a long, narrow, physiologically-relevant catheter affixed with a 28-G needle is highlighted, with hydrogel mechanics unchanged after delivery. Due to the composition of the gel, it is demonstrated that therapeutics can be differentially released with distinct elution profiles, allowing precise control over drug delivery. Finally, the cell-signaling and biocompatibility properties of this innovative hydrogel are demonstrated, revealing its wide range of therapeutic applications.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Animals , Cell Line , Drug Delivery Systems/methods , Humans , Male , Mice , NIH 3T3 Cells , Polymers/chemistry , Rats , Rats, Wistar , Tissue Engineering/methods
17.
Macromol Biosci ; 19(1): e1800275, 2019 01.
Article in English | MEDLINE | ID: mdl-30369048

ABSTRACT

Drug delivery and cell transplantation require minimally invasive deployment strategies such as injection through clinically relevant high-gauge needles. Supramolecular hydrogels comprising dodecyl-modified hydroxypropylmethylcellulose and poly(ethylene glycol)-block-poly(lactic acid) have been previously demonstrated for the delivery of drugs and proteins. Here, it is demonstrated that the rheological properties of these hydrogels allow for facile injectability, an increase of cell viability after injection when compared to cell viabilities of cells injected in phosphate-buffered saline, and homogeneous cell suspensions that do not settle. These hydrogels are injected at 1 mL min-1 with pressures less than 400 kPa, despite the solid-like properties of the gel when at rest. The cell viabilities immediately after injection are greater than 86% for adult human dermal fibroblasts, human umbilical vein cells, smooth muscle cells, and human mesenchymal stem cells. Cells are shown to remain suspended and proliferate in the hydrogel at the same rate as observed in cell media. The work expands on the versatility of these hydrogels and lays a foundation for the codelivery of drugs, proteins, and cells.


Subject(s)
Drug Delivery Systems , Hydrogels , Materials Testing , Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Cell Survival/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Polyesters/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology
18.
Circulation ; 138(19): 2130-2144, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30474423

ABSTRACT

BACKGROUND: Cardiovascular bypass grafting is an essential treatment for complex cases of atherosclerotic disease. Because the availability of autologous arterial and venous conduits is patient-limited, self-assembled cell-only grafts have been developed to serve as functional conduits with off-the-shelf availability. The unacceptably long production time required to generate these conduits, however, currently limits their clinical utility. Here, we introduce a novel technique to significantly accelerate the production process of self-assembled engineered vascular conduits. METHODS: Human aortic smooth muscle cells and skin fibroblasts were used to construct bilevel cell sheets. Cell sheets were wrapped around a 22.5-gauge Angiocath needle to form tubular vessel constructs. A thin, flexible membrane of clinically approved biodegradable tissue glue (Dermabond Advanced) served as a temporary, external scaffold, allowing immediate perfusion and endothelialization of the vessel construct in a bioreactor. Subsequently, the matured vascular conduits were used as femoral artery interposition grafts in rats (n=20). Burst pressure, vasoreactivity, flow dynamics, perfusion, graft patency, and histological structure were assessed. RESULTS: Compared with engineered vascular conduits formed without external stabilization, glue membrane-stabilized conduits reached maturity in the bioreactor in one-fifth the time. After only 2 weeks of perfusion, the matured conduits exhibited flow dynamics similar to that of control arteries, as well as physiological responses to vasoconstricting and vasodilating drugs. The matured conduits had burst pressures exceeding 500 mm Hg and had sufficient mechanical stability for surgical anastomoses. The patency rate of implanted conduits at 8 weeks was 100%, with flow rate and hind-limb perfusion similar to those of sham controls. Grafts explanted after 8 weeks showed a histological structure resembling that of typical arteries, including intima, media, adventitia, and internal and external elastic membrane layers. CONCLUSIONS: Our technique reduces the production time of self-assembled, cell sheet-derived engineered vascular conduits to 2 weeks, thereby permitting their use as bypass grafts within the clinical time window for elective cardiovascular surgery. Furthermore, our method uses only clinically approved materials and can be adapted to various cell sources, simplifying the path toward future clinical translation.


Subject(s)
Bioprosthesis , Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Femoral Artery/surgery , Muscle, Smooth, Vascular/cytology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Aorta/cytology , Blood Flow Velocity , Blood Vessel Prosthesis Implantation/adverse effects , Cells, Cultured , Coculture Techniques , Femoral Artery/pathology , Femoral Artery/physiopathology , Fibroblasts , Humans , Male , Myocytes, Smooth Muscle , Prosthesis Design , Prosthesis Failure , Rats, Nude , Regional Blood Flow , Stress, Mechanical , Tensile Strength , Time Factors , Vascular Patency
19.
Biointerphases ; 13(6): 06D406, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30231617

ABSTRACT

Poly(N-isopropyl acrylamide) (pNIPAM) is a stimulus-responsive polymer that has been of great interest to the bioengineering community. When the temperature is lowered below its lower critical solution temperature (∼32 °C), pNIPAM rapidly hydrates, and adherent cells detach as intact cell sheets. This cell-releasing behavior in a physiologically relevant temperature range has led to NIPAM's use for engineered tissues and other devices. In a previous study, however, the authors found that although most techniques used to polymerize NIPAM yield biocompatible films, some formulations from commercially-available NIPAM (cpNIPAM) can be cytotoxic. In this work, the authors investigate the reasons underlying this anomaly. The authors evaluated the response of a variety of cell types (e.g., bovine aortic endothelial cells, BAECs; monkey kidney epithelial cells, Vero cells; and mouse embryonic fibroblasts, 3T3s) after culture on substrates spin-coated with sol-gel (spNIPAM) and commercially-prepared (cpNIPAM). The relative biocompatibility of each cell type was evaluated using observations of its cell morphology and function (e.g., XTT and Live/Dead assays) after 48 and 96 h in culture. In addition, the substrates themselves were analyzed using NMR, goniometry, and XPS. The authors find that all the cell types were compromised by 96 h in culture with cpNIPAM, although the manner in which the cells are compromised differs; in particular, while Vero and 3T3 cells appear to be undergoing cytotoxic death, BAECs undergo apoptic death. The authors believe that this result is due to a combination of factors, including the presence of short chain oligomers of NIPAM in the commercially-available preparation. This work will provide valuable insights into the cytotoxicity of commercially-prepared polymer substrates for this type of bioengineering work and therefore into the applicability of cells grown on such surfaces for human subjects.


Subject(s)
Acrylic Resins/toxicity , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Fibroblasts/drug effects , Acrylic Resins/chemistry , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/physiology , Epithelial Cells/physiology , Fibroblasts/physiology , Humans , Magnetic Resonance Spectroscopy , Mice , Photoelectron Spectroscopy , Time Factors
20.
Cardiovasc Diabetol ; 16(1): 142, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29096622

ABSTRACT

BACKGROUND: Diabetes mellitus is a risk factor for coronary artery disease and diabetic cardiomyopathy, and adversely impacts outcomes following coronary artery bypass grafting. Current treatments focus on macro-revascularization and neglect the microvascular disease typical of diabetes mellitus-induced cardiomyopathy (DMCM). We hypothesized that engineered smooth muscle cell (SMC)-endothelial progenitor cell (EPC) bi-level cell sheets could improve ventricular dysfunction in DMCM. METHODS: Primary mesenchymal stem cells (MSCs) and EPCs were isolated from the bone marrow of Wistar rats, and MSCs were differentiated into SMCs by culturing on a fibronectin-coated dish. SMCs topped with EPCs were detached from a temperature-responsive culture dish to create an SMC-EPC bi-level cell sheet. A DMCM model was induced by intraperitoneal streptozotocin injection. Four weeks after induction, rats were randomized into 3 groups: control (no DMCM induction), untreated DMCM, and treated DMCM (cell sheet transplant covering the anterior surface of the left ventricle). RESULTS: SMC-EPC cell sheet therapy preserved cardiac function and halted adverse ventricular remodeling, as demonstrated by echocardiography and cardiac magnetic resonance imaging at 8 weeks after DMCM induction. Myocardial contrast echocardiography demonstrated that myocardial perfusion and microvascular function were preserved in the treatment group compared with untreated animals. Histological analysis demonstrated decreased interstitial fibrosis and increased microvascular density in the SMC-EPC cell sheet-treated group. CONCLUSIONS: Treatment of DMCM with tissue-engineered SMC-EPC bi-level cell sheets prevented cardiac dysfunction and microvascular disease associated with DMCM. This multi-lineage cellular therapy is a novel, translatable approach to improve microvascular disease and prevent heart failure in diabetic patients.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Diabetic Cardiomyopathies/prevention & control , Endothelial Progenitor Cells/transplantation , Microvessels , Myocytes, Smooth Muscle/transplantation , Tissue Engineering/methods , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/diagnostic imaging , Diabetes Mellitus, Type 1/physiopathology , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Disease Progression , Fibrosis , Microvessels/physiopathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Rodentia
SELECTION OF CITATIONS
SEARCH DETAIL
...