Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916992

ABSTRACT

Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches, and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.

2.
Mol Ther Methods Clin Dev ; 27: 452-463, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36419468

ABSTRACT

Sanfilippo syndrome type B (mucopolysaccharidosis type IIIB) is a recessive genetic disorder that severely affects the brain due to a deficiency in the enzyme α-N-acetylglucosaminidase (NAGLU), leading to intra-lysosomal accumulation of partially degraded heparan sulfate. There are no effective treatments for this disorder. In this project, we carried out an ex vivo correction of neural stem cells derived from Naglu -/- mice (iNSCs) induced pluripotent stem cells (iPSC) using a modified enzyme in which human NAGLU is fused to an insulin-like growth factor II receptor binding peptide in order to improve enzyme uptake. After brain transplantation of corrected iNSCs into Naglu -/- mice and long-term evaluation of their impact, we successfully detected NAGLU-IGFII activity in all transplanted animals. We found decreased lysosomal accumulation and reduced astrocytosis and microglial activation throughout transplanted brains. We also identified a novel neuropathological phenotype in untreated Naglu -/- brains with decreased levels of the neuronal marker Map2 and accumulation of synaptophysin-positive aggregates. Upon transplantation, we restored levels of Map2 expression and significantly reduced formation of synaptophysin-positive aggregates. Our findings suggest that genetically engineered iNSCs can be used to effectively deliver the missing enzyme to the brain and treat Sanfilippo type B-associated neuropathology.

3.
Front Endocrinol (Lausanne) ; 12: 732431, 2021.
Article in English | MEDLINE | ID: mdl-34589059

ABSTRACT

Diabetes is a complex disease that affects over 400 million people worldwide. The life-long insulin injections and continuous blood glucose monitoring required in type 1 diabetes (T1D) represent a tremendous clinical and economic burdens that urges the need for a medical solution. Pancreatic islet transplantation holds great promise in the treatment of T1D; however, the difficulty in regulating post-transplantation immune reactions to avoid both allogenic and autoimmune graft rejection represent a bottleneck in the field of islet transplantation. Cell replacement strategies have been performed in hepatic, intramuscular, omentum, and subcutaneous sites, and have been performed in both animal models and human patients. However more optimal transplantation sites and methods of improving islet graft survival are needed to successfully translate these studies to a clinical relevant therapy. In this review, we summarize the current progress in the field as well as methods and sites of islet transplantation, including stem cell-derived functional human islets. We also discuss the contribution of immune cells, vessel formation, extracellular matrix, and nutritional supply on islet graft survival. Developing new transplantation sites with emerging technologies to improve islet graft survival and simplify immune regulation will greatly benefit the future success of islet cell therapy in the treatment of diabetes.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/trends , Animals , Graft Survival , Humans , Islets of Langerhans/physiology , Islets of Langerhans Transplantation/methods
4.
J Pathol ; 255(1): 41-51, 2021 09.
Article in English | MEDLINE | ID: mdl-34050678

ABSTRACT

Down syndrome (DS), also known as trisomy 21 (T21), is the most common human chromosomal anomaly. Although DS can affect many organ systems, lung and heart disease are the leading causes of death. An abundance of existing data suggests that lung abnormalities originate postnatally in DS. However, a single report of branching insufficiency in DS has inferred a potential prenatal origin. The histology of T21 fetal lungs (n = 15) was assessed by an experienced pathologist. Spatial differences in cellular phenotypes were examined using immunohistochemistry (IHC). Comprehensive gene expression in prenatal T21 lungs (n = 19), and age-matched controls (n = 19), was performed using high-throughput RNA sequencing (RNAseq) and validated by RT-qPCR. Histopathological abnormalities were observed in approximately half of T21 prenatal lung samples analyzed, which included dilated terminal airways/acinar tubules, dilated lymphatics, and arterial wall thickening. IHC for Ki67 revealed significant reductions in epithelial and mesenchymal cell proliferation, predominantly in tissues displaying pathology. IHC demonstrated that airway smooth muscle was reduced and discontinuous in the proximal airway in conjunction with reduced SOX2. RNAseq identified 118 genes significantly dysregulated (FDR < 0.05) in T21 lung when unadjusted and 316 genes when adjusted for age. Ontology analysis showed that IFN pathway genes were appreciably upregulated, whereas complement and coagulation cascades and extracellular matrix pathway genes were downregulated. RT-qPCR confirmed the changes in genes associated with these pathways in prenatal T21 lungs. Our data demonstrate that specific histological, cellular, and molecular abnormalities occur prenatally in different compartments of human T21 lung, which could be representative of premature stage progression. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Down Syndrome/pathology , Lung/abnormalities , Fetus , Humans
5.
J Endod ; 46(9S): S101-S104, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32950181

ABSTRACT

Spontaneous healing and recovery of innervated and vascularized tissues are limited. In particular, the complexity of the central nervous system's anatomy, physiology, and pathobiology make efforts to develop effective therapeutic strategies exceptionally challenging. Repairing the brain after injury implies restoring the tissue architecture of the neural and vascular networks both morphologically and functionally. The substantial clinical burden and disability after a central nervous system injury urges the need to explore therapeutic solutions outside the confine of conventional approaches used in regenerative medicine. Recent advances in tissue engineering and material sciences have developed biomimetic materials that can be injected or implanted directly to the site of damage to provide physical support to cell infiltration and growth, promoting tissue development and de novo formation of vascular and axonal networks through cell transplantation and/or controlled release of bioactive cues. These approaches have shown promise in promoting the endogenous repair machinery of the brain and controlling the growth and development of functional vascular and neural networks in the lesion to promote long-term functional recovery. This narrative review presents a comprehensive look at recent advances using proangiogenic engineered materials and drug delivery systems for brain repair after stroke.


Subject(s)
Biocompatible Materials , Tissue Engineering , Brain , Regenerative Medicine , Wound Healing
6.
Adv Mater ; 31(33): e1900727, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31125138

ABSTRACT

The central nervous system (CNS) plays a central role in the control of sensory and motor functions, and the disruption of its barriers can result in severe and debilitating neurological disorders. Neurotrophins are promising therapeutic agents for neural regeneration in the damaged CNS. However, their penetration across the blood-brain barrier remains a formidable challenge, representing a bottleneck for brain and spinal cord therapy. Herein, a nanocapsule-based delivery system is reported that enables intravenously injected nerve growth factor (NGF) to enter the CNS in healthy mice and nonhuman primates. Under pathological conditions, the delivery of NGF enables neural regeneration, tissue remodeling, and functional recovery in mice with spinal cord injury. This technology can be utilized to deliver other neurotrophins and growth factors to the CNS, opening a new avenue for tissue engineering and the treatment of CNS disorders and neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Nanocapsules/chemistry , Nerve Growth Factors/pharmacology , Nerve Regeneration/drug effects , Spinal Cord Injuries/drug therapy , Acrylic Resins/chemistry , Animals , Biocompatible Materials/chemistry , Blood-Brain Barrier/ultrastructure , Cross-Linking Reagents/chemistry , Drug Liberation , Injections, Intravenous , Macaca mulatta , Methacrylates/chemistry , Mice, Inbred BALB C , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/blood , Nerve Growth Factors/cerebrospinal fluid , PC12 Cells , Permeability , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Polyesters/chemistry , Rats , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
7.
Nat Mater ; 17(7): 642-651, 2018 07.
Article in English | MEDLINE | ID: mdl-29784996

ABSTRACT

Stroke is the primary cause of disability due to the brain's limited ability to regenerate damaged tissue. After stroke, an increased inflammatory and immune response coupled with severely limited angiogenesis and neuronal growth results in a stroke cavity devoid of normal brain tissue. In the adult, therapeutic angiogenic materials have been used to repair ischaemic tissues through the formation of vascular networks. However, whether a therapeutic angiogenic material can regenerate brain tissue and promote neural repair is poorly understood. Here we show that the delivery of an engineered immune-modulating angiogenic biomaterial directly to the stroke cavity promotes tissue formation de novo, and results in axonal networks along thee generated blood vessels. This regenerated tissue produces functional recovery through the established axonal networks. Thus, this biomaterials approach generates a vascularized network of regenerated functional neuronal connections within previously dead tissue and lays the groundwork for the use of angiogenic materials to repair other neurologically diseased tissues.


Subject(s)
Biocompatible Materials , Brain/pathology , Neovascularization, Physiologic , Stroke/pathology , Animals , Brain/blood supply , Brain/physiopathology , Heparin/administration & dosage , Humans , Nanoparticles/administration & dosage , Neurogenesis , Recovery of Function , Stroke/physiopathology , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
8.
Nat Mater ; 16(9): 953-961, 2017 09.
Article in English | MEDLINE | ID: mdl-28783156

ABSTRACT

Integrin binding to bioengineered hydrogel scaffolds is essential for tissue regrowth and regeneration, yet not all integrin binding can lead to tissue repair. Here, we show that through engineering hydrogel materials to promote α3/α5ß1 integrin binding, we can promote the formation of a space-filling and mature vasculature compared with hydrogel materials that promote αvß3 integrin binding. In vitro, α3/α5ß1 scaffolds promoted endothelial cells to sprout and branch, forming organized extensive networks that eventually reached and anastomosed with neighbouring branches. In vivo, α3/α5ß1 scaffolds delivering vascular endothelial growth factor (VEGF) promoted non-tortuous blood vessel formation and non-leaky blood vessels by 10 days post-stroke. In contrast, materials that promote αvß3 integrin binding promoted endothelial sprout clumping in vitro and leaky vessels in vivo. This work shows that precisely controlled integrin activation from a biomaterial can be harnessed to direct therapeutic vessel regeneration and reduce VEGF-induced vascular permeability in vivo.


Subject(s)
Blood Vessel Prosthesis , Capillary Permeability , Fibronectins/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Hydrogels/chemistry , Integrin alpha3/metabolism , Integrin alpha5beta1/metabolism , Bioprosthesis , Human Umbilical Vein Endothelial Cells/cytology , Humans , Tissue Engineering/methods
9.
Adv Mater ; 29(32)2017 Aug.
Article in English | MEDLINE | ID: mdl-28650574

ABSTRACT

With the number of deaths due to stroke decreasing, more individuals are forced to live with crippling disability resulting from the stroke. To date, no therapeutics exist after the first 4.5 h after the stroke onset, aside from rest and physical therapy. Following stroke, a large influx of astrocytes and microglia releasing proinflammatory cytokines leads to dramatic inflammation and glial scar formation, affecting brain tissue's ability to repair itself. Pathological conditions, such as a stroke, trigger neural progenitor cells (NPCs) proliferation and migration toward the damaged site. However, these progenitors are often found far from the cavity or the peri-infarct tissue. Poststroke tissue remodeling results in a compartmentalized cavity that can directly accept a therapeutic material injection. Here, this paper shows that the injection of a porous hyaluronic acid hydrogel into the stroke cavity significantly reduces the inflammatory response following stroke while increasing peri-infarct vascularization compared to nonporous hydrogel controls and stroke only controls. In addition, it is shown that the injection of this material impacts NPCs proliferation and migration at the subventricular zone niche and results, for the first time, in NPC migration into the stroke site.


Subject(s)
Hydrogels/chemistry , Gliosis , Humans , Inflammation , Neural Stem Cells , Stroke
10.
Data Brief ; 10: 202-209, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27995155

ABSTRACT

This article presents data related to the research article "Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain" (P. Moshayedi, L.R. Nih, I.L. Llorente, A.R. Berg, J. Cinkornpumin, W.E. Lowry et al., 2016) [1] and focuses on the biocompatibility aspects of the hydrogel, including its stiffness and the inflammatory response of the transplanted organ. We have developed an injectable hyaluronic acid (HA)-based hydrogel for stem cell culture and transplantation, to promote brain tissue repair after stroke. This 3D biomaterial was engineered to bind bioactive signals such as adhesive motifs, as well as releasing growth factors while supporting cell growth and tissue infiltration. We used a Design of Experiment approach to create a complex matrix environment in vitro by keeping the hydrogel platform and cell type constant across conditions while systematically varying peptide motifs and growth factors. The optimized HA hydrogel promoted survival of encapsulated human induced pluripotent stem cell derived-neural progenitor cells (iPS-NPCs) after transplantation into the stroke cavity and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. The highlights of this article include: (1) Data of cell and bioactive signals addition on the hydrogel mechanical properties and growth factor diffusion, (2) the use of a design of Experiment (DOE) approach (M.W. 2 Weible and T. Chan-Ling, 2007) [2] to select multi-factorial experimental conditions, and (3) Inflammatory response and cell survival after transplantation.

11.
Biomaterials ; 105: 145-155, 2016 10.
Article in English | MEDLINE | ID: mdl-27521617

ABSTRACT

Stem cell therapies have shown promise in promoting recovery in stroke but have been limited by poor cell survival and differentiation. We have developed a hyaluronic acid (HA)-based self-polymerizing hydrogel that serves as a platform for adhesion of structural motifs and a depot release for growth factors to promote transplant stem cell survival and differentiation. We took an iterative approach in optimizing the complex combination of mechanical, biochemical and biological properties of an HA cell scaffold. First, we optimized stiffness for a minimal reaction of adjacent brain to the transplant. Next hydrogel crosslinkers sensitive to matrix metalloproteinases (MMP) were incorporated as they promoted vascularization. Finally, candidate adhesion motifs and growth factors were systemically changed in vitro using a design of experiment approach to optimize stem cell survival or proliferation. The optimized HA hydrogel, tested in vivo, promoted survival of encapsulated human neural progenitor cells (iPS-NPCs) after transplantation into the stroke core and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. This HA hydrogel can be tracked in vivo with MRI. A hydrogel can serve as a therapeutic adjunct in a stem cell therapy through selective control of stem cell survival and differentiation in vivo.


Subject(s)
Brain/pathology , Hydrogels/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Stroke/pathology , Stroke/therapy , Tissue Scaffolds , Animals , Brain/surgery , Cell Differentiation , Cell Survival , Cells, Cultured , Guided Tissue Regeneration/instrumentation , Humans , Hyaluronic Acid/chemistry , Male , Materials Testing , Mice , Mice, Inbred C57BL , Stem Cell Transplantation/instrumentation , Stem Cell Transplantation/methods , Surface Properties , Tensile Strength , Treatment Outcome , Viscosity
12.
Development ; 140(8): 1720-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23533173

ABSTRACT

Arteriogenesis requires growth of pre-existing arteriolar collateral networks and determines clinical outcome in arterial occlusive diseases. Factors responsible for the development of arteriolar collateral networks are poorly understood. The Notch ligand Delta-like 4 (Dll4) promotes arterial differentiation and restricts vessel branching. We hypothesized that Dll4 may act as a genetic determinant of collateral arterial networks and functional recovery in stroke and hind limb ischemia models in mice. Genetic loss- and gain-of-function approaches in mice showed that Dll4-Notch signaling restricts pial collateral artery formation by modulating arterial branching morphogenesis during embryogenesis. Adult Dll4(+/-) mice showed increased pial collateral numbers, but stroke volume upon middle cerebral artery occlusion was not reduced compared with wild-type littermates. Likewise, Dll4(+/-) mice showed reduced blood flow conductance after femoral artery occlusion, and, despite markedly increased angiogenesis, tissue ischemia was more severe. In peripheral arteries, loss of Dll4 adversely affected excitation-contraction coupling in arterial smooth muscle in response to vasopressor agents and arterial vessel wall adaption in response to increases in blood flow, collectively contributing to reduced flow reserve. We conclude that Dll4-Notch signaling modulates native collateral formation by acting on vascular branching morphogenesis during embryogenesis. Dll4 furthermore affects tissue perfusion by acting on arterial function and structure. Loss of Dll4 stimulates collateral formation and angiogenesis, but in the context of ischemic diseases such beneficial effects are overruled by adverse functional changes, demonstrating that ischemic recovery is not solely determined by collateral number but rather by vessel functionality.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/physiopathology , Membrane Proteins/metabolism , Microvessels/embryology , Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Analysis of Variance , Animals , Calcium-Binding Proteins , Immunohistochemistry , Ischemia/metabolism , Mice , Microvessels/physiology , Real-Time Polymerase Chain Reaction , Regional Blood Flow/physiology , X-Ray Microtomography
13.
Eur J Neurosci ; 35(8): 1208-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22512253

ABSTRACT

Pro-angiogenic cell-based therapies constitute an interesting and attractive approach to enhancing post-stroke neurogenesis and decreasing neurological deficit. However, most new stroke-induced neurons die during the first few weeks after ischemia, thus impairing total recovery. Although the neovascularization process involves different cell types and various growth factors, most cell therapy protocols are based on the biological effects of single-cell-type populations or on the administration of heterogeneous populations of progenitors, namely human cord blood-derived CD34(+) cells, with scarce vascular progenitor cells. Tight cooperation between endothelial cells and smooth muscle cells/pericytes is critical for the development of functional neovessels. We hypothesized that neuroblast survival in stroke brain depends on mature vascular network formation. In this study, we injected a combination of endothelial progenitor cells (EPCs) and smooth muscle progenitor cells (SMPCs), isolated from human umbilical cord blood, into a murine model of permanent focal ischemia induced by middle cerebral artery occlusion. The co-administration of SMPCs and EPCs induced enhanced angiogenesis and vascular remodeling in the peri-infarct and infarct areas, where vessels exhibited a more mature phenotype. This activation of vessel growth resulted in the maintenance of neurogenesis and neuroblast migration to the peri-ischemic cortex. Our data suggest that a mature vascular network is essential for neuroblast survival after cerebral ischemia, and that co-administration of EPCs and SMPCs may constitute a novel therapeutic strategy for improving the treatment of stroke.


Subject(s)
Endothelial Cells/transplantation , Infarction, Middle Cerebral Artery/therapy , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Stem Cells , Angiogenesis Inhibitors/pharmacology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endostatins/pharmacology , Endothelial Cells/physiology , Fetal Blood/cytology , Frizzled Receptors/metabolism , Functional Laterality , Humans , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myocytes, Smooth Muscle/physiology , Neovascularization, Pathologic/etiology , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Peptides/genetics , Peptides/metabolism , Permeability/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...