Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Biomater Adv ; 158: 213775, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38252986

ABSTRACT

The current paradigm of medicine is mostly designed to block or prevent pathological events. Once the disease-led tissue damage occurs, the limited endogenous regeneration may lead to depletion or loss of function for cells in the tissues. Cell therapy is rapidly evolving and influencing the field of medicine, where in some instances attempts to address cell loss in the body. Due to their biological function, engineerability, and their responsiveness to stimuli, cells are ideal candidates for therapeutic applications in many cases. Such promise is yet to be fully obtained as delivery of cells that functionally integrate with the desired tissues upon transplantation is still a topic of scientific research and development. Main known impediments for cell therapy include mechanical insults, cell viability, host's immune response, and lack of required nutrients for the transplanted cells. These challenges could be divided into three different steps: 1) Prior to, 2) during the and 3) after the transplantation procedure. In this review, we attempt to briefly summarize published approaches employing biomaterials to mitigate the above technical challenges. Biomaterials are offering an engineerable platform that could be tuned for different classes of cell transplantation to potentially enhance and lengthen the pharmacodynamics of cell therapies.


Subject(s)
Biocompatible Materials , Regenerative Medicine , Biocompatible Materials/therapeutic use , Biocompatible Materials/pharmacology , Regenerative Medicine/methods , Tissue Engineering/methods , Cell- and Tissue-Based Therapy , Cell Transplantation
2.
ACS Biomater Sci Eng ; 9(4): 1862-1890, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36877212

ABSTRACT

The promise of cell therapy has been augmented by introducing biomaterials, where intricate scaffold shapes are fabricated to accommodate the cells within. In this review, we first discuss cell encapsulation and the promising potential of biomaterials to overcome challenges associated with cell therapy, particularly cellular function and longevity. More specifically, cell therapies in the context of autoimmune disorders, neurodegenerative diseases, and cancer are reviewed from the perspectives of preclinical findings as well as available clinical data. Next, techniques to fabricate cell-biomaterials constructs, focusing on emerging 3D bioprinting technologies, will be reviewed. 3D bioprinting is an advancing field that enables fabricating complex, interconnected, and consistent cell-based constructs capable of scaling up highly reproducible cell-biomaterials platforms with high precision. It is expected that 3D bioprinting devices will expand and become more precise, scalable, and appropriate for clinical manufacturing. Rather than one printer fits all, seeing more application-specific printer types, such as a bioprinter for bone tissue fabrication, which would be different from a bioprinter for skin tissue fabrication, is anticipated in the future.


Subject(s)
Bioprinting , Tissue Engineering , Tissue Engineering/methods , Cell Encapsulation , Bioprinting/methods , Biocompatible Materials/therapeutic use , Cell Transplantation
3.
Commun Biol ; 4(1): 685, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083739

ABSTRACT

Foreign body response (FBR) to biomaterials compromises the function of implants and leads to medical complications. Here, we report a hybrid alginate microcapsule (AlgXO) that attenuated the immune response after implantation, through releasing exosomes derived from human Umbilical Cord Mesenchymal Stem Cells (XOs). Upon release, XOs suppress the local immune microenvironment, where xenotransplantation of rat islets encapsulated in AlgXO led to >170 days euglycemia in immunocompetent mouse model of Type 1 Diabetes. In vitro analyses revealed that XOs suppressed the proliferation of CD3/CD28 activated splenocytes and CD3+ T cells. Comparing suppressive potency of XOs in purified CD3+ T cells versus splenocytes, we found XOs more profoundly suppressed T cells in the splenocytes co-culture, where a heterogenous cell population is present. XOs also suppressed CD3/CD28 activated human peripheral blood mononuclear cells (PBMCs) and reduced their cytokine secretion including IL-2, IL-6, IL-12p70, IL-22, and TNFα. We further demonstrate that XOs mechanism of action is likely mediated via myeloid cells and XOs suppress both murine and human macrophages partly by interfering with NFκB pathway. We propose that through controlled release of XOs, AlgXO provide a promising new platform that could alleviate the local immune response to implantable biomaterials.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Exosomes/immunology , Immunity/immunology , Immunologic Factors/immunology , Islets of Langerhans Transplantation/methods , Animals , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Exosomes/metabolism , Humans , Immunocompromised Host/immunology , Immunologic Factors/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Rats , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Heterologous
4.
Cell Transplant ; 29: 963689720952343, 2020.
Article in English | MEDLINE | ID: mdl-33023311

ABSTRACT

Transplantation of pancreatic islets within a biomaterial device is currently under investigation in clinical trials for the treatment of patients with type 1 diabetes (T1D). Patients' preferences on such implants could guide the designs of next-generation implantable devices; however, such information is not currently available. We surveyed the preferences of 482 patients with T1D on the size, shape, visibility, and transplantation site of islet containing implants. More than 83% of participants were willing to receive autologous stem cells, and there was no significant association between implant fabricated by one's own stem cell with gender (χ2 (1, n = 468) = 0.28; P = 0.6) or with age (χ2 (4, n = 468) = 2.92; P = 0.6). Preferred location for islet transplantation within devices was under the skin (52.7%). 48.3% preferred microscopic disks, and 32.3% preferred a thin device (like a credit card). Moreover, 58.4% preferred the implant to be as small as possible, 25.4% did not care about visibility, and 16.2% preferred their implants not to be visible. Among female participants, 81% cared about the implant visibility, whereas this number was 64% for male respondents (χ2 test (1, n = 468) = 16.34; P < 0.0001). 22% of those younger than 50 years of age and 30% of those older than 50 did not care about the visibility of implant (χ2 test (4, n = 468) = 23.69; P < 0.0001). These results suggest that subcutaneous sites and micron-sized devices are preferred choices among patients with T1D who participated in our survey.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/instrumentation , Adolescent , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Surveys and Questionnaires , Young Adult
5.
ACS Biomater Sci Eng ; 6(5): 2726-2739, 2020 05 11.
Article in English | MEDLINE | ID: mdl-33463292

ABSTRACT

Various subtypes of immunocytes react against implanted biomaterials to eliminate the foreign body object from the host's body. Among these cells, dendritic cells (DCs) play a key role in early immune response, later engaging lymphocytes through antigens presentation. Due to their capability to induce tolerogenic or immunogenic responses, DCs have been considered as key therapeutic targets for immunomodulatory products. For instance, tolerogenic DCs are applied in the treatment of autoimmune diseases, rejection of allograft transplantation, and implanted biomaterial. Due to the emerging importance of DCs in immunomodulatory biomaterials, this Review summarizes DCs' responses-such as adhesion, migration, and maturation-to biomaterials. We also review some examples of key molecules and their applications in DCs' immunoengineering. These evaluations would pave the way for designing advanced biomaterials and nanomaterials to modulate the immune system, applicable in tissue engineering, transplantation, and drug delivery technologies.


Subject(s)
Biocompatible Materials , Dendritic Cells , Antigen Presentation , Immunomodulation , Tissue Engineering
6.
Methods ; 177: 50-57, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31669353

ABSTRACT

Mesenchymal stem or stromal cells are currently under clinical investigation for multiple diseases. While their mechanism of action is still not fully elucidated, vesicles secreted by MSCs are believed to recapitulate their therapeutic potentials to some extent. Microvesicles (MVs), also called as microparticles or ectosome, are among secreted vesicles that could transfer cytoplasmic cargo, including RNA and proteins, from emitting (source) cells to recipient cells. Given the importance of MVs, we here attempted to establish a method to isolate and characterize MVs secreted from unmodified human bone marrow derived MSCs (referred to as native MSCs, and their microvesicles as Native-MVs) and IFNγ stimulated MSCs (referred to as IFNγ-MSCs, and their microvesicles as IFNγ-MVs). We first describe an ultracentrifugation technique to isolate MVs from the conditioned cell culture media of MSCs. Next, we describe characterization and quality control steps to analyze the protein and RNA content of MVs. Finally, we examined the potential of MVs to exert immunomodulatory effects through induction of regulatory T cells (Tregs). Secretory vesicles from MSCs are promising alternatives for cell therapy with applications in drug delivery, regenerative medicine, and immunotherapy.


Subject(s)
Cell-Derived Microparticles/chemistry , Drug Delivery Systems/methods , Mesenchymal Stem Cells/chemistry , Proteomics/methods , Regenerative Medicine/methods , Animals , Bone Marrow Cells/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Separation/methods , Cell-Derived Microparticles/immunology , Culture Media, Conditioned/chemistry , Humans , Immunotherapy/methods , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Proteins/classification , Proteins/isolation & purification , RNA/classification , RNA/isolation & purification , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
7.
Small ; 15(34): e1902333, 2019 08.
Article in English | MEDLINE | ID: mdl-31250985

ABSTRACT

Incapability of effective cross-talk with biological environments has partly impaired the in vivo functionality of nanoparticles (NPs). Homing, biodistribution, and function of NPs could be engineered through regulating their interactions with in vivo niches. Inspired by communications in biological systems, endowing a "biological identity" to synthetic NPs is one approach to control their biodistribution, and immunonegotiation profiles. This synthetic-biological combination is referred to as biohybrid NPs, which comprise both i) engineerable, readily producible, and trackable synthetic NPs as well as ii) biological moieties with the capability to cross-talk with immunological barriers. Here, the latest understanding on the in vivo interactions of NPs, biological barriers they face, and emerging methods for quantitative measurements of NPs' biodistribution are reviewed. Some key biomolecules that have emerged as negotiators with the immune system in the context of cancer and autoimmunity, and their inspirations on biohybrid NPs are introduced. Critical design considerations for efficient cross-talk between NPs and innate and adaptive immunity followed by hybridization methods are also discussed. Finally, clinical translation challenges and future perspectives regarding biohybrid NPs are discussed.


Subject(s)
Nanoparticles/chemistry , Animals , Gene Transfer Techniques , Humans , Immunity/drug effects , Nanoparticles/toxicity , Phagocytes/cytology , Phagocytes/drug effects , Precision Medicine , Tissue Distribution/drug effects
8.
ACS Nano ; 13(6): 6670-6688, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31117376

ABSTRACT

To dissect therapeutic mechanisms of transplanted stem cells and develop exosome-based nanotherapeutics in treating autoimmune and neurodegenerative diseases, we assessed the effect of exosomes secreted from human mesenchymal stem cells (MSCs) in treating multiple sclerosis using an experimental autoimmune encephalomyelitis (EAE) mouse model. We found that intravenous administration of exosomes produced by MSCs stimulated by IFNγ (IFNγ-Exo) (i) reduced the mean clinical score of EAE mice compared to PBS control, (ii) reduced demyelination, (iii) decreased neuroinflammation, and (iv) upregulated the number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords of EAE mice. Co-culture of IFNγ-Exo with activated peripheral blood mononuclear cells (PBMCs) cells in vitro reduced PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines including IL-6, IL-12p70, IL-17AF, and IL-22 yet increased levels of immunosuppressive cytokine indoleamine 2,3-dioxygenase. IFNγ-Exo could also induce Tregs in vitro in a murine splenocyte culture, likely mediated by a third-party accessory cell type. Further, IFNγ-Exo characterization by deep RNA sequencing suggested that IFNγ-Exo contains anti-inflammatory RNAs, where their inactivation partially hindered the exosomes potential to induce Tregs. Furthermore, we found that IFNγ-Exo harbors multiple anti-inflammatory and neuroprotective proteins. These results not only shed light on stem cell therapeutic mechanisms but also provide evidence that MSC-derived exosomes can potentially serve as cell-free therapies in creating a tolerogenic immune response to treat autoimmune and central nervous system disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Exosomes/transplantation , Mesenchymal Stem Cell Transplantation/methods , Animals , Cells, Cultured , Exosomes/metabolism , Female , Humans , Interferon-gamma/pharmacology , Interleukins/genetics , Interleukins/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Monocytes/immunology , T-Lymphocytes, Regulatory/immunology
9.
Cytokine Growth Factor Rev ; 43: 54-68, 2018 10.
Article in English | MEDLINE | ID: mdl-30170892

ABSTRACT

Cytokine therapies have emerged during the past decade as promising noninvasive treatments for heart disease. In general, current drug treatments are directed towards symptom control and prevention of disease progression; however, many agents also produce cause side effects that alter quality of life. Cytokine based therapies have the potential to reduce post-infarct heart failure and chronic ischemia by stimulating the proliferation and differentiation of endothelial cells and bone marrow hematopoietic stem cells and mobilizing these cells toward ischemic tissue. In turn, these mobilized cell populations contribute to myocardial regeneration. In contrast, over-expression of several cytokines has been linked to a variety of heart diseases; thus, therapies targeting and monitoring these cytokines are of great interest. Here we summarize results from clinical studies on cytokines as therapeutic agents or therapeutic targets in the treatment for heart disease as well as cytokines involved in the evolution of heart disease.


Subject(s)
Cytokines , Heart Diseases , Animals , Cytokines/immunology , Cytokines/therapeutic use , Heart Diseases/drug therapy , Heart Diseases/immunology , Humans
10.
Front Immunol ; 9: 1354, 2018.
Article in English | MEDLINE | ID: mdl-29963051

ABSTRACT

Recent advances on using immune and stem cells as two-pronged approaches for type 1 diabetes mellitus (T1DM) treatment show promise for advancement into clinical practice. As T1DM is thought to arise from autoimmune attack destroying pancreatic ß-cells, increasing treatments that use biologics and cells to manipulate the immune system are achieving better results in pre-clinical and clinical studies. Increasingly, focus has shifted from small molecule drugs that suppress the immune system nonspecifically to more complex biologics that show enhanced efficacy due to their selectivity for specific types of immune cells. Approaches that seek to inhibit only autoreactive effector T cells or enhance the suppressive regulatory T cell subset are showing remarkable promise. These modern immune interventions are also enabling the transplantation of pancreatic islets or ß-like cells derived from stem cells. While complete immune tolerance and body acceptance of grafted islets and cells is still challenging, bioengineering approaches that shield the implanted cells are also advancing. Integrating immunotherapy, stem cell-mediated ß-cell or islet production and bioengineering to interface with the patient is expected to lead to a durable cure or pave the way for a clinical solution for T1DM.

11.
Sci Rep ; 8(1): 4286, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523826

ABSTRACT

Despite preliminary confidence on biosafety of polymer coated iron oxide nanoparticles (SPIONs), toxicity concerns have hampered their clinical translation. SPIONs toxicity is known to be due to catalytic activity of their surface and release of toxic Fe ions originating from the core biodegradation, leading to the generation of reactive oxygen species (ROS). Here, we hypothesized that a double-layer polymeric corona comprising of dextran as an interior, and polyethylene glycol (PEG) as an exterior layer better shields the core SPIONs. We found that ROS generation was cell specific and depended on SPIONs concentration, although it was reduced by sufficient PEG immobilization or 100 µM deferoxamine. 24 h following injection, PEGylated samples showed reduction of biodistribution in liver, heterogenous biodistribution profile in spleen, and no influence on NPs blood retention. Sufficient surface masking or administration of deferoxamine could be beneficial strategies in designing and clinical translation of future biomedical SPIONs.


Subject(s)
Dextrans/chemistry , Iron/pharmacokinetics , Metal Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Animals , Cells, Cultured , Colloids/chemistry , Deferoxamine/pharmacology , Drug Liberation , Female , Ferric Compounds/chemistry , Iron/toxicity , Iron Chelating Agents/pharmacology , Liver/drug effects , Liver/metabolism , Metal Nanoparticles/adverse effects , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Spleen/drug effects , Spleen/metabolism , Tissue Distribution
12.
J Mater Chem B ; 5(22): 3995-4018, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-32264132

ABSTRACT

The last twenty years have witnessed great advances in biology, medicine, and materials science, leading to the development of various nanoparticle (NP)-mediated drug delivery systems. Innovation in materials science has led the generation of biodegradable, biocompatible, stimuli-responsive, and targeted delivery systems. However, currently available nanotherapeutic technologies are not efficient, which has culminated in the failure of their clinical trials. Despite huge efforts devoted to drug delivery nanotherapeutics, only a small amount of the injected material could reach the desired target. One promising strategy to enhance the efficiency of NP drug delivery is to hybridize multiple materials, where each component could play a critical role in an efficient multipurpose delivery system. This review aims to comprehensively cover different techniques, materials, advantages, and drawbacks of various systems to develop hybrid nano-vesicles for drug delivery. Attention is finally given to the hybridization benefits in overcoming the biological barriers for drug delivery. It is believed that the advent of modern nano-formulations for multifunctional hybrid carriers paves the way for future advances to achieve more efficient drug delivery systems.

SELECTION OF CITATIONS
SEARCH DETAIL
...