Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Immunol Rev ; 313(1): 91-103, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36258635

RESUMEN

A number of clinical treatment modalities involve contact between blood and biomaterials: these include extracorporeal circuits such as hemodialysis, cardiopulmonary bypass, plasmapheresis, and intravascular treatments. Common side effects arising from these treatments are caused by activation of the cascade systems of the blood. Many of these side effects are mediated via the complement system, including thromboinflammatory reactions and rejection of implants. Depending on the composition of the materials, complement activation is triggered via all the activation pathways but is by far mostly driven by the alternative pathway amplification loop. On biomaterial surfaces the alternative pathway amplification is totally unregulated and leads under optimal conditions to deposition of complement fragments, mostly C3b, on the surface leading to a total masking of the underlying surface. In this review, we discuss the mechanism of the complement activation, clinical consequences of the activation, and potential strategies for therapeutic regulation of the activation, using hemodialysis as demonstrator.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Humanos , Vía Alternativa del Complemento
2.
Sci Technol Adv Mater ; 23(1): 845-857, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36518982

RESUMEN

Liposome surface coating has been studied to avoid the immunological responses caused by the complement system, and alternative materials to poly(ethylene glycol) (PEG) have been explored recently since the production of anti-PEG IgM antibodies has been found in humans. We previously reported a liposome coating with poly(2-methacryloyloxyethyl phosphorylcholine) (poly(MPC))-conjugated lipids (PMPC-lipids) and demonstrated its protective effect on blood protein interactions. Here, we attempted to modify the liposome surface by exogenously adding PMPC-lipids, which were spontaneously incorporated into the outer membrane via hydrophobic interactions. The polymerization degree of the PMPC segment was regulated from 10 to 100. The incorporated ratio of PMPC-lipid increased with a decrease in the degree of PMPC polymerization. Due to surface modification with PMPC-lipids, increase in the length of the PMPC-chain increased the size of the liposomes. The modified liposomes were kept stable for 14 d in terms of their size, polydispersity, and surface properties, where approximately 70% of PMPC-lipids were incorporated into the liposome surface. We demonstrated that liposome surface modification with PMPC-lipids can inhibit protein adsorption when exposed to serum, regardless of the degree of polymerization of PMPC. In addition, the PMPC-lipid modified surface was not recognized by the anti-PEG IgM antibody, whereas PEG-lipid was recognized by the antibody. Thus, we successfully fabricated an inert liposome surface via spontaneous modification with PMPC-lipids, where only the outer bilayer surface was modified. This technique can be available for full loading of water-soluble active pharmaceutical ingredient inside the modified liposome.

3.
Langmuir ; 37(32): 9711-9723, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34342462

RESUMEN

The use of amphiphilic molecules such as poly(ethylene glycol)-conjugated phospholipid (PEG-lipid) enables incorporation into liposome surfaces by exogenous addition as a result of the self-assembly with lipids. This technique can be applicable for manipulation of both liposomes and cells. In this study, we aimed to characterize Tat peptide (YGRKKRRQRRR)-conjugated PEG-lipids when used to exogenously surface modify liposomes (size: ca. 100 nm). We earlier reported that cells, which were surface modified with Tat peptides conjugated to PEG-lipids could attach spontaneously to material surfaces without any chemical modification. Here, we synthesized different types of Tat-PEG-lipids by combining PEG of different molecular weights (5 and 40 kDa) with different lipids with three acyl chains (myristoyl, palmitoyl, and stearoyl, respectively) and then studied the spontaneous adsorption of modified liposomes onto a substrate surface induced by the different Tat-PEG-lipids. The amount of adsorbed liposomes strongly depended on the number of incorporated Tat-PEG-lipid moieties: a decrease in both the PEG and the acyl chain lengths led to adsorption of higher amounts of liposomes. Furthermore, when a collagenase-cleavable amino acid sequence was inserted between the Tat sequence and the PEG segment, adsorbed liposomes could be harvested from the substrate by collagenase treatment with no difference in desorption efficiency between the different Tat-PEG-lipids. Thus, Tat-PEG-lipid can be a suitable tool for the manipulation of liposomes and cells.


Asunto(s)
Péptidos de Penetración Celular , Liposomas , Adsorción , Humanos , Fosfolípidos , Polietilenglicoles
4.
Biomacromolecules ; 22(5): 1980-1989, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33813822

RESUMEN

Mesenchymal stem/stromal cells (MSCs) evoke great excitement for treating different human diseases due to their ability to home inflamed tissues, suppress inflammation, and promote tissue regeneration. Despite great promises, clinical trial results are disappointing as allotransplantation of MSCs trigger thrombotic activity and are damaged by the complement system, compromising their survival and function. To overcome this, a new strategy is presented by the silencing of tissue factor (TF), a transmembrane protein that mediates procoagulant activity. Novel Pluronic-based micelles are designed with the pendant pyridyl disulfide group, which are used to conjugate TF-targeting siRNA by the thiol-exchange reaction. This nanocarrier design effectively delivered the payload to MSCs resulting in ∼72% TF knockdown (KD) without significant cytotoxicity. Hematological evaluation of MSCs and TF-KD MSCs in an ex vivo human whole blood model revealed a significant reduction in an instant-blood-mediated-inflammatory reaction as evidenced by reduced platelet aggregation (93% of free platelets in the TF-KD group, compared to 22% in untreated bone marrow-derived MSCs) and thrombin-antithrombin complex formation. Effective TF silencing induced higher MSC differentiation in osteogenic and adipogenic media and showed stronger paracrine suppression of proinflammatory cytokines in macrophages and higher stimulation in the presence of endotoxins. Thus, TF silencing can produce functional cells with higher fidelity, efficacy, and functions.


Asunto(s)
Células Madre Mesenquimatosas , Diferenciación Celular , Células Cultivadas , Humanos , Micelas , Comunicación Paracrina , Poloxámero , Tromboplastina/genética
5.
Langmuir ; 36(41): 12088-12106, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32927948

RESUMEN

The regulation of the cellular surface with biomaterials can contribute to the progress of biomedical applications. In particular, the cell surface is exposed to immunological surveillance and reactions in transplantation therapy, and modulation of cell surface properties might improve transplantation outcomes. The transplantation of therapeutic cells, tissue, and organs is an effective and fundamental treatment and has contributed to saving lives and improving quality of life. Because of shortages, donor cells, tissues, and organs are carefully transplanted with the goal of retaining activity and viability. However, some issues remain to be resolved in terms of reducing side effects, improving graft survival, managing innate and adaptive immune responses, and improving transplant storage and procedures. Given that the transplantation process involves multiple steps and is technically complicated, an engineering approach together with medical approaches to resolving these issues could enhance success. In particular, cell surface engineering with biocompatible polymers looks promising for improving transplantation therapy and has potential for other biomedical applications. Here we review the significance of polymer-based surface modification of cells and organs for biomedical applications, focusing on the following three topics: Cell protection: cellular protection through local immune regulation using cell surface modification with biocompatible polymers. This protection could extend to preventing attack by the host immune system, freeing recipients from taking immunosuppressive drugs, and avoiding a second transplantation. Cell attachment: cell manipulation, which is an important technique for delivery of therapeutic cells and their alignment for recellularization of decellularized tissues and organs in regenerative therapy. Cell fusion: fusion of different cells, which can lead to the formation of new functional cells that could be useful for generating, e.g., immunologically competent or metabolically active cells.


Asunto(s)
Polímeros , Calidad de Vida , Materiales Biocompatibles , Propiedades de Superficie , Ingeniería de Tejidos
6.
Semin Immunol ; 28(3): 268-77, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27211838

RESUMEN

Therapeutic medicine today includes a vast number of procedures involving the use of biomaterials, transplantation of therapeutic cells or cell clusters, as well as of solid organs. These treatment modalities are obviously of great benefit to the patient, but also present a great challenge to the innate immune system, since they involve direct exposure of non-biological materials, cells of non-hematological origin as well as endothelial cells, damaged by ischemia-perfusion in solid organs to proteins and cells in the blood. The result of such an exposure may be an inappropriate activation of the complement and contact/kallikrein systems, which produce mediators capable of triggering the platelets and PMNs and monocytes, which can ultimately result in thrombotic and inflammatory (i.e., a thrombo-inflammatory) response to the treatment modality. In this concept review, we give an overview of the mechanisms of recognition within the innate immunity system, with the aim to identify suitable points for intervention. Finally, we discuss emerging and promising techniques for surface modification of biomaterials and cells with specific inhibitors in order to diminish thromboinflammation and improve clinical outcome.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Plaquetas/inmunología , Proteínas del Sistema Complemento/metabolismo , Inmunoterapia/métodos , Inflamación/terapia , Trombocitosis/terapia , Materiales Biocompatibles/efectos adversos , Activación de Complemento , Humanos , Inmunidad Innata , Inflamación/inmunología , Terapia Molecular Dirigida , Trombocitosis/inmunología
7.
Immunol Rev ; 274(1): 245-269, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782319

RESUMEN

Innate immunity is fundamental to our defense against microorganisms. Physiologically, the intravascular innate immune system acts as a purging system that identifies and removes foreign substances leading to thromboinflammatory responses, tissue remodeling, and repair. It is also a key contributor to the adverse effects observed in many diseases and therapies involving biomaterials and therapeutic cells/organs. The intravascular innate immune system consists of the cascade systems of the blood (the complement, contact, coagulation, and fibrinolytic systems), the blood cells (polymorphonuclear cells, monocytes, platelets), and the endothelial cell lining of the vessels. Activation of the intravascular innate immune system in vivo leads to thromboinflammation that can be activated by several of the system's pathways and that initiates repair after tissue damage and leads to adverse reactions in several disorders and treatment modalities. In this review, we summarize the current knowledge in the field and discuss the obstacles that exist in order to study the cross-talk between the components of the intravascular innate immune system. These include the use of purified in vitro systems, animal models and various types of anticoagulants. In order to avoid some of these obstacles we have developed specialized human whole blood models that allow investigation of the cross-talk between the various cascade systems and the blood cells. We in particular stress that platelets are involved in these interactions and that the lectin pathway of the complement system is an emerging part of innate immunity that interacts with the contact/coagulation system. Understanding the resulting thromboinflammation will allow development of new therapeutic modalities.


Asunto(s)
Plaquetas/inmunología , Proteínas del Sistema Complemento/metabolismo , Células Endoteliales/fisiología , Inflamación/inmunología , Trombosis/inmunología , Animales , Coagulación Sanguínea , Homeostasis , Humanos , Inmunidad Innata , Calicreínas/metabolismo , Cininas/metabolismo
8.
Sci Technol Adv Mater ; 20(1): 688-698, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31275460

RESUMEN

In this review article, we focus on activation of the soluble components of the innate immune system triggered by nonbiological compounds and stress variances in activation due to the difference in size between nanoparticles (NPs) and larger particles or bulk material of the same chemical and physical composition. We then discuss the impact of the so-called protein corona which is formed on the surface of NPs when they come in contact with blood or other body fluids. For example, NPs which bind inert proteins, proteins which are prone to activate the contact system (e.g., factor XII), which may lead to clotting and fibrin formation or the complement system (e.g., IgG or C3), which may result in inflammation and vascular damage. Furthermore, we describe a whole blood model which we have developed to monitor activation and interaction between different components of innate immunity: blood protein cascade systems, platelets, leukocytes, cytokine generation, which are induced by NPs. Finally, we describe our own studies on innate immunity system activation induced by three fundamentally different species of NPs (two types of engineered NPs and diesel NPs) as demonstrator of the utility of an initial determination of the composition of the protein corona formed on NPs exposed to ethylenediaminetetraacetic acid (EDTA) plasma and subsequent analysis in our whole blood model.

9.
Sci Technol Adv Mater ; 17(1): 677-684, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27877914

RESUMEN

We studied real-time interaction between poly(ethylene glycol)-conjugated phospholipids (PEG-lipids) and a supported lipid membrane by surface plasmon resonance (SPR) spectroscopy to understand dynamic behaviors of PEG-lipids on living cell membranes. Supported lipid membranes formed on a hydrophobic surface were employed as a model of living cell membrane. We prepared three kinds of PEG-lipids that carried alkyl chains of different lengths for SPR measurements and also performed fluorescence recovery after photobleaching (FRAP) to study the influence of acyl chain length on dynamics on the supported membrane. PEG-lipids were uniformly anchored to lipid membranes with high fluidity without clustering. Incorporation and dissociation rates of PEG-lipids into supported membranes strongly depended on the length of acyl chains; longer acyl chains reduced the incorporation rate and the dissociation rate of PEG-lipid. Furthermore, protein adsorption experiment with bovine serum albumin indicated that PEG modification prevented the adsorption of bovine serum albumin on such supported membrane.

10.
Sci Technol Adv Mater ; 17(1): 807-815, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27933120

RESUMEN

Since circulating tumor cells (CTCs) are tumor cells which are found in the blood of cancer patients, CTCs are potential tumor markers, so a rapid isolation of CTCs is desirable for clinical applications. In this paper, a three-dimensional polystyrene (PS) microfiber fabric with vacuum aspiration system was developed for capturing CTCs within a short time. Various microfiber fabrics with different diameters were prepared by the electrospinning method and optimized for contact frequency with cells. Vacuum aspiration utilizing these microfiber fabrics could filter all cells within seconds without mechanical damage. The microfiber fabric with immobilized anti-EpCAM antibodies was able to specifically capture MCF-7 cells that express EpCAM on their surfaces. The specificity of the system was confirmed by monitoring the ability to isolate MCF-7 cells from a mixture containing CCRF-CEM cells that do not express EpCAM. Furthermore, the selective capture ability of the microfiber was retained even when the microfiber was exposed to the whole blood of pigs spiked with MCF-7 cells. The specific cell capture ratio of the vacuum aspiration system utilizing microfiber fabric could be improved by increasing the thickness of the microfiber fabric through electrospinning time.

11.
Stem Cells ; 32(9): 2430-42, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24805247

RESUMEN

We have recently reported that therapeutic mesenchymal stromal cells (MSCs) have low engraftment and trigger the instant blood mediated inflammatory reaction (IBMIR) after systemic delivery to patients, resulting in compromised cell function. In order to optimize the product, we compared the immunomodulatory, blood regulatory, and therapeutic properties of freeze-thawed and freshly harvested cells. We found that freeze-thawed MSCs, as opposed to cells harvested from continuous cultures, have impaired immunomodulatory and blood regulatory properties. Freeze-thawed MSCs demonstrated reduced responsiveness to proinflammatory stimuli, an impaired production of anti-inflammatory mediators, increased triggering of the IBMIR, and a strong activation of the complement cascade compared to fresh cells. This resulted in twice the efficiency in lysis of thawed MSCs after 1 hour of serum exposure. We found a 50% and 80% reduction in viable cells with freshly detached as opposed to thawed in vitro cells, indicating a small benefit for fresh cells. In evaluation of clinical response, we report a trend that fresh cells, and cells of low passage, demonstrate improved clinical outcome. Patients treated with freshly harvested cells in low passage had a 100% response rate, twice the response rate of 50% observed in a comparable group of patients treated with freeze-thawed cells at higher passage. We conclude that cryobanked MSCs have reduced immunomodulatory and blood regulatory properties directly after thawing, resulting in faster complement-mediated elimination after blood exposure. These changes seem to be paired by differences in therapeutic efficacy in treatment of immune ailments after hematopoietic stem cell transplantation.


Asunto(s)
Criopreservación/métodos , Inmunoterapia/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Adolescente , Adulto , Anciano , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Inmunomodulación , Inmunofenotipificación/métodos , Lactante , Masculino , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Adulto Joven
12.
Adv Exp Med Biol ; 865: 189-209, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26306451

RESUMEN

Transplantation of the pancreatic islets of Langerhans (islets) is a promising cell therapy for treating insulin-dependent type 1 diabetes mellitus. Islet transplantation is a minimally-invasive technique involving relatively simple surgery. However, after intraportal transplantation, the transplanted islets are attacked by the recipient's immune system, because they activate a number of systems, including coagulation, complement response, inflammation, immune rejection, and recurrence of autoimmune disease. We have developed a surface modification and microencapsulation technique that protects cells and islets with biomaterials and bioactive substances, which may be useful in clinical settings. This approach employs amphiphilic polymers, which can interact with lipid bilayer membranes, without increasing cell volume. Molecules attached to these polymers can protect transplanted cells and islets from attack by the host immune system. We expect that this surface modification technique will improve graft survival in clinical islet transplantation.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Rechazo de Injerto/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/química , Membrana Dobles de Lípidos/química , Polietilenglicoles/química , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Supervivencia de Injerto , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/inmunología , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Isogénico , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/inmunología
13.
Adv Exp Med Biol ; 865: 3-17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26306440

RESUMEN

Thromboinflammation is primarily triggered by the humoral innate immune system, which mainly consists of the cascade systems of the blood, i.e., the complement, contact/coagulation and fibrinolytic systems. Activation of these systems subsequently induces activation of endothelial cells, leukocytes and platelets, finally resulting in thrombotic and inflammatory reactions. Such reactions are triggered by a number of medical procedures, e.g., treatment with biomaterials or drug delivery devices as well as in transplantation with cells, cell clusters or whole vascularized organs. Here, we (1) describe basic mechanisms for thromboinflammation; (2) review thromboinflammatory reactions in therapeutic medicine; and (3) discuss emerging strategies to dampen thromboinflammation.


Asunto(s)
Anticoagulantes/uso terapéutico , Rechazo de Injerto/prevención & control , Factores Inmunológicos/uso terapéutico , Trombosis/prevención & control , Trasplante de Tejidos , Materiales Biocompatibles/efectos adversos , Coagulación Sanguínea/efectos de los fármacos , Factores de Coagulación Sanguínea/inmunología , Factores de Coagulación Sanguínea/metabolismo , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Leucocitos/citología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Trombosis/inmunología , Trombosis/patología
14.
Biomacromolecules ; 15(6): 2012-8, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24761752

RESUMEN

In this study, we fabricated lectin-tagged fluorescent polymeric nanoparticles approximately 35 nm in diameter using biocompatible polymers conjugated with lectins for the purpose of detecting sialic acid on a living cell surface, which is one of the most important biomarkers for cancer diagnosis. Through cellular experiments, we successfully detected sialic acid overexpression on cancerous cells with high specificity. These fluorescent polymeric nanoparticles can be useful as a potential bioimaging probe for detecting diseased cells.


Asunto(s)
Membrana Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Colorantes Fluorescentes/administración & dosificación , Ácido N-Acetilneuramínico , Nanopartículas/administración & dosificación , Lectinas de Plantas/administración & dosificación , Polímeros/administración & dosificación , Animales , Membrana Celular/metabolismo , Colorantes Fluorescentes/metabolismo , Células HeLa , Humanos , Células MCF-7 , Ratones , Ácido N-Acetilneuramínico/metabolismo , Nanopartículas/metabolismo , Corteza de la Planta , Extractos Vegetales/administración & dosificación , Extractos Vegetales/metabolismo , Lectinas de Plantas/metabolismo , Polímeros/metabolismo , Sambucus nigra
15.
Sci Rep ; 14(1): 15340, 2024 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961142

RESUMEN

Although stem cell-based regenerative medicine has been extensively studied, it remains difficult to reconstruct three dimensional tissues and organs in combination with vascular systems in vitro. One clinically successful therapy is transplantation of mesenchymal stem cells (MSC) into patients with graft versus host disease. However, transplanted cells are immediately damaged and destroyed because of innate immune reactions provoked by thrombogenic inflammation, and patients need to take immunosuppressive drugs for the immunological regulation of allogeneic cells. This reduces the benefits of stem cell transplantation. Therefore, alternative therapies are more realistic options for clinical use. In this study, we aimed to take advantage of the therapeutic efficacy of MSC and use multiple cytokines released from MSC, that is, stem cells from human exfoliated deciduous teeth (SHEDs). Here, we purified components from conditioned media of immortalized SHED (IM-SHED-CM) and evaluated the activities of intracellular dehydrogenase, cell migration, and antioxidative stress by studying the cells. The immortalization of SHED could make the stable supply of CM possible. We found that the fractionated component of 50-100 kD from IM-SHED-CM had higher efficacy than the original IM-SHED-CM in terms of intracellular dehydrogenase and cell migration in which intracellular signal transduction was activated via receptor tyrosine kinases, and the glutathione peroxidase and reductase system was highly active. Although antioxidative stress activities in the fractionated component of 50-100 kD had slightly lower than that of original IM-SHE-CM, the fraction still had the activity. Thus, the use of fractionated components of 50-100 kD from IM-SHED-CM could be an alternative choice for MSC transplantation because the purified components from CM could maintain the effect of cytokines from SHED.


Asunto(s)
Movimiento Celular , Células Madre Mesenquimatosas , Estrés Oxidativo , Diente Primario , Humanos , Diente Primario/citología , Diente Primario/metabolismo , Movimiento Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Células Cultivadas , Antioxidantes/farmacología , Antioxidantes/metabolismo , Células Madre/metabolismo , Células Madre/citología , Transducción de Señal/efectos de los fármacos
16.
Stem Cells ; 30(7): 1565-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22522999

RESUMEN

Multipotent mesenchymal stromal cells (MSCs) are tested in numerous clinical trials. Questions have been raised concerning fate and function of these therapeutic cells after systemic infusion. We therefore asked whether culture-expanded human MSCs elicit an innate immune attack, termed instant blood-mediated inflammatory reaction (IBMIR), which has previously been shown to compromise the survival and function of systemically infused islet cells and hepatocytes. We found that MSCs expressed hemostatic regulators similar to those produced by endothelial cells but displayed higher amounts of prothrombotic tissue/stromal factors on their surface, which triggered the IBMIR after blood exposure, as characterized by formation of blood activation markers. This process was dependent on the cell dose, the choice of MSC donor, and particularly the cell-passage number. Short-term expanded MSCs triggered only weak blood responses in vitro, whereas extended culture and coculture with activated lymphocytes increased their prothrombotic properties. After systemic infusion to patients, we found increased formation of blood activation markers, but no formation of hyperfibrinolysis marker D-dimer or acute-phase reactants with the currently applied dose of 1.0-3.0 × 10(6) cells per kilogram. Culture-expanded MSCs trigger the IBMIR in vitro and in vivo. Induction of IBMIR is dose-dependent and increases after prolonged ex vivo expansion. Currently applied doses of low-passage clinical-grade MSCs elicit only minor systemic effects, but higher cell doses and particularly higher passage cells should be handled with care. This deleterious reaction can compromise the survival, engraftment, and function of these therapeutic cells.


Asunto(s)
Células Madre Mesenquimatosas/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Células Endoteliales/citología , Humanos , Células Madre Mesenquimatosas/inmunología , Trombosis/sangre
17.
J Mater Chem B ; 11(46): 11121-11134, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-37953734

RESUMEN

Surface modification with heparin is a powerful biomaterial coating strategy that protects against innate immunity activation since heparin is a part of the proteoglycan heparan sulfate on cell surfaces in the body. We studied the heparinization of cellular and material surfaces via lipid conjugation to a heparin-binding peptide. In the present study, we synthesized fragmented heparin (fHep)-conjugated phospholipids and studied their regulation of the innate immune system on a lipid bilayered surface using liposomes. Liposomes have versatile applications, such as drug-delivery systems, due to their ability to carry a wide range of molecules. Owing to their morphological similarity to cell membranes, they can also be used to mimic a simple cell-membrane to study protein-lipid interactions. We investigated the interaction of complement-regulators, factor H and C4b-binding protein (C4BP), as well as the coagulation inhibitor antithrombin (AT), with fHep-lipids on the liposomal surface. Herein, we studied the ability of fHep-lipids to recruit factor H, C4BP, and AT using a quartz crystal microbalance with dissipation monitoring. With dynamic light scattering, we demonstrated that liposomes could be modified with fHep-lipids and were stable up to 60 days at 4 °C. Using a capillary western blot-based method (Wes), we showed that fHep-liposomes could recruit factor H in a model system using purified proteins and assist in the degradation of the active complement protein C3b to iC3b. Furthermore, we found that fHep-liposomes could recruit factor H and AT from human plasma. Therefore, the use of fHep-lipids could be a potential coating for liposomes and cell surfaces to regulate the immune system on the lipid surface.


Asunto(s)
Heparina , Liposomas , Humanos , Liposomas/química , Factor H de Complemento , Membrana Dobles de Lípidos , Antitrombinas/farmacología , Anticoagulantes , Sistema Inmunológico/metabolismo
18.
Biomater Adv ; 147: 213331, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36773382

RESUMEN

Mesenchymal stem cells (MSCs) therapy is a promising approach for treating inflammatory diseases due to their immunosuppressive and tissue repair characteristics. However, allogenic transplantation of MSCs induces thrombotic complications in some patients which limits its potential for clinical translation. To address this challenge, we have exploited the bioactivity of heparin, a well-known anticoagulant and immunosuppressive polysaccharide that is widely used in clinics. We have developed a smart layer-by-layer (LbL) coating strategy using gelatin and heparin polymers exploiting their overall positive and negative charges that enabled efficient complexation with the MSCs' glycocalyx. The stable coating of MSCs suppressed complement attack and mitigated thrombotic activation as demonstrated in human whole blood. Gratifyingly, the MSC coating retained its immunosuppressive properties and differentiation potential when exposed to inflammatory conditions and differentiation factors. We believe the simple coating procedure of MSCs will increase allogenic tolerance and circumvent the major challenge of MSCs transplantation.


Asunto(s)
Biomimética , Células Madre Mesenquimatosas , Humanos , Polielectrolitos , Heparina , Diferenciación Celular , Inmunosupresores
19.
Regen Ther ; 22: 90-98, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36712957

RESUMEN

Exosomes (diameter 30-200 nm) are a subtype of extracellular vesicles secreted by cells containing DNA, microRNA (miRNA), and proteins. Exosomes are expected to be valuable as a means of delivering drugs or functional miRNAs in treatment of diseases. However, the delivery of exosomes is not sufficiently effective, even though exosomes have intrinsic delivery functions. Cell-penetrating peptides (CPPs) are short peptide families that facilitate cellular intake of molecules and vesicles. We previously reported that the modification of cells, and liposomes with CPP-conjugated-lipids, CPPs conjugated with poly (ethylene glycol)-conjugated phospholipids (PEG-lipid), that induce adhesion by CPPs, can be useful for cell-based assays and harvesting liposomes. In this study, we aimed to modulate the exosome surface using Tat peptide (YGRKKRRQRRR)-PEG-lipids to improve intracellular delivery to endothelial cells. We isolated and characterized exosomes from the medium of HEK 293 T cell cultures. Tat conjugated PEG-lipids with different spacer molecular weights and lipid types were incorporated into exosomes using fluorescein isothiocyanate labeling to optimize the number of Tat-PEG-lipids immobilized on the exosome surface. The exosomes modified with Tat-PEG-lipids were incubated with human umbilical vein endothelial cells (HUVECs) to study the interaction. Tat conjugated with 5 kDa PEG and C16 lipids incorporated on the exosome surface were highly detected inside HUVECs by flow cytometry. Fluorescence was negligible in HUVECs for control groups. Thus, Tat-PEG-lipids can be modified on the exosome surface, improving the intracellular delivery of exosomes.

20.
J Mater Chem B ; 10(14): 2561-2569, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-34878485

RESUMEN

The effects of differentiated cells on stem cell differentiation were analyzed via co-culturing using a cell-encapsulated double-layered hydrogel system. As a polymer hydrogel matrix, a water-soluble zwitterionic polymer having both a 2-methacryloyloxyethyl phosphorylcholine unit and a p-vinylphenylboronic acid unit (PMBV), was complexed spontaneously with poly(vinyl alcohol) (PVA) under mild cell culture conditions. The creep modulus of the hydrogel was controlled by changing the composition of the polymer in the solution. Mouse mesenchymal stem cells (MSCs), C3H10T1/2 cells, were encapsulated into PMBV/PVA hydrogels and cultured. In the PMBV/PVA hydrogel with a lower creep modulus (0.40 kPa), proliferation of C3H10T1/2 cells occurred, and the formation of cell aggregates was observed. On the other hand, a higher creep modulus (1.7 kPa) of the hydrogel matrix prevented cell proliferation. Culturing C3H10T1/2 cells encapsulated in the PMBV/PVA hydrogel in the presence of bone morphogenetic protein-2 increased the activity of intracellular alkaline phosphatase (ALP). This indicated that C3H10T1/2 cells differentiated into mature osteoblasts. When the C3H10T1/2 cells encapsulated in the PMBV/PVA hydrogel were cultured in combination with the mature osteoblasts in the hydrogel by a close contacting double-layered hydrogel structure, higher ALP activity was observed compared with the cells cultured separately. It was considered that the differentiation of C3H10T1/2 cells in the hydrogel layer was induced by cytokines diffused from mature osteoblasts encapsulated in another hydrogel layer. It could be concluded that the PMBV/PVA hydrogel system provides a good way to observe the effects of the surrounding cells on cell function in three-dimensional culture.


Asunto(s)
Hidrogeles , Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Hidrogeles/química , Hidrogeles/farmacología , Metacrilatos , Ratones , Fosforilcolina/análogos & derivados , Polímeros/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda