Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
ACS Biomater Sci Eng ; 10(5): 2911-2924, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38657240

RESUMEN

Macrophage uptake of nanoparticles is highly dependent on the physicochemical characteristics of those nanoparticles. Here, we have created a collection of lipid-polymer nanoparticles (LPNPs) varying in size, stiffness, and lipid makeup to determine the effects of these factors on uptake in murine bone marrow-derived macrophages. The LPNPs varied in diameter from 232 to 812 nm, in storage modulus from 21.2 to 287 kPa, and in phosphatidylserine content from 0 to 20%. Stiff, large nanoparticles with a coating containing phosphatidylserine were taken up by macrophages to a much higher degree than any other formulation (between 9.3× and 166× higher than other LPNPs). LPNPs with phosphatidylserine were taken up most by M2-polarized macrophages, while those without were taken up most by M1-polarized macrophages. Differences in total LPNP uptake were not dependent on endocytosis pathway(s) other than phagocytosis. This work acts as a basis for understanding how the interactions between nanoparticle physicochemical characteristics may act synergistically to facilitate particle uptake.


Asunto(s)
Lípidos , Macrófagos , Nanopartículas , Polímeros , Nanopartículas/química , Animales , Macrófagos/metabolismo , Ratones , Polímeros/química , Polímeros/metabolismo , Lípidos/química , Tamaño de la Partícula , Fagocitosis , Endocitosis , Fosfatidilserinas/metabolismo , Fosfatidilserinas/química
2.
J Biomed Mater Res A ; 112(9): 1388-1398, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38270241

RESUMEN

Stem cell therapy and skin substitutes address the stalled healing of chronic wounds in order to promote wound closure; however, the high cost and regulatory hurdles of these treatments limit patient access. A low-cost method to induce bioactive healing has the potential to substantially improve patient care and prevent wound-induced limb loss. A previous study reported that bioactive factors derived from apoptotic-like mesenchymal stem cells (MSCs) demonstrated anti-inflammatory and proangiogenic effects and improved ischemic muscle regeneration. In this work, these MSC-derived bioactive factors were loaded into a hydrogel foam to harness immunomodulatory and angiogenic properties from MSC components to facilitate chronic wound healing without the high cost and translational challenges of cell therapies. After incorporation of bioactive factors, the hydrogel foam retained high absorbency, moisture retention, and target water vapor transmission rate. High loading efficiency was confirmed and release studies indicated that over 90% of loaded factors were released within 24 h. Ethylene oxide sterilization and 4-week storage did not affect the bioactive factor release profile or physical properties of the hydrogel foam dressing. Bioactivity retention of the released factors was also confirmed for as-sterilized, 4°C-stored, and -20°C-stored bioactive hydrogel foams as determined by relevant gene expression levels in treated pro-inflammatory (M1) macrophages. These results support the use of the bioactive dressings as an off-the-shelf product. Overall, this work reports a new method to achieve a first-line wound dressing with the potential to reduce persistent inflammation and promote angiogenesis in chronic wounds.


Asunto(s)
Vendajes , Hidrogeles , Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Hidrogeles/química , Hidrogeles/farmacología , Animales , Humanos , Ratones , Inductores de la Angiogénesis/farmacología , Cicatrización de Heridas/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factores Inmunológicos/farmacología
3.
Cytotherapy ; 25(4): 375-386, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36543717

RESUMEN

BACKGROUND AIMS: Skeletal muscle regeneration after severe damage is reliant on local stem cell proliferation and differentiation, processes that are tightly regulated by macrophages. Peripheral artery disease is a globally prevalent cardiovascular disease affecting millions of people. Progression of the disease leads to intermittent claudication, subsequent critical limb ischemia and muscle injury. Tissue-derived and ex vivo-expanded mesenchymal stromal cells (MSCs) for skeletal muscle regeneration have been studied, but pre-clinical and clinical results have not been consistent. As a result, the potential therapeutic efficacy and associated repair mechanisms of MSCs remain unclear. Numerous studies have demonstrated the vulnerability of delivered MSCs, with a precipitous drop in cell viability upon transplantation. This has prompted investigation into the therapeutic benefit of apoptotic cells, microvesicles, exosomes and soluble signals that are released upon cell death. METHODS: In this study, we characterized various components produced by MSCs after cell death induction under different conditions. We discovered anti-inflammatory and pro-regenerative effects produced by cell components following a freeze and thaw (F&T) process on macrophage polarization in vitro. We further investigated the underlying mechanisms of macrophage polarization by those components resulting from severe cell death induction. RESULTS: We found potent therapeutic effects from F&T-induced cell debris are dependent on the externalization of phosphatidylserine on the plasma membrane. In contrast, effects from the supernatant of F&T-induced cell death primarily depends on the released protein content. We then applied the F&T-induced cell supernatant to an animal model of peripheral artery disease to treat muscle injury caused by severe ischemia. Treatment with the F&T supernatant but not the vulnerable MSCs resulted in significantly improved recovery of muscle function, blood flow and morphology and inflammation resolution in the affected muscles 2 weeks after injury. CONCLUSIONS: This study validates the therapeutic potential of F&T-induced supernatant obviating the need for a viable population from vulnerable MSCs to treat injury, thus providing a roadmap for cell-free therapeutic approaches for tissue regeneration.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Enfermedad Arterial Periférica , Animales , Inflamación/terapia , Inflamación/metabolismo , Isquemia/terapia , Enfermedad Arterial Periférica/terapia , Músculos , Trasplante de Células Madre Mesenquimatosas/métodos
4.
Int J Pharm ; 618: 121634, 2022 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-35247497

RESUMEN

Chronic inflammation is a significant pathological process found in a range of disease states. Treatments to reduce inflammation in this family of diseases may improve symptoms and disease progression, but are largely limited by variable response rates, cost, and off-target effects. Macrophages are implicated in many inflammatory diseases for their critical role in the maintenance and resolution of inflammation. Macrophages exhibit significant plasticity to direct the inflammatory response by taking on an array of pro- and anti-inflammatory phenotypes based on extracellular cues. In this work, a nanoparticle has been developed to target sites of inflammation and reduce the inflammatory macrophage phenotype by mimicking the anti-inflammatory effect of apoptotic cell engulfment. The nanoparticle, comprised of a poly(lactide-co-glycolide) core, is coated with phosphatidylserine (PS)-supplemented cell plasma membrane to emulate key characteristics of the apoptotic cell surface. The particle surface is additionally functionalized with an acid-sensitive sheddable polyethylene glycol (PEG) moiety to increase the delivery of the nanoparticles to low pH environments such as those of chronic inflammation. In a mouse model of lipopolysaccharide-induced inflammation, particles were preferentially taken up by macrophages at the site and promoted an anti-inflammatory phenotype shift. This PEGylated membrane coating increased the delivery of nanoparticles to sites of inflammation and may be used as a tool alone or as a delivery scheme for additional cargo to reduce macrophage-associated inflammatory response.


Asunto(s)
Inflamación , Nanopartículas , Animales , Antiinflamatorios/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Macrófagos , Ratones , Fenotipo
5.
Tissue Eng Part B Rev ; 28(2): 279-294, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33528306

RESUMEN

Inflammation is a crucial part of wound healing and pathogen clearance. However, it can also play a role in exacerbating chronic diseases and cancer progression when not regulated properly. A subset of current innate immune engineering research is focused on how molecules such as lipids, proteins, and nucleic acids native to a healthy inflammatory response can be harnessed in the context of biomaterial design to promote healing, decrease disease severity, and prolong survival. The engineered biomaterials in this review inhibit inflammation by releasing anti-inflammatory cytokines, sequestering proinflammatory cytokines, and promoting phenotype switching of macrophages in chronic inflammatory disease models. Conversely, other biomaterials discussed here promote inflammation by mimicking pathogen invasion to inhibit tumor growth in cancer models. The form that these biomaterials take spans a spectrum from nanoparticles to large-scale hydrogels to surface coatings on medical devices. Cell-inspired molecules have been incorporated in a variety of creative ways, including loaded into or onto the surface of biomaterials or used as the biomaterials themselves. Impact statement Chronic inflammatory diseases and cancers are widespread health care concerns. Treatment plans for these diseases can be complicated and the outcomes are often mixed due to off-target effects. Current research efforts in immune engineering and biomaterials are focused on utilizing the body's native immune response to return to homeostasis as a therapeutic approach. This review collects many of the most current findings in the field as a resource for future research.


Asunto(s)
Materiales Biocompatibles , Neoplasias , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Citocinas , Humanos , Inflamación , Macrófagos/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia
6.
ACS Biomater Sci Eng ; 7(6): 2605-2614, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33949850

RESUMEN

Hydrogels made from self-assembling peptides have significant advantages in tissue engineering, namely a biocompatible nature and large molecular repertoire. Short peptides in particular allow for straightforward synthesis, self-assembly, and reproducibility. Applications are currently limited, however, due to potential toxicity of the chemical modifications that drive self-assembly and harsh gelation conditions. Peptides conjugated to nucleobases present one opportunity for a naturally derived species to minimize cytotoxicity. We have developed a hydrogel-formation environment for nucleopeptide gelation modulated entirely by biological buffers and salts. Self-assembly in this system is dependent on buffer and ion identity mediated by pKa and formulation in the former and by valency and ionicity in the latter. Solutions at physiological pH and osmolarity, and in turn compatible with cell culture, initiate hydrogel formation and analytical and computational methods are used to explore pH and salt effects at the molecular and structural level. The mechanism of nucleopeptide self-assembly enables tuning of mechanical properties through the addition of divalent cations and one order of magnitude increase in hydrogel storage modulus. The stability of these constructs therefore provides an opportunity for long-term cell culture, and we demonstrate survival and proliferation of fibroblasts on hydrogel surfaces. This novel, biological buffer-mediated gelation methodology expands opportunities for tissue engineering applications of short peptides and their derivatives.


Asunto(s)
Hidrogeles , Ingeniería de Tejidos , Técnicas de Cultivo de Célula , Péptidos , Reproducibilidad de los Resultados
7.
Nanoscale ; 13(6): 3644-3653, 2021 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-33538275

RESUMEN

Recent advances in immunotherapy have highlighted a need for therapeutics that initiate immunogenic cell death in tumors to stimulate the body's immune response to cancer. This study examines whether laser-generated bubbles surrounding nanoparticles ("nanobubbles") induce an immunogenic response for cancer treatment. A single nanosecond laser pulse at 1064 nm generates micron-sized bubbles surrounding gold nanorods in the cytoplasm of breast cancer cells. Cell death occurred in cells treated with nanorods and irradiated, but not in cells with irradiation treatment alone. Cells treated with nanorods and irradiation had increased damage-associated molecular patterns (DAMPs), including increased expression of chaperone proteins human high mobility group box 1 (HMGB1), adenosine triphosphate (ATP), and heat shock protein 70 (HSP70). This enhanced expression of DAMPs led to the activation of dendritic cells. Overall, this treatment approach is a rapid and highly specific method to eradicate tumor cells with simultaneous immunogenic cell death signaling, showing potential as a combination strategy for immunotherapy.


Asunto(s)
Neoplasias de la Mama , Proteína HMGB1 , Neoplasias de la Mama/terapia , Calreticulina/metabolismo , Humanos , Muerte Celular Inmunogénica , Rayos Láser
8.
Methods Mol Biol ; 2183: 489-498, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32959262

RESUMEN

Aluminum salts are used as an adjuvant in many human and veterinary vaccines. However, aluminum salt-adjuvanted vaccines are sensitive to temperature change and must be stored at 2-8 °C. Inadvertently exposing them to slow freezing temperatures can cause irreversible aggregation of aluminum salt microparticles and loss of potency and/or immunogenicity of the vaccines. There have been efforts to overcome this limitation by either adding stabilizing agents to the liquid vaccine or converting the vaccine from a liquid to a dry powder. Thin-film freeze-drying (TFFD) has proven to be an effective process to convert aluminum salt-adjuvanted vaccines from liquid to dry powder without causing particle aggregation or loss of immunogenicity upon reconstitution. This chapter provides a review of the TFFD process and examples for preparing stable aluminum salt-adjuvanted vaccine dry powders using TFFD.


Asunto(s)
Adyuvantes Inmunológicos , Aluminio , Criopreservación , Vacunas , Aluminio/química , Animales , Antígenos/inmunología , Criopreservación/métodos , Estabilidad de Medicamentos , Liofilización , Humanos , Ratones , Vacunas/inmunología
9.
Biomater Sci ; 8(11): 3130-3137, 2020 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-32352097

RESUMEN

Self-assembled nucleo-peptide hydrogels have a nanofibril structure composed of noncovalent molecular interactions between peptide groups as well as π-π stacking and Watson-Crick interactions via complementary nucleobases. These hydrogels have specific benefits for biomedical applications due to their DNA-like interactions in addition to the well-known advantages of peptide biomaterials: biocompatibility, extracellular matrix (ECM)-like structure, and bottom-up design. Inspired by the nucleobase stacking structure, we hypothesized that nucleo-peptides would be able to deliver the DNA-intercalating chemotherapeutic, doxorubicin (Dox) in a sustained manner when delivered locally to a solid tumor. Ade-FFF nucleo-peptide hydrogels were able to load a high concentration of Dox (1 mM) and demonstrated continuous release under in vitro degradation conditions. We adopted an in vivo tumor-bearing mouse model to evaluate the delivery of Dox by Ade-FFF hydrogels. We found that Dox-containing hydrogels reduced tumor growth and resulted in greater apoptosis-mediated cell death in the tumor as evidenced by caspase-3 expression. Pharmacokinetics and biodistribution studies also supported the observation that Dox delivery by an Ade-FFF hydrogel improves sustained delivery in the local tumor site. This study demonstrates the potential of self-assembled nucleo-peptides in biomedical applications by using their distinctive DNA-like structure.


Asunto(s)
Adenina/análogos & derivados , Adenina/administración & dosificación , Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Hidrogeles/administración & dosificación , Neoplasias/tratamiento farmacológico , Péptidos/administración & dosificación , Adenina/química , Adenina/farmacocinética , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Hidrogeles/química , Hidrogeles/farmacocinética , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Péptidos/química , Péptidos/farmacocinética
10.
Integr Biol (Camb) ; 12(6): 161-174, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32472133

RESUMEN

Soft tissue tumors, including breast cancer, become stiffer throughout disease progression. This increase in stiffness has been shown to correlate to malignant phenotype and epithelial-to-mesenchymal transition (EMT) in vitro. Unlike current models, utilizing static increases in matrix stiffness, our group has previously created a system that allows for dynamic stiffening of an alginate-matrigel composite hydrogel to mirror the native dynamic process. Here, we utilize this system to evaluate the role of matrix stiffness on EMT and metastasis both in vitro and in vivo. Epithelial cells were seen to lose normal morphology and become protrusive and migratory after stiffening. This shift corresponded to a loss of epithelial markers and gain of mesenchymal markers in both the cell clusters and migrated cells. Furthermore, stiffening in a murine model reduced tumor burden and increased migratory behavior prior to tumor formation. Inhibition of FAK and PI3K in vitro abrogated the morphologic and migratory transformation of epithelial cell clusters. This work demonstrates the key role extracellular matrix stiffening has in tumor progression through integrin signaling and, in particular, its ability to drive EMT-related changes and metastasis.


Asunto(s)
Movimiento Celular , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Metástasis de la Neoplasia , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Hidrogeles/química , Técnicas In Vitro , Integrinas/metabolismo , Neoplasias Mamarias Animales/metabolismo , Ratones , Microscopía Confocal , Invasividad Neoplásica , Neoplasias/metabolismo , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
11.
Acta Biomater ; 108: 250-260, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32251779

RESUMEN

Macrophages play a critical role in the initiation, maintenance, and resolution of inflammation because of their diverse and plastic phenotypic responses to extracellular stimuli. Inflammatory stimuli drive the recruitment and activation of inflammatory (M1) macrophages, capable of significant cytokine production that potentiates inflammation. Local environmental signals including apoptotic cell efferocytosis drive a phenotypic transition toward pro-reparative (M2) macrophages to facilitate the resolution of inflammation. However, prolonged or dysregulated inflammatory macrophage response contributes to many disease states and tissue damage. We have developed a nanoparticle to help resolve macrophage-mediated inflammation by mimicking the anti-inflammatory effect of apoptotic cell engulfment. The nanoparticle, comprised of a poly(lactide-co-glycolide) core, is coated in phosphatidylserine (PS)-supplemented cell plasma membrane to emulate key characteristics of the apoptotic cell surface. These apoptotic body-inspired PS/membrane-coated nanoparticles (PS-MNPs) reduce inflammatory cytokine expression to promote an anti-inflammatory, phenotypic shift in macrophages in vitro, without the use of small molecule inhibitors or other drugs. Specifically, PS-MNP treatment before lipopolysaccharide (LPS)-induced inflammatory challenge resulted in a 2.5-fold reduction in secreted tumor necrosis factor α (TNFα) at 24 h, with co-treatment of PS-MNPs and LPS demonstrating a 5-fold TNFα reduction compared to LPS alone. Reduced TNFα production, as well as gene expression of several pro-inflammatory cytokines, correlated with a reduction in NFκB activation from PS-MNP treatment. The development of a nanoparticle to reduce the production of multiple inflammatory cytokines and transition away from an inflammatory macrophage phenotype, through the use of a physiologic anti-inflammatory pathway, illustrates a new potential strategy in creating anti-inflammatory therapeutics. STATEMENT OF SIGNIFICANCE: Macrophages propagate inflammation as the major source of cytokine production in the body. In inflammatory diseases, pro-inflammatory macrophages persist in the site of inflammation and exacerbate tissue destruction. Current anti-inflammatory drugs have significant drawbacks, including variable response rates and off-target effects. Here, we have developed an apoptotic-body inspired nanoparticle to modulate inflammatory macrophage phenotype. This polymeric nanoparticle is coated with phosphatidylserine-supplemented cell plasma membrane to mimic the anti-inflammatory effect of apoptotic cell engulfment. Nanoparticle delivery reduces inflammatory cytokine production and promotes an anti-inflammatory phenotypic macrophage shift. The capacity of these nanoparticles to help resolve macrophage-mediated inflammation may be a useful tool to study macrophage-apoptotic cell interactions, the role of macrophages in inflammatory diseases, and in the design of anti-inflammatory therapeutics.


Asunto(s)
Macrófagos , Nanopartículas , Citocinas , Humanos , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología
12.
Nanoscale Adv ; 2(9): 3849-3857, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36132778

RESUMEN

Identifying pro-inflammatory macrophages (M1) is of immense importance to diagnose, monitor, and treat various pathologies. In addition, adoptive cell therapies, where harvested cells are isolated, modified to express an M1-like phenotype, then re-implanted to the patient, are also becoming more prevalent to treat diseases such as cancer. In a step toward identifying, labeling, and monitoring macrophage phenotype for adoptive cell therapies, we developed a reactive oxygen species (ROS)-sensitive, gold nanoparticle (AuNP) that fluorescently labels M1 macrophages. AuNPs are electrostatically coated with a proteolysis resistant, fluorescein isothiocyanate-conjugated, poly-d-lysine (PDL-FITC) that is susceptible to backbone cleavage by ROS. When PDL-FITC is bound to AuNPs, fluorescence is quenched via a combination of nanoparticle surface (NSET) and Forster resonance (FRET) energy transfer mechanisms. Upon ROS-induced cleavage of PDL-FITC, up to a 7-fold change in fluorescence is demonstrated. PDL-FITC AuNPs were loaded into RAW 264.7 macrophages (RAWs) and primary bone marrow- derived macrophages (BMDMs) prior to in vitro polarization. For both cell types, detectable differences in intracellular fluorescence were observed between M1 polarized and non-stimulated (M0) control groups after 24 h using both confocal imaging and flow cytometry. PDL-FITC AuNPs can potentially be useful in identifying M1 macrophages within diverse cell populations and provide longitudinal macrophage response data to external cues.

13.
ACS Nano ; 13(7): 7791-7799, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31250647

RESUMEN

Adult stem cell therapy has demonstrated improved outcomes for treating cardiovascular diseases in preclinical trials. The development of imaging tools may increase our understanding of the mechanisms of stem cell therapy, and a variety of imaging tools have been developed to image transplanted stem cells in vivo; however, they lack the ability to interrogate stem cell function longitudinally. Here, we report the use of a nanoparticle-based contrast agent that can track stem cell viability using photoacoustic imaging. The contrast agent consists of inert gold nanorods coated with IR775c, a reactive oxygen species (ROS) sensitive near-infrared dye. Upon cell death, stem cells produce ROS to degrade the cell. Using this feature of stem cells, the viability can be measured by comparing the IR775c signal to the ROS insensitive gold nanorod signal, which can also be used to track stem cell location. The nanoprobe was successfully loaded into mesenchymal stem cells (MSCs), and then, MSCs were transplanted into the lower limb of a mouse and imaged using combined ultrasound and photoacoustic imaging. MSC viability was assessed using the nanoprobe and displayed significant cell death within 24 h and an estimated 5% viability after 10 days. This nanoparticle system allows for longitudinal tracking of MSC viability in vivo with high spatial and temporal resolution which other imaging modalities currently cannot achieve.


Asunto(s)
Rastreo Celular , Colorantes Fluorescentes/química , Células Madre Mesenquimatosas/citología , Nanopartículas/química , Técnicas Fotoacústicas , Animales , Supervivencia Celular , Células Cultivadas , Femenino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie
14.
J Biomed Mater Res B Appl Biomater ; 107(2): 314-323, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29656592

RESUMEN

Three-dimensional bioprinting is an innovative technique in tissue engineering, to create layer-by-layer structures, required for mimicking body tissues. However, synthetic bioinks do not generally possess high printability and biocompatibility at the same time. So, there is an urgent need for naturally derived bioinks that can exhibit such optimized properties. We used furfuryl-gelatin as a novel, visible-light crosslinkable bioink for fabricating cell-laden structures with high viability. Hyaluronic acid was added as a viscosity enhancer and either Rose Bengal or Riboflavin was used as a visible-light crosslinker. Crosslinking was done by exposing the printed structure for 2.5 min to visible light and confirmed using Fourier transform infrared spectroscopy and rheometry. Scanning electron microscopy revealed a highly porous networked structure. Three different cell types were successfully bioprinted within these constructs. Mouse mesenchymal stem cells printed within monolayer and bilayer sheets showed viability, network formation and proliferation (∼5.33 times) within 72 h of culture. C2C12 and STO cells were used to print a double layered structure, which showed evidence of the viability of both cells and heterocellular clusters within the construct. This furfuryl-gelatin based bioink can be used for tissue engineering of complex tissues and help in understanding how cellular crosstalk happens in vivo during normal or diseased pathology. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 314-323, 2019.


Asunto(s)
Bioimpresión , Gelatina/química , Ácido Hialurónico/química , Tinta , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Tejidos , Animales , Línea Celular , Células Madre Mesenquimatosas/citología , Ratones , Porcinos
15.
Front Oncol ; 8: 337, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30234012

RESUMEN

The persistence of drug resistant cell populations following chemotherapeutic treatment is a significant challenge in the clinical management of cancer. Resistant subpopulations arise via both cell intrinsic and extrinsic mechanisms. Extrinsic factors in the microenvironment, including neighboring cells, glycosaminoglycans, and fibrous proteins impact therapy response. Elevated levels of extracellular fibrous proteins are associated with tumor progression and cause the surrounding tissue to stiffen through changes in structure and composition of the extracellular matrix (ECM). We sought to determine how this progressively stiffening microenvironment affects the sensitivity of breast cancer cells to chemotherapeutic treatment. MDA-MB-231 triple negative breast carcinoma cells cultured in a 3D alginate-based hydrogel system displayed a stiffness-dependent response to the chemotherapeutic doxorubicin. MCF7 breast carcinoma cells cultured in the same conditions did not exhibit this stiffness-dependent resistance to the drug. This differential therapeutic response was coordinated with nuclear translocation of YAP, a marker of mesenchymal differentiation. The stiffness-dependent response was lost when cells were transferred from 3D to monolayer cultures, suggesting that endpoint ECM conditions largely govern the response to doxorubicin. To further examine this response, we utilized a platform capable of dynamic ECM stiffness modulation to allow for a change in matrix stiffness over time. We found that MDA-MB-231 cells have a stiffness-dependent resistance to doxorubicin and that duration of exposure to ECM stiffness is sufficient to modulate this response. These results indicate the need for additional tools to integrate mechanical stiffness with therapeutic response and inform decisions for more effective use of chemotherapeutics in the clinic.

16.
J Vasc Surg ; 67(6): 1908-1920.e1, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29273298

RESUMEN

OBJECTIVE: Peripheral arterial disease can cause not only ischemia but also skeletal muscle damage. It has been known that macrophages (MPs) play an important role in coordinating muscle repair; however, phenotype transition of monocyte-MP in ischemic muscle has not been well defined. Hence, the purpose of this study was to examine the temporal recruitment of MPs and to explore their therapeutic effect on ischemic muscle regeneration. METHODS: Unilateral femoral artery excision was performed on C57BL/6 mice. Myeloid cells were isolated from the ischemic muscles, characterized using flow cytometry. Bone marrow-derived MPs were injected (2 × 106 cells) into the ischemic gastrocnemius muscle 24 hours after injury. Blood flow recovery was measured using laser speckle imaging. Functional outcome was evaluated by assessing the contractile force of ischemic muscles. Histologic analysis included quantification of myofiber size, collagen deposition, number of inflammatory and MyoD-expressing cells, and capillary density. RESULTS: Neutrophils and inflammatory monocytes-MPs were present at day 1 after injury. The mature MPs then remained elevated as the dominant population from day 5 to day 21 with the observation of regenerating fibers. Functional measurements revealed that the force production was significantly enhanced after treatment with proinflammatory M1 MPs (94.9% vs 77.9%; P < .05), and this was consistent with increased myofiber size, capillary- fiber ratio, and perfusion (78.6% vs 39.9%; P < .05). Moreover, the percentage of MyoD-expressing nuclei was significantly higher at day 4, indicating that M1 MPs may hasten muscle repair. Whereas early delivery of anti-inflammatory M2 MPs improved myofiber size, this was accompanied by persistent fibrosis suggesting ongoing tissue remodeling, and lower force production was observed. CONCLUSIONS: We demonstrated the dynamics of myeloid cells in skeletal muscle after ischemic insult, and the administration of exogenous M1 MPs in a temporally coordinated manner successfully improved angiogenesis and skeletal muscle regeneration. Our results suggested that cell therapy using MPs may be a promising adjunctive therapeutic approach for peripheral arterial disease.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Macrófagos/trasplante , Músculo Esquelético/patología , Animales , Velocidad del Flujo Sanguíneo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Isquemia/patología , Isquemia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Músculo Esquelético/fisiopatología
17.
Regen Med ; 12(2): 153-167, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28244825

RESUMEN

AIM: Progressive ischemia due to peripheral artery disease causes muscle damage and reduced strength of the lower extremities. Autologous cell therapy is an attractive treatment to restore perfusion and improve muscle function. Adipose-derived stem cells (ASCs) have therapeutic potential in tissue repair, including polarizing effects on macrophages (MPs). MATERIALS & METHODS: Co-culture systems of ASCs and MPs were analyzed for gene and protein expression modifications in ASC-conditioned MPs. Co-transplantation of MPs/ASCs in vivo led to improved skeletal muscle regeneration in a mouse model of peripheral artery disease. RESULTS: ASCs/MPs therapy restored muscle function, increased perfusion and reduced inflammatory infiltrate. CONCLUSION: Combined MPs/ASCs cell therapy is a promising approach to restore muscle function and stimulate local angiogenesis in the ischemic limb.


Asunto(s)
Tejido Adiposo/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Isquemia/terapia , Macrófagos/citología , Músculo Esquelético/citología , Regeneración/fisiología , Células Madre/citología , Animales , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Neovascularización Fisiológica , Enfermedad Arterial Periférica/terapia , Trasplante de Células Madre
18.
Cell Mol Bioeng ; 10(1): 114-123, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31719853

RESUMEN

Tumors are much stiffer than healthy tissue, and progressively stiffen as the cancer develops. Tumor stiffening is largely the result of extracellular matrix (ECM) remodeling, for example, deposition and crosslinking of collagen I. Well established in vitro models have demonstrated the influence of the microenvironment in regulating tissue homeostasis, with matrix stiffness being a particularly influential mediator. Non-malignant MCF10A mammary epithelial cells (MECs) lose their epithelial characteristics and become invasive when cultured in stiff microenvironments, leading to the hypothesis that tumor stiffening could contribute directly to disease progression. However, previous studies demonstrating MCF10A invasion have been performed in gels with constant mechanical properties, unlike the dynamically stiffening tumor microenvironment. Here, we employ a temporally stiffening hydrogel platform to demonstrate that matrix stiffening induces invasion from and proliferation in MCF10A mammary acini. After allowing MCF10A acini to form in soft hydrogels for 14 days, the gels were stiffened to the level of a malignant tumor, giving rise to a proliferative and invasive phenotype. Cells were observed to collectively migrate away from mammary acini while maintaining cell-cell contacts. Small molecule inhibition of PI3K and Rac1 pathways was sufficient to significantly reduce the number and size of invasive acini after stiffening. Our results demonstrate that temporal matrix stiffening can induce invasion from mammary acini and supports the notion that tumor stiffening could be implicated in disease progression and metastasis.

19.
Biomaterials ; 102: 9-19, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27318932

RESUMEN

The intent of the current study was to investigate the therapeutic contribution of MSCs to vascular regeneration and functional recovery of ischemic tissue. We used a rodent hind limb ischemia model and intramuscularly delivered MSCs within a PEGylated fibrin gel matrix. Within this model, we demonstrated that MSC therapy, when delivered in PEGylated fibrin, results in significantly higher mature blood vessel formation, which allows for greater functional recovery of skeletal muscle tissue as assessed using force production measurements. We observed initial signs of vascular repair at early time points when MSCs were delivered without PEGylated fibrin, but this did not persist or lead to recovery of the tissue in the long-term. Furthermore, animals which were treated with PEGylated fibrin alone exhibited a greater number of mature blood vessels, but they did not arterialize and did not show improvements in force production. These results demonstrate that revascularization of ischemic tissue may be a necessary but not sufficient step to complete functional repair of the injured tissue. This work has implications on stem cell therapies for ischemic diseases and also potentially on how such therapies are evaluated.


Asunto(s)
Fibrina/química , Geles/química , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Polietilenglicoles/química , Animales , Materiales Biocompatibles/química , Células Cultivadas , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Isquemia/patología , Neovascularización Fisiológica , Ratas , Ratas Endogámicas Lew , Andamios del Tejido/química
20.
PLoS One ; 10(12): e0145550, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26717325

RESUMEN

Skeletal muscle regeneration following acute injury is a multi-step process involving complex changes in tissue microenvironment. Macrophages (MPs) are one of the key cell types involved in orchestration and modulation of the repair process. Multiple studies highlight the essential role of MPs in the control of the myogenic program and inflammatory response during skeletal muscle regeneration. A variety of MP phenotypes have been identified and characterized in vitro as well as in vivo. As such, MPs hold great promise for cell-based therapies in the field of regenerative medicine. In this study we used bone-marrow derived in vitro LPS/IFN-y-induced M1 MPs to enhance functional muscle recovery after tourniquet-induced ischemia/reperfusion injury (TK-I/R). We detected a 15% improvement in specific tension and force normalized to mass after M1 (LPS/IFN-γ) MP transplantation 24 hours post-reperfusion. Interestingly, we found that M0 bone marrow-derived unpolarized MPs significantly impaired muscle function highlighting the complexity of temporally coordinated skeletal muscle regenerative program. Furthermore, we show that delivery of M1 (LPS/IFN-γ) MPs early in regeneration accelerates myofiber repair, decreases fibrotic tissue deposition and increases whole muscle IGF-I expression.


Asunto(s)
Macrófagos/trasplante , Músculo Esquelético/lesiones , Músculo Esquelético/fisiopatología , Animales , Peso Corporal/efectos de los fármacos , Células de la Médula Ósea/citología , Polaridad Celular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interferón gamma/farmacología , Antígenos Comunes de Leucocito/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología , Coloración y Etiquetado , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA