Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Pharmacol Transl Sci ; 6(2): 320-333, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36798475

RESUMEN

Biomaterials derived from brain extracellular matrix (ECM) have the potential to promote neural tissue regeneration by providing instructive cues that can direct cell survival, proliferation, and differentiation. This study focused on the development and characterization of microcarriers derived from decellularized brain tissue (DBT) as a platform for neural progenitor cell culture. First, a novel detergent-free decellularization protocol was established that effectively reduced the cellular content of porcine and rat brains, with a >97% decrease in the dsDNA content, while preserving collagens (COLs) and glycosaminoglycans (GAGs). Next, electrospraying methods were applied to generate ECM-derived microcarriers incorporating the porcine DBT that were stable without chemical cross-linking, along with control microcarriers fabricated from commercially sourced bovine tendon COL. The DBT microcarriers were structurally and biomechanically similar to the COL microcarriers, but compositionally distinct, containing a broader range of COL types and higher sulfated GAG content. Finally, we compared the growth, phenotype, and neurotrophic factor gene expression levels of rat brain-derived progenitor cells (BDPCs) cultured on the DBT or COL microcarriers within spinner flask bioreactors over 2 weeks. Both microcarrier types supported BDPC attachment and expansion, with immunofluorescence staining results suggesting that the culture conditions promoted BDPC differentiation toward the oligodendrocyte lineage, which may be favorable for cell therapies targeting remyelination. Overall, our findings support the further investigation of the ECM-derived microcarriers as a platform for neural cell derivation for applications in regenerative medicine.

2.
J Biomed Mater Res A ; 111(3): 415-434, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36210786

RESUMEN

Recognizing the cell-instructive capacity of the extracellular matrix (ECM), this study investigated the effects of expanding human adipose-derived stromal cells (hASCs) on ECM-derived microcarriers fabricated from decellularized adipose tissue (DAT) or decellularized cartilage tissue (DCT) within spinner flask bioreactors. Protocols were established for decellularizing porcine auricular cartilage and electrospraying methods were used to generate microcarriers comprised exclusively of DAT or DCT, which were compositionally distinct, but had matching Young's moduli. Both microcarrier types supported hASC attachment and growth over 14 days within a low-shear spinner culture system, with a significantly higher cell density observed on the DCT microcarriers at 7 and 14 days. Irrespective of the ECM source, dynamic culture on the microcarriers altered the expression of genes and proteins associated with cell adhesion and ECM remodeling. Label-free mass spectrometry analysis showed upregulation of proteins associated with cartilage development and ECM in the hASCs expanded on the DCT microcarriers. Based on Luminex analysis, the hASCs expanded on the DCT microcarriers secreted significantly higher levels of IL-8 and PDGFAA, supporting that the ECM source can modulate hASC paracrine factor secretion. Finally, the hASCs expanded on the microcarriers were extracted for analysis of adipogenic and chondrogenic differentiation relative to baseline controls. The microcarrier-cultured hASCs showed enhanced intracellular lipid accumulation at 7 days post-induction of adipogenic differentiation. In the chondrogenic studies, a low level of differentiation was observed in all groups. Future studies are warranted using alternative cell sources with greater chondrogenic potential to further assess the chondro-inductive properties of the DCT microcarriers.


Asunto(s)
Adipocitos , Tejido Adiposo , Animales , Humanos , Reactores Biológicos , Diferenciación Celular , Células Cultivadas , Células del Estroma , Porcinos
3.
Front Bioeng Biotechnol ; 10: 937239, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36237211

RESUMEN

Bioscaffolds derived from the extracellular matrix (ECM) have shown the capacity to promote regeneration by providing tissue-specific biological instructive cues that can enhance cell survival and direct lineage-specific differentiation. This study focused on the development and characterization of two-dimensional (2-D) and three-dimensional (3-D) cell culture platforms incorporating decellularized nucleus pulposus (DNP). First, a detergent-free protocol was developed for decellularizing bovine nucleus pulposus (NP) tissues that was effective at removing cellular content while preserving key ECM constituents including collagens, glycosaminoglycans, and the cell-adhesive glycoproteins laminin and fibronectin. Next, novel 2-D coatings were generated using the DNP or commercially-sourced bovine collagen type I (COL) as a non-tissue-specific control. In addition, cryo-milled DNP or COL particles were incorporated within methacrylated chondroitin sulphate (MCS) hydrogels as a 3-D cell culture platform for exploring the effects of ECM particle composition. Culture studies showed that the 2-D coatings derived from the DNP could support cell attachment and growth, but did not maintain or rescue the phenotype of primary bovine NP cells, which de-differentiated when serially passaged in monolayer culture. Similarly, while bovine NP cells remained highly viable following encapsulation and 14 days of culture within the hydrogel composites, the incorporation of DNP particles within the MCS hydrogels was insufficient to maintain or rescue changes in NP phenotype associated with extended in vitro culture based on gene expression patterns. Overall, DNP produced with our new decellularization protocol was successfully applied to generate both 2-D and 3-D bioscaffolds; however, further studies are required to assess if these platforms can be combined with additional components of the endogenous NP microenvironment to stimulate regeneration or lineage-specific cell differentiation.

4.
J Biomed Mater Res A ; 110(3): 559-575, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34581474

RESUMEN

Stirred bioreactor systems integrating microcarriers represent a promising approach for therapeutic cell manufacturing. While a variety of microcarriers are commercially available, current options do not integrate the tissue-specific composition of the extracellular matrix (ECM), which can play critical roles in directing cell function. The current study sought to generate microcarriers comprised exclusively of ECM from multiple tissue sources. More specifically, porcine decellularized dermis, porcine decellularized myocardium, and human decellularized adipose tissue were digested with α-amylase to obtain ECM suspensions that could be electrosprayed into liquid nitrogen to generate 3D microcarriers that were stable over a range of ECM concentrations without the need for chemical crosslinking or other additives. Characterization studies confirmed that all three microcarrier types had similar soft and compliant mechanical properties and were of a similar size range, but that their composition varied depending on the native tissue source. In vivo testing in immunocompetent mice revealed that the microcarriers integrated into the host tissues, supporting the infiltration of host cells including macrophages and endothelial cells at 2 weeks post-implantation. In vitro cell culture studies validated that the novel microcarriers supported the attachment of tissue-specific stromal cell populations under dynamic culture conditions within spinner flasks, with a significant increase in live cell numbers observed over 1 week on the dermal- and adipose-derived microcarriers. Overall, the findings demonstrate the versatility of the electrospraying methods and support the further development of the microcarriers as cell culture and delivery platforms.


Asunto(s)
Ingeniería de Tejidos , Andamios del Tejido , Tejido Adiposo , Animales , Células Endoteliales , Matriz Extracelular/química , Ratones , Porcinos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
5.
Wound Repair Regen ; 30(1): 45-63, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34708478

RESUMEN

In the skin-healing field, porcine models are regarded as a useful analogue for human skin due to their numerous anatomical and physiological similarities. Despite the widespread use of porcine models in skin healing studies, the initial origin, recruitment and transition of fibroblasts to matrix-secreting contractile myofibroblasts are not well defined for this model. In this review, we discuss the merit of the pig as an animal for studying myofibroblast origin, as well as the challenges associated with assessing their contributions to skin healing. Although a variety of wound types (incisional, partial thickness, full thickness, burns) have been investigated in pigs in attempts to mimic diverse injuries in humans, direct comparison of human healing profiles with regards to myofibroblasts shows evident differences. Following injury in porcine models, which often employ juvenile animals, myofibroblasts are described in the developing granulation tissue at 4 days, peaking at Days 7-14, and persisting at 60 days post-wounding, although variations are evident depending on the specific pig breed. In human wounds, the presence of myofibroblasts is variable and does not correlate with the age of the wound or clinical contraction. Our comparison of porcine myofibroblast-mediated healing processes with those in humans suggests that further validation of the pig model is essential. Moreover, we identify several limitations evident in experimental design that need to be better controlled, and standardisation of methodologies would be beneficial for the comparison and interpretation of results. In particular, we discuss anatomical location of the wounds, their size and depth, as well as the healing microenvironment (wet vs. moist vs. dry) in pigs and how this could influence myofibroblast recruitment. In summary, although a widespread model used in the skin healing field, further research is required to validate pigs as a useful analogue for human healing with regards to myofibroblasts.


Asunto(s)
Miofibroblastos , Cicatrización de Heridas , Animales , Modelos Animales de Enfermedad , Tejido de Granulación , Piel , Porcinos
6.
FASEB Bioadv ; 3(7): 541-557, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34258523

RESUMEN

Recent studies have highlighted the functional diversity of dermal fibroblast populations in health and disease, with part of this diversity linked to fibroblast lineage and embryonic origin. Fibroblasts derived from foxd1-expressing progenitors contribute to the myofibroblast populations present in lung and kidney fibrosis in mice but have not been investigated in the context of dermal wound repair. Using a Cre/Lox system to genetically track populations derived from foxd1-expressing progenitors, lineage-positive fibroblasts were identified as a subset of the dermal fibroblast population. During development, lineage-positive cells were most abundant within the dorsal embryonic tissues, contributing to the developing dermal fibroblast population, and remaining in this niche into adulthood. In adult mice, assessment of fibrosis-related gene expression in lineage-positive and lineage-negative populations isolated from wounded and unwounded dorsal skin was performed, identifying an enrichment of transcripts associated with matrix synthesis and remodeling in the lineage-positive populations. Using a novel excisional wound model, ventral skin healed with a greatly reduced frequency of foxd1 lineage-positive cells. This work supports that the embryonic origin of fibroblasts is an important predictor of fibroblast function, but also highlights that within disparate regions, fibroblasts of different lineages likely undergo convergent differentiation contributing to phenotypic similarities.

7.
Biomaterials ; 275: 120978, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34182328

RESUMEN

With the goal of establishing a new clinically-relevant bioscaffold format to enable the delivery of high densities of human adipose-derived stromal cells (ASCs) for applications in soft tissue regeneration, a novel "cell-assembly" method was developed to generate robust 3-D scaffolds comprised of fused networks of decellularized adipose tissue (DAT)-derived beads. In vitro studies confirmed that the assembly process was mediated by remodelling of the extracellular matrix by the seeded ASCs, which were well distributed throughout the scaffolds and remained highly viable after 8 days in culture. The ASC density, sulphated glycosaminoglycan content and scaffold stability were enhanced under culture conditions that included growth factor preconditioning. In vivo testing was performed to compare ASCs delivered within the cell-assembled DAT bead foams to an equivalent number of ASCs delivered on a previously-established pre-assembled DAT bead foam platform in a subcutaneous implant model in athymic nude mice. Scaffolds were fabricated with human ASCs engineered to stably co-express firefly luciferase and tdTomato to enable long-term cell tracking. Longitudinal bioluminescence imaging showed a significantly stronger signal associated with viable human ASCs at timepoints up to 7 days in the cell-assembled scaffolds, although both implant groups were found to retain similar densities of human ASCs at 28 days. Notably, the infiltration of CD31+ murine endothelial cells was enhanced in the cell-assembled implants at 28 days. Moreover, microcomputed tomography angiography revealed that there was a marked reduction in vascular permeability in the cell-assembled group, indicating that the developing vascular network was more stable in the new scaffold format. Overall, the novel cell-assembled DAT bead foams represent a promising platform to harness the pro-regenerative paracrine functionality of human ASCs and warrant further investigation as a clinically-translational approach for volume augmentation.


Asunto(s)
Células Madre Mesenquimatosas , Tejido Adiposo , Animales , Células Endoteliales , Ratones , Ratones Desnudos , Andamios del Tejido , Microtomografía por Rayos X
8.
Biomaterials ; 274: 120867, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33992837

RESUMEN

Cellular therapies to stimulate therapeutic angiogenesis in individuals with critical limb ischemia (CLI) remain under intense investigation. In this context, the efficacy of cell therapy is dependent on the survival, biodistribution, and pro-angiogenic paracrine signaling of the cells transplanted. Hematopoietic progenitor cells (HPC) purified from human umbilical cord blood using high aldehyde dehydrogenase-activity (ALDHhi cells) and expanded ex vivo, represent a heterogeneous mixture of progenitor cells previously shown to support limb revascularization in mouse models of CLI. The objectives of this study were to investigate the utility of bioscaffolds derived from human decellularized adipose tissue (DAT) to guide the differentiation of seeded HPC in vitro and harness the pro-angiogenic capacity of HPC at the site of ischemia after implantation in vivo. Probing whether the DAT scaffolds altered HPC differentiation, label-free quantitative mass spectrometry and flow cytometric phenotype analyses indicated that culturing the HPC on the DAT scaffolds supported their differentiation towards the pro-angiogenic monocyte/macrophage lineage at the expense of megakaryopoiesis. Moreover, implantation of HPC in DAT scaffolds within a unilateral hindlimb ischemia model in NOD/SCID mice increased cell retention at the site of ischemia relative to intramuscular injection, and accelerated the recovery of limb perfusion, improved functional limb use and augmented CD31+ capillary density when compared to DAT implantation alone or saline-injected controls. Collectively, these data indicate that cell-instructive DAT scaffolds can direct therapeutic HPC differentiation towards the monocyte/macrophage lineage and represent a promising delivery platform for improving the efficacy of cell therapies for CLI.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Andamios del Tejido , Tejido Adiposo , Animales , Diferenciación Celular , Miembro Posterior , Isquemia/terapia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Fisiológica , Regeneración , Distribución Tisular
9.
Front Bioeng Biotechnol ; 9: 642465, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816453

RESUMEN

Cell-based therapies involving the delivery of adipose-derived stromal cells (ASCs) on decellularized adipose tissue (DAT) scaffolds are a promising approach for soft tissue augmentation and reconstruction. Our lab has recently shown that culturing human ASCs on DAT scaffolds within a perfusion bioreactor prior to implantation can enhance their capacity to stimulate in vivo adipose tissue regeneration. Building from this previous work, the current study investigated the effects of bioreactor preconditioning on the ASC phenotype and secretory profile in vitro, as well as host cell recruitment following implantation in an athymic nude mouse model. Immunohistochemical analyses indicated that culturing within the bioreactor increased the percentage of ASCs co-expressing inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1), as well as tumor necrosis factor-alpha (TNF-α) and interleukin-10 (IL-10), within the peripheral regions of the DAT relative to statically cultured controls. In addition, bioreactor culture altered the expression levels of a range of immunomodulatory factors in the ASC-seeded DAT. In vivo testing revealed that culturing the ASCs on the DAT within the perfusion bioreactor prior to implantation enhanced the infiltration of host CD31+ endothelial cells and CD26+ cells into the DAT implants, but did not alter CD45+F4/80+CD68+ macrophage recruitment. However, a higher fraction of the CD45+ cell population expressed the pro-regenerative macrophage marker CD163 in the bioreactor group, which may have contributed to enhanced remodeling of the scaffolds into host-derived adipose tissue. Overall, the findings support that bioreactor preconditioning can augment the capacity of human ASCs to stimulate regeneration through paracrine-mediated mechanisms.

10.
Tissue Eng Part A ; 27(9-10): 618-630, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32873224

RESUMEN

Decellularized adipose tissue (DAT) scaffolds represent a promising cell-instructive platform for soft tissue engineering. While recent work has highlighted that mesenchymal stromal cells, including adipose-derived stromal cells (ASCs), can be combined with decellularized scaffolds to augment tissue regeneration, the mechanisms involved require further study. The objective of this work was to probe the roles of syngeneic donor ASCs and host-derived macrophages in tissue remodeling of DAT scaffolds within an immunocompetent mouse model. Dual transgenic reporter mouse strains were employed to track and characterize the donor ASCs and host macrophages within the DAT implants. More specifically, ASCs isolated from dsRed mice were seeded on DAT scaffolds, and the seeded and unseeded control scaffolds were implanted subcutaneously into MacGreen transgenic mice for up to 8 weeks. ASC seeding was shown to augment cell infiltration into the DAT implants at 8 weeks, and this was linked to significantly enhanced angiogenesis relative to the unseeded controls. Immunohistochemical staining demonstrated long-term retention of the syngeneic donor ASCs over the duration of the 8-week study, providing evidence that the DAT scaffolds are a cell-supportive delivery platform. Notably, newly formed adipocytes within the DAT implants were not dsRed+, indicating that the donor ASCs supported fat formation through indirect mechanisms. Immunohistochemical tracking of host macrophages through costaining for enhanced green fluorescent protein with the macrophage marker Iba1 revealed that ASC seeding significantly increased the number of infiltrating macrophages within the DAT implants at 3 weeks, while the fraction of macrophages relative to the total cellular infiltrate was similar between the groups at 1, 3, and 8 weeks. Consistent with the tissue remodeling response that was observed, western blotting demonstrated that there was significantly augmented expression of CD163 and CD206, markers of constructive M2-like macrophages, within the ASC-seeded DAT implants. Overall, our results demonstrate that exogenous ASCs enhance tissue regeneration within DAT scaffolds indirectly through multimodal mechanisms that include host cell recruitment and immunomodulation. These data provide further evidence to support the use of decellularized scaffolds as a delivery platform for ASCs in tissue engineering.


Asunto(s)
Adipocitos , Tejido Adiposo , Animales , Ratones , Células del Estroma , Ingeniería de Tejidos , Andamios del Tejido
11.
J Tissue Eng Regen Med ; 14(12): 1827-1840, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33009723

RESUMEN

Tissue-engineering approaches hold promise to address the need in plastic and reconstructive surgery for new therapies that promote stable adipose tissue regeneration. Previous studies have demonstrated the potential of combining decellularized adipose tissue (DAT) scaffolds with adipose-derived stromal cells (ASCs) for volume augmentation applications. With the goal of enhancing in vivo angiogenesis and adipogenesis, this study evaluated the effects of culturing human ASCs on DAT scaffolds within a perfusion bioreactor. Using this system, the impact of both dynamic culture and hypoxic preconditioning were explored in vitro and in vivo. Initial studies compared the effects of 14 days of culture within the perfusion bioreactor under hypoxia (2% O2 ) or normoxia (~20% O2 ) on human ASC expansion and expression of hypoxia inducible factor-1 alpha (HIF-1α) in vitro relative to static cultured controls. The findings indicated that culturing within the bioreactor under hypoxia significantly increased ASC proliferation on the DAT, with a higher cell density observed in the scaffold periphery. Subsequent characterization in a subcutaneous implant model in athymic nude mice revealed that in vivo angiogenesis and adipogenesis were markedly enhanced when the ASCs were cultured on the DAT within the perfusion bioreactor under hypoxia for 14 days prior to implantation relative to the other culture conditions, as well as freshly seeded and unseeded DAT control groups. Overall, dynamic culture within the perfusion bioreactor system under hypoxia represents a promising approach for preconditioning ASCs on DAT scaffolds to enhance their capacity to stimulate angiogenesis and host-derived adipose tissue regeneration.


Asunto(s)
Tejido Adiposo/citología , Reactores Biológicos , Perfusión , Regeneración/fisiología , Andamios del Tejido/química , Animales , Recuento de Células , Diferenciación Celular , Hipoxia de la Célula , Células Cultivadas , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Neovascularización Fisiológica , Implantación de Prótesis , Células del Estroma/citología , Tejido Subcutáneo/fisiología
12.
Biotechnol J ; 15(3): e1900118, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31657515

RESUMEN

While extracellular matrix (ECM)-derived coatings have the potential to direct the response of cell populations in culture, there is a need to investigate the effects of ECM sourcing and processing on substrate bioactivity. To develop improved cell culture models for studying adipogenesis, the current study examines the proliferation and adipogenic differentiation of human adipose-derived stem/stromal cells (ASCs) on a range of ECM-derived coatings. Human decellularized adipose tissue (DAT) and commercially available bovine tendon collagen (COL) are digested with α-amylase or pepsin to prepare the coatings. Physical characterization demonstrates that α-amylase digestion generates softer, thicker, and more stable coatings, with a fibrous tissue-like ultrastructure that is lost in the pepsin-digested thin films. ASCs cultured on the α-amylase-digested ECM have a more spindle-shaped morphology, and proliferation is significantly enhanced on the α-amylase-digested DAT coatings. Further, the α-amylase-digested DAT provides a more pro-adipogenic microenvironment, based on higher levels of adipogenic gene expression, glycerol-3-phosphate dehydrogenase (GPDH) enzyme activity, and perilipin staining. Overall, this study supports α-amylase digestion as a new approach for generating bioactive ECM-derived coatings, and demonstrates tissue-specific bioactivity using adipose-derived ECM to enhance ASC proliferation and adipogenic differentiation.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/enzimología , alfa-Amilasas/metabolismo , Adipogénesis/genética , Adipogénesis/fisiología , Tejido Adiposo/ultraestructura , Animales , Bovinos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Colágeno/química , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Microscopía de Fuerza Atómica , Microscopía Electroquímica de Rastreo , Tendones/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
13.
J Biomed Mater Res A ; 107(3): 571-585, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30390406

RESUMEN

Cell-based therapies involving the injection of adipose-derived stem/stromal cells (ASCs) within rationally designed biomaterials are a promising approach for stimulating angiogenesis. With this focus, the current work explored the effects of incorporating integrin-binding RGD or IKVAV peptides within in situ-gelling N-methacrylate glycol chitosan (MGC) hydrogels on the response of encapsulated human ASCs. Initial studies focused on hydrogel characterization to validate that the MGC, MGC-RGD, and MGC-IKVAV hydrogels had similar biomechanical properties. ASC viability following encapsulation and culture under 2% O2 was significantly impaired in the MGC-IKVAV group relative to the MGC and MGC-RGD groups. In contrast, sustained viability, along with enhanced cell spreading and metabolic activity were observed in the MGC-RGD group. Investigation of angiogenic transcription suggested that the incorporation of the peptide groups did not substantially alter the pro-angiogenic gene expression profile of the encapsulated ASCs after 7 days of culture under 2% O2. Consistent with the in vitro findings, preliminary in vivo characterization following subcutaneous implantation into NOD/SCID mice showed that ASC retention was enhanced in the MGC-RGD hydrogels relative to the MGC-IKVAV group at 14 days. Further, the encapsulated ASCs in the MGC and MGC-RGD groups promoted murine CD31+ endothelial cell recruitment to the peri-implant region. Overall, the results indicate that the MGC-RGD and MGC hydrogels are promising platforms for ASC delivery, and suggest that strategies that support long-term ASC viability can augment in vivo angiogenesis through paracrine mechanisms. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 571-585, 2019.


Asunto(s)
Tejido Adiposo/metabolismo , Células Inmovilizadas , Quitosano , Hidrogeles , Neovascularización Fisiológica , Oligopéptidos , Trasplante de Células Madre , Células Madre/metabolismo , Tejido Adiposo/citología , Animales , Supervivencia Celular , Células Inmovilizadas/citología , Células Inmovilizadas/metabolismo , Células Inmovilizadas/trasplante , Quitosano/química , Quitosano/farmacología , Xenoinjertos , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oligopéptidos/química , Oligopéptidos/farmacología , Células Madre/citología
14.
Acta Biomater ; 83: 199-210, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385224

RESUMEN

There is a substantial need for new strategies to stimulate cutaneous tissue repair in the treatment of chronic wounds. To address this challenge, our team is developing modular biomaterials termed "bead foams", comprised of porous beads synthesized exclusively of extracellular matrix (ECM) and assembled into a cohesive three-dimensional (3-D) network. In the current study, bead foams were fabricated from human decellularized adipose tissue (DAT) or commercially-sourced bovine tendon collagen (COL) to explore the effects of ECM composition on human wound edge dermal fibroblasts (weDF) sourced from chronic wound tissues. The DAT and COL bead foams were shown to be structurally similar, but compositionally distinct, containing different levels of glycosaminoglycan content and collagen types IV, V, and VI. In vitro testing under conditions simulating stresses within the chronic wound microenvironment indicated that weDF survival and angiogenic marker expression were significantly enhanced in the DAT bead foams as compared to the COL bead foams. These findings were corroborated through in vivo assessment in a subcutaneous athymic mouse model. Taken together, the results demonstrate that weDF survival and paracrine function can be modulated by the matrix source applied in the design of ECM-derived scaffolds and that the DAT bead foams hold promise as cell-instructive biological wound dressings. STATEMENT OF SIGNIFICANCE: Biological wound dressings derived from the extracellular matrix (ECM) can be designed to promote the establishment of a more permissive microenvironment for healing in the treatment of chronic wounds. In the current work, we developed modular biomaterials comprised of fused networks of porous ECM-derived beads fabricated from human decellularized adipose tissue (DAT) or commercially-available bovine collagen. The bioscaffolds were designed to be structurally similar to provide a platform for investigating the effects of ECM composition on human dermal fibroblasts isolated from chronic wounds. Testing in in vitro and in vivo models demonstrated that cell survival and pro-angiogenic function were enhanced in the adipose-derived bioscaffolds, which contained higher levels of glycosaminoglycans and collagen types IV, V, and VI. Our findings support that the complex matrix composition within DAT can induce a more pro-regenerative cellular response for applications in wound healing.


Asunto(s)
Dermis/metabolismo , Matriz Extracelular/química , Fibroblastos/metabolismo , Neovascularización Fisiológica , Andamios del Tejido/química , Heridas y Lesiones/metabolismo , Supervivencia Celular , Microambiente Celular , Enfermedad Crónica , Colágeno/química , Dermis/patología , Femenino , Fibroblastos/patología , Humanos , Heridas y Lesiones/patología
15.
ACS Biomater Sci Eng ; 5(12): 6655-6666, 2019 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33423484

RESUMEN

Melt electrowriting (MEW) is an additive manufacturing technology that produces readily handleable fibrous scaffolds with controlled geometry to support cell infiltration. Although MEW scaffolds have excellent potential for cell delivery in regenerative medicine applications, studies to date have primarily focused on polymers such as poly(ε-caprolactone) (PCL) that lack bioactive cues to affect cell function. To address this aspect, MEW scaffolds with extracellular matrix (ECM) coatings were developed as a proadipogenic platform for human mesenchymal stromal cells (hMSCs). More specifically, highly flexible PCL scaffolds fabricated through MEW were coated with a complex ECM suspension prepared from human decellularized adipose tissue (DAT), purified fibronectin, or laminin to determine the effects of two key bioactive proteins present within adipose-derived ECM. In vitro studies exploring the response of human bone marrow-derived mesenchymal stromal cells cultured under adipogenic differentiation conditions indicated a high level of differentiation on all substrates studied, including unmodified PCL scaffolds and two-dimensional controls. To more fully assess the intrinsic proadipogenic capacity of the composite biomaterials, a modified culture regime was established that involved a short-term adipogenic induction in differentiation medium, followed by continued culture in maintenance medium supplemented with insulin for up to 3 weeks. Under these conditions, adipogenic differentiation was enhanced on all fiber scaffolds as compared to the tissue culture controls. Notably, the highest adipogenic response was consistently observed on the PCL + DAT scaffolds, based on the analysis of multiple markers including adipogenic gene [lipoprotein lipase, fatty acid binding protein 4 (FABP4), adiponectin, perilipin 1] and protein (FABP4, leptin) expression and intracellular triglyceride accumulation. Taken together, the PCL scaffolds incorporating DAT provide an adipoinductive microenvironment for the hMSCs, with particular applicability of this cell-instructive delivery platform for applications in plastic and reconstructive surgery.

16.
Artículo en Inglés | MEDLINE | ID: mdl-31921807

RESUMEN

While it has been postulated that tissue-specific bioscaffolds derived from the extracellular matrix (ECM) can direct stem cell differentiation, systematic comparisons of multiple ECM sources are needed to more fully assess the benefits of incorporating tissue-specific ECM in stem cell culture and delivery platforms. To probe the effects of ECM sourced from decellularized adipose tissue (DAT) or decellularized trabecular bone (DTB) on the adipogenic and osteogenic differentiation of human adipose-derived stem/stromal cells (ASCs), a novel detergent-free decellularization protocol was developed for bovine trabecular bone that complemented our established detergent-free decellularization protocol for human adipose tissue and did not require specialized equipment or prolonged incubation times. Immunohistochemical and biochemical characterization revealed enhanced sulphated glycosaminoglycan content in the DTB, while the DAT contained higher levels of collagen IV, collagen VI and laminin. To generate platforms with similar structural and biomechanical properties to enable assessment of the compositional effects of the ECM on ASC differentiation, micronized DAT and DTB were encapsulated with human ASCs within methacrylated chondroitin sulfate (MCS) hydrogels through UV-initiated crosslinking. High ASC viability (>90%) was observed over 14 days in culture. Adipogenic differentiation was enhanced in the MCS+DAT composites relative to the MCS+DTB composites and MCS controls after 14 days of culture in adipogenic medium. Osteogenic differentiation studies revealed a peak in alkaline phosphatase (ALP) enzyme activity at 7 days in the MCS+DTB group cultured in osteogenic medium, suggesting that the DTB had bioactive effects on osteogenic protein expression. Overall, the current study suggests that tissue-specific ECM sourced from DAT or DTB can act synergistically with soluble differentiation factors to enhance the lineage-specific differentiation of human ASCs within 3-D hydrogel systems.

17.
Sci Rep ; 8(1): 16166, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385873

RESUMEN

Pannexin 1 (Panx1) is a channel-forming glycoprotein important in paracrine signaling and cellular development. In this study, we discovered that mice globally lacking Panx1 (KO) have significantly greater total fat mass and reduced lean mass compared to wild type (WT) mice under a normal diet. Despite having higher fat content, Panx1 KO mice on a high fat diet exhibited no differences in weight gain and blood markers of obesity as compared to WT controls, except for an increase in glucose and insulin levels. However, metabolic cage data revealed that these Panx1 KO mice display significantly increased activity levels, higher ambulatory activity, and reduced sleep duration relative to their WT littermates on a high-fat diet. To uncover the cellular mechanism responsible for the increased fat content in the KO, we isolated primary cultures of adipose-derived stromal cells (ASCs) from WT and KO fat pads. In WT ASCs we observed that Panx1 protein levels increase upon induction into an adipogenic lineage. ASCs isolated from Panx1 KO mice proliferate less but demonstrate enhanced adipogenic differentiation with increased intracellular lipid accumulation, glycerol-3-phosphate dehydrogenase (GPDH) enzyme activity, and adipokine secretion, as compared to WT ASCs. This was consistent with the increased adipocyte size and decreased adipocyte numbers observed in subcutaneous fat of the Panx1 KO mice compared to WT. We concluded that Panx1 plays a key role in adipose stromal cells during the early stages of adipogenic proliferation and differentiation, regulating fat accumulation in vivo.


Asunto(s)
Adipogénesis/genética , Conexinas/genética , Metabolismo de los Lípidos/genética , Proteínas del Tejido Nervioso/genética , Obesidad/genética , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Obesidad/patología , Células del Estroma/citología , Células del Estroma/metabolismo , Grasa Subcutánea/crecimiento & desarrollo , Grasa Subcutánea/metabolismo , Grasa Subcutánea/patología
18.
Tissue Eng Part A ; 24(23-24): 1784-1797, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29882474

RESUMEN

Injectable hydrogels have the potential to enhance stem cell-based therapies by improving cell localization, retention, and survival after transplantation. The inflammatory response to both the hydrogel and the encapsulated cells is a critical aspect of this strategy, with macrophages being highly involved in the process of hydrogel remodeling, angiogenesis, and tissue regeneration. As a step toward the development of a cell-based strategy for therapeutic angiogenesis, this work compared the intramuscular injection of allogeneic rat adipose-derived stem/stromal cells (rASCs) in an in situ gelling hydrogel with the injection of the hydrogel alone and rASCs in saline in an immunocompetent rat model by immunohistochemical analysis over 4 weeks. rASCs delivered in the hydrogel were retained intramuscularly at significantly higher densities as compared with cells delivered in saline. The encapsulated rASCs modulated the inflammatory response, promoting CD68+ macrophage recruitment, with the majority of infiltrating cells expressing the M1 marker CCR7, as well as a higher fraction of CD163+ M2c macrophages surrounding the hydrogel. Furthermore, rASCs reduced the initial expression of inducible nitric oxide synthase and promoted arginase-1 expression in the infiltrating macrophages over time, consistent with a shift toward a more proregenerative phenotype. Coincident with the enhanced macrophage infiltration, significantly more CD31+ lumens were observed surrounding and within the hydrogels with rASCs at 2 and 4 weeks as compared with the hydrogels alone. Overall, these results are a promising indication that encapsulated rASCs can have immunomodulatory effects and may enhance angiogenic processes after intramuscular injection, promoting a regenerative macrophage response and blood vessel formation.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular , Hidrogeles , Macrófagos/metabolismo , Trasplante de Células Madre , Células Madre/metabolismo , Adipocitos/citología , Animales , Células Inmovilizadas/citología , Células Inmovilizadas/metabolismo , Células Inmovilizadas/trasplante , Femenino , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Ratas , Ratas Wistar , Células Madre/citología
19.
Methods Mol Biol ; 1773: 53-71, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29687381

RESUMEN

Surgically discarded adipose tissue is not only an abundant source of multipotent adipose-derived stem/stromal cells (ASCs) but can also be decellularized to obtain a biomimetic microenvironment for tissue engineering applications. The decellularization methods involve processing excised fat through a series of chemical, mechanical, and enzymatic treatment stages designed to extract cells, cellular components, and lipid from the tissues. This process yields a complex 3D bioscaffold enriched in collagens that mimics the biochemical and biomechanical properties of the native extracellular matrix (ECM). For ASC culture and delivery, decellularized adipose tissue (DAT) provides a cell-supportive platform that is conducive to adipogenesis. While DAT can be applied in its intact form as an off-the-shelf adipogenic matrix, it can also be used as an ECM source for the fabrication of an array of other scaffold formats including adipose ECM-derived microcarriers and porous foams. In this chapter, we describe the methods developed in our lab to decellularize human adipose tissue and to further process it into a variety of scaffolding materials for a range of applications in soft tissue regeneration, wound healing, and cell culture.


Asunto(s)
Tejido Adiposo/citología , Regeneración Tisular Dirigida , Células Madre Mesenquimatosas/citología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Liofilización , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Nitrógeno/farmacología , Porosidad , Ingeniería de Tejidos , Andamios del Tejido , Cicatrización de Heridas
20.
Biomaterials ; 159: 146-160, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29324306

RESUMEN

A promising strategy for treating peripheral ischemia involves the delivery of stem cells to promote angiogenesis through paracrine signaling. Treatment success depends on cell localization, retention, and survival within the mechanically dynamic intramuscular (IM) environment. Herein we describe an injectable, in situ-gelling hydrogel for the IM delivery of adipose-derived stem/stromal cells (ASCs), specifically designed to withstand the dynamic loading conditions of the lower limb and facilitate cytokine release from encapsulated cells. Copolymers of poly(trimethylene carbonate)-b-poly(ethylene glycol)-b-poly(trimethylene carbonate) diacrylate were used to modulate the properties of methacrylated glycol chitosan hydrogels crosslinked by thermally-initiated polymerization using ammonium persulfate and N,N,N',N'-tetramethylethylenediamine. The scaffolds had an ultimate compressive strain over 75% and maintained mechanical properties during compressive fatigue testing at physiological levels. Rapid crosslinking (<3 min) was achieved at low initiator concentration (5 mM). Following injection and crosslinking within the scaffolds, human ASCs demonstrated high viability (>90%) over two weeks in culture under both normoxia and hypoxia. Release of angiogenic and chemotactic cytokines was enhanced from encapsulated cells under sustained hypoxia, in comparison to normoxic and tissue culture polystyrene controls. When delivered by IM injection in a mouse model of hindlimb ischemia, human ASCs were well retained in the scaffold over 28 days and significantly increased the IM vascular density compared to untreated controls.


Asunto(s)
Citocinas/metabolismo , Células Madre/metabolismo , Andamios del Tejido/química , Tejido Adiposo/citología , Animales , Células Cultivadas , Femenino , Humanos , Hidrogeles/química , Inmunohistoquímica , Ratones , Enfermedad Arterial Periférica/metabolismo , Ingeniería de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...