Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Cancer Res ; 84(9): 1517-1533, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38587552

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by an immunosuppressive tumor microenvironment enriched with cancer-associated fibroblasts (CAF). This study used a convergence approach to identify tumor cell and CAF interactions through the integration of single-cell data from human tumors with human organoid coculture experiments. Analysis of a comprehensive atlas of PDAC single-cell RNA sequencing data indicated that CAF density is associated with increased inflammation and epithelial-mesenchymal transition (EMT) in epithelial cells. Transfer learning using transcriptional data from patient-derived organoid and CAF cocultures provided in silico validation of CAF induction of inflammatory and EMT epithelial cell states. Further experimental validation in cocultures demonstrated integrin beta 1 (ITGB1) and vascular endothelial factor A (VEGFA) interactions with neuropilin-1 mediating CAF-epithelial cell cross-talk. Together, this study introduces transfer learning from human single-cell data to organoid coculture analyses for experimental validation of discoveries of cell-cell cross-talk and identifies fibroblast-mediated regulation of EMT and inflammation. SIGNIFICANCE: Adaptation of transfer learning to relate human single-cell RNA sequencing data to organoid-CAF cocultures facilitates discovery of human pancreatic cancer intercellular interactions and uncovers cross-talk between CAFs and tumor cells through VEGFA and ITGB1.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Técnicas de Cocultura , Transição Epitelial-Mesenquimal , Inflamação , Integrina beta1 , Neoplasias Pancreáticas , Análise de Célula Única , Microambiente Tumoral , Humanos , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Inflamação/patologia , Inflamação/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Organoides/patologia , Organoides/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Neuropilina-1/metabolismo , Neuropilina-1/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Comunicação Celular
2.
Artigo em Inglês | MEDLINE | ID: mdl-38654098

RESUMO

Cellular senescence is a state of terminal growth arrest associated with the upregulation of different cell cycle inhibitors, mainly p16 and p21, structural and metabolic alterations, chronic DNA damage responses, and a hypersecretory state known as the senescence-associated secretory phenotype (SASP). The SASP is the major mediator of the paracrine effects of senescent cells in their tissue microenvironment and of various local and systemic biological functions. In this Review, we discuss the composition, dynamics and heterogeneity of the SASP as well as the mechanisms underlying its induction and regulation. We describe the various biological properties of the SASP, its beneficial and detrimental effects in different physiological and pathological settings, and its impact on overall health span. Finally, we discuss the use of the SASP as a biomarker and of SASP inhibitors as senomorphic interventions to treat cancer and other age-related conditions.

3.
J Neurosurg Spine ; 39(1): 113-121, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37021767

RESUMO

OBJECTIVE: Infuse bone graft is a widely used osteoinductive adjuvant; however, the simple collagen sponge scaffold used in the implant has minimal inherent osteoinductive properties and poorly controls the delivery of the adsorbed recombinant human bone morphogenetic protein-2 (rhBMP-2). In this study, the authors sought to create a novel bone graft substitute material that overcomes the limitations of Infuse and compare the ability of this material with that of Infuse to facilitate union following spine surgery in a clinically translatable rat model of spinal fusion. METHODS: The authors created a polydopamine (PDA)-infused, porous, homogeneously dispersed solid mixture of extracellular matrix and calcium phosphates (BioMim-PDA) and then compared the efficacy of this material directly with Infuse in the setting of different concentrations of rhBMP-2 using a rat model of spinal fusion. Sixty male Sprague Dawley rats were randomly assigned to each of six equal groups: 1) collagen + 0.2 µg rhBMP-2/side, 2) BioMim-PDA + 0.2 µg rhBMP-2/side, 3) collagen + 2.0 µg rhBMP-2/side, 4) BioMim-PDA + 2.0 µg rhBMP-2/side, 5) collagen + 20 µg rhBMP-2/side, and 6) BioMim-PDA + 20 µg rhBMP-2/side. All animals underwent posterolateral intertransverse process fusion at L4-5 using the assigned bone graft. Animals were euthanized 8 weeks postoperatively, and their lumbar spines were analyzed via microcomputed tomography (µCT) and histology. Spinal fusion was defined as continuous bridging bone bilaterally across the fusion site evaluated via µCT. RESULTS: The fusion rate was 100% in all groups except group 1 (70%) and group 4 (90%). Use of BioMim-PDA with 0.2 µg rhBMP-2 led to significantly greater results for bone volume (BV), percentage BV, and trabecular number, as well as significantly smaller trabecular separation, compared with the use of the collagen sponge with 2.0 µg rhBMP-2. The same results were observed when the use of BioMim-PDA with 2.0 µg rhBMP-2 was compared with the use of the collagen sponge with 20 µg rhBMP-2. CONCLUSIONS: Implantation of rhBMP-2-adsorbed BioMim-PDA scaffolds resulted in BV and bone quality superior to that afforded by treatment with rhBMP-2 concentrations 10-fold higher implanted on a conventional collagen sponge. Using BioMim-PDA (vs a collagen sponge) for rhBMP-2 delivery could significantly lower the amount of rhBMP-2 required for successful bone grafting clinically, improving device safety and decreasing costs.


Assuntos
Fusão Vertebral , Masculino , Ratos , Humanos , Animais , Fusão Vertebral/métodos , Transplante Ósseo/métodos , Microtomografia por Raio-X , Biomimética , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/uso terapêutico , Proteína Morfogenética Óssea 2/farmacologia , Colágeno/farmacologia , Proteínas Recombinantes/farmacologia , Vértebras Lombares/cirurgia
4.
Geroscience ; 45(4): 2559-2587, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37079217

RESUMO

Cellular senescence is a state of permanent growth arrest that plays an important role in wound healing, tissue fibrosis, and tumor suppression. Despite senescent cells' (SnCs) pathological role and therapeutic interest, their phenotype in vivo remains poorly defined. Here, we developed an in vivo-derived senescence signature (SenSig) using a foreign body response-driven fibrosis model in a p16-CreERT2;Ai14 reporter mouse. We identified pericytes and "cartilage-like" fibroblasts as senescent and defined cell type-specific senescence-associated secretory phenotypes (SASPs). Transfer learning and senescence scoring identified these two SnC populations along with endothelial and epithelial SnCs in new and publicly available murine and human data single-cell RNA sequencing (scRNAseq) datasets from diverse pathologies. Signaling analysis uncovered crosstalk between SnCs and myeloid cells via an IL34-CSF1R-TGFßR signaling axis, contributing to tissue balance of vascularization and matrix production. Overall, our study provides a senescence signature and a computational approach that may be broadly applied to identify SnC transcriptional profiles and SASP factors in wound healing, aging, and other pathologies.


Assuntos
Envelhecimento , Senescência Celular , Humanos , Camundongos , Animais , Senescência Celular/genética , Envelhecimento/genética , Fenótipo , Fibroblastos , Aprendizado de Máquina
5.
NPJ Regen Med ; 7(1): 6, 2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35031598

RESUMO

Soft tissue reconstruction remains an intractable clinical challenge as current surgical options and synthetic implants may produce inadequate outcomes. Soft tissue deficits may be surgically reconstructed using autologous adipose tissue, but these procedures can lead to donor site morbidity, require multiple procedures, and have highly variable outcomes. To address this clinical need, we developed an "off-the-shelf" adipose extracellular matrix (ECM) biomaterial from allograft human tissue (Acellular Adipose Tissue, AAT). We applied physical and chemical processing methods to remove lipids and create an injectable matrix that mimicked the properties of lipoaspirate. Biological activity was assessed using cell migration and adipogenesis assays. Characterization of regenerative immune properties in a murine muscle injury model revealed that allograft and xenograft AAT induced pro-regenerative CD4+ T cells and macrophages with xenograft AAT additionally attracting eosinophils secreting interleukin 4 (Il4). In immunocompromised mice, AAT injections retained similar volumes as human fat grafts but lacked cysts and calcifications seen in the fat grafts. The combination of AAT with human adipose-derived stem cells (ASCs) resulted in lower implant volumes. However, tissue remodeling and adipogenesis increased significantly in combination with ASCs. Larger injected volumes of porcine-derived AAT demonstrated biocompatibility and greater retention when applied allogeneicly in Yorkshire cross pigs. AAT was implanted in healthy volunteers in abdominal tissue that was later removed by elective procedures. AAT implants were well tolerated in all human subjects. Implants removed between 1 and 18 weeks demonstrated increasing cellular infiltration and immune populations, suggesting continued tissue remodeling and the potential for long-term tissue replacement.

6.
Biomaterials ; 241: 119880, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32097748

RESUMO

As the outermost layer of the eye, the cornea is vulnerable to physical and chemical trauma, which can result in loss of transparency and lead to corneal blindness. Given the global corneal donor shortage, there is an unmet need for biocompatible corneal substitutes that have high transparency, mechanical integrity and regenerative potentials. Herein we engineered a dual-layered collagen vitrigel containing biomimetic synthetic Bowman's membrane (sBM) and stromal layer (sSL). The sBM supported rapid epithelial cell migration, maturation and multilayer formation, and the sSL containing tissue-derived extracellular matrix (ECM) microparticles presented a biomimetic lamellar ultrastructure mimicking the native corneal stroma. The incorporation of tissue-derived microparticles in sSL layer significantly enhanced the mechanical properties and suturability of the implant without compromising the transparency after vitrification. In vivo performance of the vitrigel in a rabbit anterior lamellar keratoplasty model showed full re-epithelialization within 14 days and integration of the vitrigel with the host tissue stroma by day 30. The migrated epithelial cells formed functional multilayer with limbal stem cell marker p63 K14 expressed in the lower layer, epithelial marker K3 and K12 expressed through the layers and tight junction protein ZO-1 expressed by the multilayers. Corneal fibroblasts migrated into the implants to facilitate host/implant integration and corneal stromal regeneration. In summary, these results suggest that the multi-functional layers of this novel collagen vitrigel exhibited significantly improved biological performance as corneal substitute by harnessing a fast re-epithelialization and stromal regeneration potential.


Assuntos
Lâmina Limitante Anterior , Transplante de Córnea , Animais , Biomimética , Colágeno , Córnea , Substância Própria , Coelhos
7.
Cancer Res ; 80(3): 377-378, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32015156

RESUMO

Synthetic biomaterials implanted in the body induce a foreign body reaction characterized by chronic inflammation and fibrosis. In this issue of Cancer Research, Oakes and colleagues used biomaterial implants and their associated immunologic activity to develop a "metastasis sensor" for detection of tumor burden at distal sites. A scoring system was developed from computational analysis of gene expression patterns from implant biopsies that could predict the presence of tumor. This unexpected use of biomaterials for early detection of cancer provides a more accurate systemic sampling compared with blood or liquid biopsies and alleviates the need for inefficient imaging and biopsy sampling from potential metastatic target tissues.See related article by Oakes et al., p. 602.


Assuntos
Materiais Biocompatíveis , Células Mieloides , Carvão Mineral , Progressão da Doença , Humanos , Resultado do Tratamento
8.
Cartilage ; 11(4): 447-457, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-30280586

RESUMO

OBJECTIVE: To determine performance and repair kinetics of the ChonDux hydrogel scaffold for treating focal articular cartilage defects in the knee over 24 months. DESIGN: This assessor-blinded trial evaluates ChonDux hydrogel scaffold implantation in combination with microfracture in 18 patients across 6 sites. Male and female patients 18 to 65 years of age with full-thickness femoral condyle defects 2 to 4 cm2 in area were enrolled. Eligible patients received ChonDux treatment followed by rehabilitation. Defect volume fill was evaluated after 3, 6 (primary outcome), 12, 18, and 24 months by assessor blinded magnetic resonance imaging (MRI) analysis. Secondary outcomes were T2-weighted MRI relaxation time and patient surveys via visual analogue scale (VAS) pain and International Knee Documentation Committee (IKDC) knee function scoring. RESULTS: ChonDux maintained durable tissue restoration over 24 months with final defect percent fill of 94.2% ± 16.3% and no significant loss of fill volume at any time points. Tissues treated with ChonDux maintained T2 relaxation times similar to uninjured cartilage between 12 and 24 months. VAS pain scoring decreased between 1 and 6 weeks, and IKDC knee function scores improved by approximately 30.1 with ChonDux over 24 months. CONCLUSION: ChonDux treatment is a safe adjunct to microfracture therapy and promotes stable restoration of full thickness articular cartilage defects for at least 24 months.


Assuntos
Doenças das Cartilagens/tratamento farmacológico , Cartilagem Articular/lesões , Fraturas de Estresse/reabilitação , Hidrogéis/administração & dosagem , Traumatismos do Joelho/reabilitação , Adolescente , Adulto , Idoso , Doenças das Cartilagens/etiologia , Doenças das Cartilagens/reabilitação , Cartilagem Articular/diagnóstico por imagem , Cartilagem Articular/fisiopatologia , Terapia Combinada , Avaliação da Deficiência , Feminino , Fêmur , Seguimentos , Fraturas de Estresse/complicações , Fraturas de Estresse/fisiopatologia , Humanos , Cinética , Traumatismos do Joelho/complicações , Traumatismos do Joelho/fisiopatologia , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Medição da Dor , Recuperação de Função Fisiológica , Método Simples-Cego , Resultado do Tratamento , Adulto Jovem
9.
Sci Rep ; 9(1): 16287, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31705048

RESUMO

Three-dimensional cell spheroid models can be used to predict the effect of drugs and therapeutics and to model tissue development and regeneration. The utility of these models is enhanced by high throughput 3D spheroid culture technologies allowing researchers to efficiently culture numerous spheroids under varied experimental conditions. Detailed analysis of high throughput spheroid culture is much less efficient and generally limited to narrow outputs, such as metabolic viability. We describe a microarray approach that makes traditional histological embedding/sectioning/staining feasible for large 3D cell spheroid sample sets. Detailed methodology to apply this technology is provided. Analysis of the technique validates the potential for efficient histological analysis of up to 96 spheroids in parallel. By integrating high throughput 3D spheroid culture technologies with advanced immunohistochemical techniques, this approach will allow researchers to efficiently probe expression of multiple biomarkers with spatial localization within 3D structures. Quantitative comparison of staining will have improved inter- and intra-experimental reproducibility as multiple samples are collectively processed, stained, and imaged on a single slide.


Assuntos
Técnicas de Cultura de Células , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Esferoides Celulares , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Camundongos
10.
Sci Immunol ; 4(40)2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604843

RESUMO

Biomaterials induce an immune response and mobilization of macrophages, yet identification and phenotypic characterization of functional macrophage subsets in vivo remain limited. We performed single-cell RNA sequencing analysis on macrophages sorted from either a biologic matrix [urinary bladder matrix (UBM)] or synthetic biomaterial [polycaprolactone (PCL)]. Implantation of UBM promotes tissue repair through generation of a tissue environment characterized by a T helper 2 (TH2)/interleukin (IL)-4 immune profile, whereas PCL induces a standard foreign body response characterized by TH17/IL-17 and fibrosis. Unbiased clustering and pseudotime analysis revealed distinct macrophage subsets responsible for antigen presentation, chemoattraction, and phagocytosis, as well as a small population with expression profiles of both dendritic cells and skeletal muscle after UBM implantation. In the PCL tissue environment, we identified a CD9hi+IL-36γ+ macrophage subset that expressed TH17-associated molecules. These macrophages were virtually absent in mice lacking the IL-17 receptor, suggesting that they might be involved in IL-17-dependent immune and autoimmune responses. Identification and comparison of the unique phenotypical and functional macrophage subsets in mouse and human tissue samples suggest broad relevance of the new classification. These distinct macrophage subsets demonstrate previously unrecognized myeloid phenotypes involved in different tissue responses and provide targets for potential therapeutic modulation in tissue repair and pathology.


Assuntos
Fibrose/imunologia , Interleucina-17/imunologia , Interleucina-1/imunologia , Macrófagos/imunologia , Animais , Modelos Animais de Doenças , Feminino , Interleucina-17/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
11.
Biomater Sci ; 7(11): 4472-4481, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31424059

RESUMO

The immune system has evolved as a powerful tool for our body to combat infections, and is being engineered for new treatments in cancer and autoimmune disease. More recently, the complex role of the immune system is being recognized in tissue repair, regenerative medicine and biomaterial responses. From these combined interests, the field of immunoengineering is rapidly growing. However, bridging immunology with engineering poses numerous challenges including the biological complexity, language of immunology and accurately leveraging the powerful techniques of immunology to new applications. Elucidating the identity and function of immune cell populations responding to engineering systems will be required for continued advancement. Multi-color flow cytometry is a central technique used by immunologists for this purpose that requires careful control of variables, data acquisition, and interpretation. Here, we present methods for multi-color flow cytometry experimental design and analysis focused on characterizing the scaffold immune microenvironment in regenerative medicine research.


Assuntos
Doenças Autoimunes/imunologia , Materiais Biocompatíveis/análise , Citometria de Fluxo , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Animais , Doenças Autoimunes/patologia , Humanos , Neoplasias/patologia , Medicina Regenerativa , Alicerces Teciduais
12.
Sci Transl Med ; 11(477)2019 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-30700576

RESUMO

Biomaterials in regenerative medicine are designed to mimic and modulate tissue environments to promote repair. Biologic scaffolds (derived from decellularized tissue extracellular matrix) promote a wound-healing (proregenerative) immune phenotype and are used clinically to treat tissue loss, including in the context of tumor resection. It is unknown whether a biomaterial microenvironment that encourages tissue formation may also promote tumor development. We implanted a urinary bladder matrix (UBM) scaffold, which is used clinically for wound management, with syngeneic cancer cell lines in mice to study how wound-healing immune responses affect tumor formation and sensitivity to immune checkpoint blockade. The UBM scaffold created an immune microenvironment that inhibited B16-F10 melanoma tumor formation in a CD4+ T cell-dependent and macrophage-dependent manner. In-depth immune characterization revealed an activated type 2-like immune response that was distinct from the classical tumor microenvironment, including activated type 2 T helper T cells, a unique macrophage phenotype, eosinophil infiltration, angiogenic factors, and complement. Tumor growth inhibition by PD-1 and PD-L1 checkpoint blockade was potentiated in the UBM scaffold immune microenvironment. Engineering the local tumor microenvironment to promote a type 2 wound-healing immune signature may serve as a therapeutic target to improve immunotherapy efficacy.


Assuntos
Materiais Biocompatíveis/farmacologia , Carcinogênese/imunologia , Carcinogênese/patologia , Imunoterapia , Alicerces Teciduais/química , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Inflamação/patologia , Interleucina-4/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Melanoma Experimental/patologia , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Fenótipo , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Bexiga Urinária/fisiologia , Bexiga Urinária/ultraestrutura , Cicatrização/efeitos dos fármacos
13.
Biomaterials ; 192: 405-415, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30500722

RESUMO

The immune system plays a critical role in wound healing and the response to biomaterials. Biomaterials-directed regenerative immunology is an immunoengineering strategy that targets the immune system to promote tissue repair. Biomaterial scaffolds employed in regenerative medicine can be broadly classified as biological (such as those derived from the tissue extracellular matrix) or synthetic. Here, we show in depth the divergent myeloid response to biological versus synthetic biomaterial scaffolds. While neutrophil depletion and changes in physical properties such as shape and mechanics can modulate the pro-inflammatory myeloid immune response to synthetic materials to a degree, the overall general divergent myeloid responses persist. Biologic scaffolds elicit a type-2-like immune response with upregulation of genes such as Il4, Cd163, Mrc1 and Chil3, as well as genes associated with damage-associated molecular patterns providing another possible mechanism by which ECM scaffolds promote wound healing via amplification of endogenous wound-associated signaling pathways. Synthetic materials recruit a high proportion of neutrophils which is compounded by material stiffness and by the presence of an injury. Understanding the complex immune response to biomaterial classes will help in the efficient design of immunoengineering strategies and optimizing regenerative and reducing foreign body fibrotic responses to scaffolds.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Inflamação/etiologia , Macrófagos/imunologia , Alicerces Teciduais/efeitos adversos , Animais , Materiais Biocompatíveis/química , Feminino , Imunidade , Inflamação/imunologia , Camundongos Endogâmicos C57BL , Alicerces Teciduais/química
14.
Acta Biomater ; 85: 192-202, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30579044

RESUMO

Biological materials derived from the extracellular matrix (ECM) of tissues serve as scaffolds for rebuilding tissues and for improved wound healing. Cornea trauma represents a wound healing challenge as the default repair pathway can result in fibrosis and scar formation that limit vision. Effective treatments are needed to reduce inflammation, promote tissue repair, and retain the tissue's native transparency and vision capacity. Tissue microparticles derived from cornea, cartilage and lymph nodes were processed and screened in vitro for their ability to reduce inflammation in ocular surface cells isolated from the cornea stroma, conjunctiva, and lacrimal gland. Addition of ECM particles to the media reduced expression of inflammatory genes and restored certain tear film protein production in vitro. Particles derived from lymph nodes were then applied to a rabbit lamellar keratectomy corneal injury model. Application of the tissue particles in a fibrin glue carrier decreased expression of inflammatory and fibrotic genes and scar formation as measured through imaging, histology and immunohistochemistry. In sum, immunomodulatory tissue microparticles may provide a new therapeutic tool for reducing inflammation in the cornea and ocular surface and promoting tissue repair. STATEMENT OF SIGNIFICANCE: Damaged cornea will result in scar tissue formation that impedes vision, and new therapies are needed to enhance wound healing in the cornea and to prevent fibrosis. We evaluated the effects of biological scaffolds derived extracellular matrix (ECM) during corneal wound healing. These ECM particles reduced inflammatory gene expression and restored tear film production in vitro, and reduced scar formation and fibrosis genes in the wounded cornea, when applied to in vivo lamellar keratectomy injury model. The immunomodulatory tissue microparticles may provide a new therapeutic tool for reducing inflammation in the cornea and ocular surface and promoting proper tissue repair.


Assuntos
Micropartículas Derivadas de Células/patologia , Córnea/patologia , Inflamação/patologia , Cicatrização , Animais , Cicatriz/patologia , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Fibrose , Imunomodulação , Ceratectomia , Coelhos
15.
J Biol Chem ; 293(40): 15594-15605, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30139748

RESUMO

Recent studies have reported that the immune system significantly mediates skeletal muscle repair and regeneration. Additionally, biological scaffolds have been shown to play a role in polarizing the immune microenvironment toward pro-myogenic outcomes. Moreover, myostatin inhibitors are known to promote muscle regeneration and ameliorate fibrosis in animal models of Duchenne muscular dystrophy (DMD), a human disease characterized by chronic muscle degeneration. Biological scaffolds and myostatin inhibition can potentially influence immune-mediated regeneration in the dystrophic environment, but have not been evaluated together. Toward this end, here we created an injectable biological scaffold composed of hyaluronic acid and processed skeletal muscle extracellular matrix. This material formed a cytocompatible hydrogel at physiological temperatures in vitro When injected subfascially above the tibialis anterior muscles of both WT and dystrophic mdx-5Cv mice, a murine model of DMD, the hydrogel spreads across the entire muscle before completely degrading at 3 weeks in vivo We found that the hydrogel is associated with CD206+ pro-regenerative macrophage polarization and elevated anti-inflammatory cytokine expression in both WT and dystrophic mice. Co-injection of both hydrogel and myostatin inhibitor significantly increased FoxP3+ regulatory T cell modulation and Foxp3 gene expression in the scaffold immune microenvironment. Finally, delivery of myostatin inhibitor with the hydrogel increased its bioactivity in vivo, and transplantation of immortalized human myoblasts with the hydrogel promoted their survival in vivo This study identifies a key role for biological scaffolds and myostatin inhibitors in modulating a pro-regenerative immune microenvironment in dystrophic muscle.


Assuntos
Anticorpos Monoclonais/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Imunidade Inata/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Miostatina/antagonistas & inibidores , Regeneração/efeitos dos fármacos , Implantes Absorvíveis , Animais , Matriz Extracelular/química , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica , Humanos , Ácido Hialurônico/química , Hidrogéis/química , Imunidade Inata/genética , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Endogâmicos mdx , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/imunologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/imunologia , Distrofia Muscular Animal/patologia , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/imunologia , Miostatina/genética , Miostatina/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Regeneração/genética , Regeneração/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Alicerces Teciduais
16.
ACS Appl Mater Interfaces ; 10(17): 14559-14569, 2018 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-29613762

RESUMO

Severe damage to the ocular surface can result in limbal stem cell (LSC) deficiency, which contributes to loss of corneal clarity, potential vision loss, chronic pain, photophobia, and keratoplasty failure. Human amniotic membrane (AM) is the most effective substrate for LSC transplantation to treat patients with LSC deficiency. However, the widespread use of the AM in the clinic remains a challenge because of the high cost for preserving freshly prepared AM and the weak mechanical strength of lyophilized AM. Here, we developed a novel composite membrane consisting of an electrospun bioabsorbable polymer fiber mesh bonded to a decellularized AM (dAM) sheet through interfacial conjugation. This membrane engineering approach drastically improved the tensile property and toughness of dAM, preserved similar levels of bioactivities as the dAM itself in supporting LSC attachment, growth, and maintenance, and retained significant anti-inflammatory capacity. These results demonstrate that the lyophilized nanofiber-dAM composite membrane offers superior mechanical properties for easy handling and suturing to the dAM, while presenting biochemical cues and basement membrane structure to facilitate LSC transplantation. This composite membrane exhibits major advantages for clinical applications in treating soft tissue damage and LSC deficiency.


Assuntos
Nanofibras/química , Âmnio , Membrana Basal , Córnea , Humanos
17.
J Tissue Eng Regen Med ; 12(3): 821-829, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29049869

RESUMO

Structural and biochemical cues of extracellular matrix can substantially influence the differentiation and maturation of cultured retinal pigment epithelial (RPE) cells. In this study, thin collagen vitrigels were engineered to create collagen nanofibrillar structures of different fibril densities in an effort to evaluate the maturation of human embryonic stem cell-derived retinal pigment epithelial (hESC-RPE) cells. The ultrastructure of the different collagen vitrigels was characterized by transmission electron microscopy, and the mechanical properties were evaluated by tensile testing. The pigmentation and polarization of cells, in addition to key RPE marker gene and protein expression levels, were analyzed to determine the differentiation of hESCs on the gels. The hESC-RPE differentiation was most significant in collagen vitrigels with low fibril density with mature collagen fibrils with diameter of around 60 nm and Young's modulus of 2.41 ± 0.13 MPa. This study provides insight into the influence of collagen nanofibrillar structures on hESC-RPE maturation and presents a potential bioengineered substratum for hESC-RPE for future preclinical and clinical applications.


Assuntos
Diferenciação Celular , Colágeno/farmacologia , Células Epiteliais/citologia , Géis/farmacologia , Epitélio Pigmentado da Retina/citologia , Vitrificação , Animais , Bovinos , Polaridade Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Reagentes de Ligações Cruzadas/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/ultraestrutura , Humanos , Pigmentação/efeitos dos fármacos , Poliésteres/farmacologia
18.
Invest Ophthalmol Vis Sci ; 58(11): 4514-4523, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28873177

RESUMO

Purpose: Disappointing results from clinical studies assessing the efficacy of therapies targeting vascular endothelial growth factor (VEGF) for the treatment of pterygia suggest that other angiogenic mediators may also play a role in its development. We therefore explore the relative contribution of VEGF, hypoxia-inducible factor (HIF)-1α (the transcription factor that regulates VEGF expression in ocular neovascular disease), and a second HIF-regulated mediator, angiopoietin-like 4 (ANGPTL4), to the angiogenic phenotype of pterygia. Methods: Expression of HIF-1α, VEGF, and ANGPTL4 were examined in surgically excised pterygia, and in immortalized human (ih) and primary rabbit (pr) conjunctival epithelial cells (CjECs). Endothelial cell (EC) tubule formation assays using media conditioned by ihCjECs in the presence or absence of inducers/inhibitors of HIF-1 or RNA interference (RNAi) targeting VEGF, ANGPTL4, or both were used to assess their relative contribution to the angiogenic potential of these cells. Results: HIF-1α and VEGF expression were detected in 6/6 surgically excised pterygia and localized to CjECs. Accumulation of HIF-1α in was confirmed in ihCjECs and prCjECs, including stratified prCjECs grown on collagen vitrigel, and resulted in expression of VEGF and the promotion of EC tubule formation; the latter effect was partially blocked using RNAi targeting VEGF mRNA expression. We demonstrate expression of a second HIF-regulated angiogenic mediator, ANGPTL4, in CjECs in culture and in surgically excised pterygia. RNAi targeting ANGPTL4 inhibited EC tubule formation and was additive to RNAi targeting VEGF. Conclusions: Our results support the development of therapies targeting both ANGPTL4 and VEGF for the treatment of patients with pterygia.


Assuntos
Angiopoietinas/metabolismo , Túnica Conjuntiva/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neovascularização Patológica/metabolismo , Pterígio/metabolismo , Moduladores da Angiogênese , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Animais , Western Blotting , Células Cultivadas , Digoxina/farmacologia , Endotélio Vascular/fisiologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Fenótipo , Interferência de RNA , RNA Mensageiro/genética , Coelhos , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Adv Drug Deliv Rev ; 114: 184-192, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28712923

RESUMO

The compatibility of biomaterials is critical to their structural and biological function in medical applications. The immune system is the first responder to tissue trauma and to a biomaterial implant. The innate immune effector cells, most notably macrophages, play a significant role in the defense against foreign bodies and the formation of a fibrous capsule around synthetic implants. Alternatively, macrophages participate in the pro-regenerative capacity of tissue-derived biological scaffolds. Research is now elucidating the role of the adaptive immune system, and T cells in particular, in directing macrophage response to synthetic and biological materials. Here, we review basic immune cell types and discuss recent research on the role of the immune system in tissue repair and its potential relevance to scaffold design. We will also discuss new emerging immune cell types relevant to biomaterial responses and tissue repair. Finally, prospects for specifically targeting and modulating the immune response to biomaterial scaffolds for enhancing tissue repair and regeneration will be presented.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Reação a Corpo Estranho/imunologia , Macrófagos/imunologia , Medicina Regenerativa , Linfócitos T/imunologia , Alicerces Teciduais/efeitos adversos , Animais , Humanos , Imunidade Inata , Regeneração/imunologia , Medicina Regenerativa/métodos
20.
Nat Med ; 23(6): 775-781, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28436958

RESUMO

Senescent cells (SnCs) accumulate in many vertebrate tissues with age and contribute to age-related pathologies, presumably through their secretion of factors contributing to the senescence-associated secretory phenotype (SASP). Removal of SnCs delays several pathologies and increases healthy lifespan. Aging and trauma are risk factors for the development of osteoarthritis (OA), a chronic disease characterized by degeneration of articular cartilage leading to pain and physical disability. Senescent chondrocytes are found in cartilage tissue isolated from patients undergoing joint replacement surgery, yet their role in disease pathogenesis is unknown. To test the idea that SnCs might play a causative role in OA, we used the p16-3MR transgenic mouse, which harbors a p16INK4a (Cdkn2a) promoter driving the expression of a fusion protein containing synthetic Renilla luciferase and monomeric red fluorescent protein domains, as well as a truncated form of herpes simplex virus 1 thymidine kinase (HSV-TK). This mouse strain allowed us to selectively follow and remove SnCs after anterior cruciate ligament transection (ACLT). We found that SnCs accumulated in the articular cartilage and synovium after ACLT, and selective elimination of these cells attenuated the development of post-traumatic OA, reduced pain and increased cartilage development. Intra-articular injection of a senolytic molecule that selectively killed SnCs validated these results in transgenic, non-transgenic and aged mice. Selective removal of the SnCs from in vitro cultures of chondrocytes isolated from patients with OA undergoing total knee replacement decreased expression of senescent and inflammatory markers while also increasing expression of cartilage tissue extracellular matrix proteins. Collectively, these findings support the use of SnCs as a therapeutic target for treating degenerative joint disease.


Assuntos
Senescência Celular/genética , Condrócitos/metabolismo , Osteoartrite do Joelho/genética , Regeneração/genética , Animais , Ligamento Cruzado Anterior/cirurgia , Lesões do Ligamento Cruzado Anterior/complicações , Antivirais/farmacologia , Cartilagem Articular/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Imunofluorescência , Ganciclovir/farmacologia , Glicosaminoglicanos/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suporte de Carga
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA