Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neurourol Urodyn ; 41(2): 592-600, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35094431

RESUMO

AIMS: To measure the force applied along the anterior and posterior vaginal walls in a cohort of 46 patients measured by a fiber-optic pressure sensor and determine if this correlates with vaginal parity and pelvic organ prolapse (POP). METHODS: An intravaginal fiber-optic sensor measured pressure at nine locations along the anterior and posterior vaginal walls during a maximal voluntary pelvic floor muscle contraction (MVC). An automated probe dilation cycle measured the tissue resistance incorporating the vagina and surrounding anatomy. MVC and resting tissue resistance (RTR) were assessed between subjects grouped by the number of vaginal births and prolapse stage. RESULTS: A previous vaginal birth was associated with a significant threefold decrease in the overall anterior pressure measurement during MVC. Decreased anterior pressure measurements were observed at Sensors 1 and 3 (distal vagina) and, posteriorly at Sensors 4-6 (midvagina). Women with Stage 2 posterior prolapse exhibited a decreased MVC pressure in the midvagina than those with Stage 0/1. In this pilot study, there was no difference in the vaginal wall RTR according to previous vaginal birth or stage of prolapse. CONCLUSION: This pilot study found that a decrease in vaginal pressure measured during MVC is associated with vaginal birth and with posterior POP. Greater sample size is required to assess the role of resting tissue pressure measurement.


Assuntos
Diafragma da Pelve , Prolapso de Órgão Pélvico , Feminino , Humanos , Contração Muscular/fisiologia , Diafragma da Pelve/fisiologia , Projetos Piloto , Gravidez , Vagina
2.
Cell Tissue Res ; 385(3): 803-815, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33961124

RESUMO

Mesenchymal stem cells (MSCs) that meet the International Society for Cellular Therapy (ISCT) criteria are obtained from placental tissue by plastic adherence. Historically, no known single marker was available for isolating placental MSCs (pMSCs) from the decidua basalis. As the decidua basalis is derived from the regenerative endometrium, we hypothesised that SUSD2, an endometrial perivascular MSC marker, would purify maternal perivascular pMSC. Perivascular pMSCs were isolated from the maternal placenta using SUSD2 magnetic bead sorting and assessed for the colony-forming unit-fibroblasts (CFU-F), surface markers, and in vitro differentiation into mesodermal lineages. Multi-colour immunofluorescence was used to colocalise SUSD2 and α-SMA, a perivascular marker in the decidua basalis. Placental stromal cell suspensions comprised 5.1%SUSD2+ cells. SUSD2 magnetic bead sorting of the placental stromal cells increased their purity approximately two-fold. SUSD2+ pMSCs displayed greater CFU-F activity than SUSD2- stromal fibroblasts (pSFs). However, both SUSD2+ pMSC and SUSD2- pSF underwent mesodermal differentiation in vitro, and both expressed the ISCT surface markers. Higher percentages of cultured SUSD2+ pMSCs expressed the perivascular markers CD146, CD140b, and SUSD2 than SUSD2- pSFs. These findings suggest that SUSD2 is a single marker that enriches maternal pMSCs, suggesting they may originate from eMSC. Placental decidua basalis can be used as an alternative source of MSC for clinical translation in situations where there is no access to endometrial tissue.


Assuntos
Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Gravidez
3.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567756

RESUMO

Aloe vera (AV), a succulent plant belonging to the Liliaceae family, has been widely used for biomedical and pharmaceutical application. Its popularity stems from several of its bioactive components that have anti-oxidant, anti-microbial, anti-inflammatory and even immunomodulatory effects. Given such unique multi-modal biological impact, AV has been considered as a biomaterial for regenerative medicine and tissue engineering applications, where tissue repair and neo-angiogenesis are vital. This review outlines the growing scientific evidence that demonstrates the advantage of AV as tissue engineering scaffolds. We particularly highlight the recent advances in the application of AV-based scaffolds. From a tissue engineering perspective, it is pivotal that the implanted scaffolds strike an appropriate foreign body response to be well-accepted in the body without complications. Herein, we highlight the key cellular processes that regulate the foreign body response to implanted scaffolds and underline the immunomodulatory effects incurred by AV on the innate and adaptive system. Given that AV has several beneficial components, we discuss the importance of delving deeper into uncovering its action mechanism and thereby improving material design strategies for better tissue engineering constructs for biomedical applications.


Assuntos
Aloe/química , Materiais Biocompatíveis/química , Imunomodulação , Medicina Regenerativa , Alicerces Teciduais/química , Cicatrização , Animais , Humanos
4.
Biomacromolecules ; 20(1): 454-468, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30512928

RESUMO

The current urogynecological clinical meshes trigger unfavorable foreign body response which leads to graft failure in the long term. To overcome the present challenge, we applied a tissue engineering strategy using endometrial SUSD2+ mesenchymal stem cells (eMSCs) with high regenerative properties. This study delves deeper into foreign body response to SUSD2+ eMSC based degradable PLACL/gelatin nanofiber meshes using a mouse model targeted at understanding immunomodulation and mesh integration in the long term. Delivery of cells with nanofiber mesh provides a unique topography that enables entrapment of therapeutic cells for up to 6 weeks that promotes substantial cellular infiltration of host anti-inflammatory macrophages. As a result, degradation rate and tissue integration are highly impacted by eMSCs, revealing an unexpected level of implant integration over 6 weeks in vivo. From a clinical perspective, such immunomodulation may aid in overcoming the current challenges and provide an alternative to an unmet women's urogynecological health need.


Assuntos
Endométrio/citologia , Procedimentos Cirúrgicos em Ginecologia/instrumentação , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Implantes Absorvíveis/efeitos adversos , Animais , Anti-Inflamatórios/química , Células Cultivadas , Feminino , Gelatina/química , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Poliésteres/química , Telas Cirúrgicas/efeitos adversos , Alicerces Teciduais/efeitos adversos
5.
Curr Opin Urol ; 29(4): 450-457, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31008783

RESUMO

PURPOSE OF REVIEW: Nondegradable transvaginal polypropylene meshes for treating pelvic organ prolapse (POP) are now generally unavailable or banned. In this review, we summarize recent developments using tissue engineering approaches combining alternate degradable scaffolds with a novel source of mesenchymal stem/stromal cells from human endometrium (eMSC). RECENT FINDINGS: Tissue engineering constructs comprising immunomodulatory, reparative eMSC and biomimetic materials with nanoarchitecture are a promising approach for vaginal repair and improving outcomes of POP surgery. Culture expansion of eMSC that maintains them (and other MSC) in the undifferentiated state has been achieved using a small molecule transforming growth factor-ß receptor inhibitor, A83-01. The mechanism of action of A83-01 has been determined and its suitability for translation into the clinic explored. Novel blends of electrospun synthetic and natural polymers combined with eMSC shows this approach promotes host cell infiltration and slows biomaterial degradation that has potential to strengthen the vaginal wall during healing. Improving the preclinical ovine transvaginal surgical model by adapting the human clinical POP-Quantification system for selection of multiparous ewes with vaginal wall weakness enables assessment of this autologous eMSC/nanobiomaterial construct. SUMMARY: A tissue engineering approach using autologous eMSC with degradable nanobiomaterials offers a new approach for treating women with POP.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Prolapso de Órgão Pélvico/cirurgia , Células Estromais/transplante , Engenharia Tecidual/métodos , Implantes Absorvíveis , Animais , Modelos Animais de Doenças , Endométrio/citologia , Feminino , Humanos , Células-Tronco Mesenquimais/imunologia , Nanoestruturas/uso terapêutico , Pirazóis/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Ovinos , Telas Cirúrgicas , Tiossemicarbazonas/farmacologia , Alicerces Teciduais , Transplante Autólogo , Vagina/cirurgia
6.
Neurourol Urodyn ; 38(8): 2264-2272, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31385355

RESUMO

AIMS: Pelvic floor disorders (PFDs) in women are a major public health concern. Current clinical methods for assessing PFDs are either subjective or confounded by interference from intra-abdominal pressure (IAP). This study introduces an intravaginal probe that can determine distributed vaginal pressure during voluntary exercises and measures the degree of vaginal tissue support independent of IAP fluctuations. METHODS: An intravaginal probe was fabricated with 18 independent fiber-optic pressure transducers positioned along its upper and lower blades. Continuous pressure measurement along the anterior and posterior vaginal walls during the automated expansion of the probe enabled the resistance of the tissue to be evaluated as a function of displacement, in a manner reflecting the elastic modulus of the tissue. After validation in a simulated vaginal phantom, in vivo measurements were conducted in the relaxed state and during a series of voluntary exercises to gauge the utility of the device in women. RESULTS: The probe reliably detected variations in the composition of sub-surface material in the vaginal phantom. During in-vivo measurements the probe detected distributed tissue elasticity in the absence of IAP change. In addition, the distribution of pressure along both anterior and posterior vaginal walls during cough, Valsalva and pelvic floor contraction was clearly resolved with a large variation observed between subjects. CONCLUSIONS: Our data highlight the potential for the probe to assess the integrity of the vagina wall and support structures as an integrated functional unit. Further in vivo trials are needed to correlate data with clinical findings to assist in the assessment of PFDs.


Assuntos
Exame Ginecológico/instrumentação , Exame Ginecológico/métodos , Vagina/patologia , Adolescente , Adulto , Idoso , Tosse/fisiopatologia , Elasticidade , Exercício Físico , Feminino , Tecnologia de Fibra Óptica , Humanos , Pessoa de Meia-Idade , Contração Muscular , Diafragma da Pelve , Distúrbios do Assoalho Pélvico/diagnóstico , Distúrbios do Assoalho Pélvico/patologia , Imagens de Fantasmas , Pressão , Transdutores , Manobra de Valsalva , Adulto Jovem
7.
Am J Obstet Gynecol ; 214(2): 260.e1-260.e8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26348376

RESUMO

BACKGROUND: Pelvic organ prolapse (POP) is a multifactorial disease that manifests as the herniation of the pelvic organs into the vagina. Surgical methods for prolapse repair involve the use of a synthetic polypropylene mesh. The use of this mesh has led to significantly higher anatomical success rates compared with native tissue repairs, and therefore, despite recent warnings by the Food and Drug Administration regarding the use of vaginal mesh, the number of POP mesh surgeries has increased over the last few years. However, mesh implantation is associated with higher postsurgery complications, including pain and erosion, with higher consecutive rates of reoperation when placed vaginally. Little is known on how the mechanical properties of the implanted mesh itself change in vivo. It is assumed that the mechanical properties of these meshes remain unchanged, with any differences in mechanical properties of the formed mesh-tissue complex attributed to the attached tissue alone. It is likely that any changes in mesh mechanical properties that do occur in vivo will have an impact on the biomechanical properties of the formed mesh-tissue complex. OBJECTIVE: The objective of the study was to assess changes in the multiaxial mechanical properties of synthetic clinical prolapse meshes implanted abdominally for up to 90 days, using a rat model. Another objective of the study was to assess the biomechanical properties of the formed mesh-tissue complex following implantation. STUDY DESIGN: Three nondegradable polypropylene clinical synthetic mesh types for prolapse repair (Gynemesh PS, Polyform Lite, and Restorelle) and a partially degradable polypropylene/polyglecaprone mesh (UltraPro) were mechanically assessed before and after implantation (n = 5/ mesh type) in Sprague Dawley rats for 30 (Gynemesh PS, Polyform Lite, and Restorelle) and 90 (UltraPro and Polyform Lite) days. Stiffness and permanent extension following cyclic loading, and breaking load, of the preimplanted mesh types, explanted mesh-tissue complexes, and explanted meshes were assessed using a multi-axial (ball-burst) method. RESULTS: The 4 clinical meshes varied from each other in weight, thickness, porosity, and pore size and showed significant differences in stiffness and breaking load before implantation. Following 30 days of implantation, the mechanical properties of some mesh types altered, with significant decreases in mesh stiffness and breaking load, and increased permanent extension. After 90 days these changes were more obvious, with significant decreases in stiffness and breaking load and increased permanent extension. Similar biomechanical properties of formed mesh-tissue complexes were observed for mesh types of different preimplant stiffness and structure after 90 days implantation. CONCLUSION: This is the first study to report on intrinsic changes in the mechanical properties of implanted meshes and how these changes have an impact on the estimated tissue contribution of the formed mesh-tissue complex. Decreased mesh stiffness, strength, and increased permanent extension following 90 days of implantation increase the biomechanical contribution of the attached tissue of the formed mesh-tissue complex more than previously thought. This needs to be considered when using meshes for prolapse repair.


Assuntos
Teste de Materiais , Fenômenos Mecânicos , Polipropilenos , Telas Cirúrgicas , Animais , Procedimentos Cirúrgicos em Ginecologia/instrumentação , Prolapso de Órgão Pélvico/cirurgia , Ratos , Ratos Sprague-Dawley
8.
Appl Microbiol Biotechnol ; 98(4): 1807-15, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24402415

RESUMO

Recently, a different class of collagen-like molecules has been identified in numerous bacteria. Initial studies have shown that these collagens are readily produced in Escherichia coli and they have been isolated and purified by various small-scale chromatography approaches. These collagens are non-cytotoxic, are non-immunogenic, and can be produced in much higher yields than mammalian collagens, making them potential new collagens for biomedical materials. One of the major drawbacks with large-scale fermentation of collagens has been appropriate scalable down-stream processing technologies. Like other collagens, the triple helical domains of bacterial collagens are particularly resistant to proteolysis. The present study describes the development and optimization of a simple, scalable procedure using a combination of acid precipitation of the E. coli host proteins, followed by proteolysis of residual host proteins to produce purified collagens in large scale without the use of chromatographic methods.


Assuntos
Biotecnologia/métodos , Colágeno/metabolismo , Proteínas Recombinantes/metabolismo , Biotecnologia/economia , Colágeno/genética , Proteínas Recombinantes/genética
9.
Microb Cell Fact ; 11: 146, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23126526

RESUMO

BACKGROUND: Collagen has proved valuable as biomedical materials for a range of clinical applications, particularly in wound healing. It is normally produced from animal sources, such as from bovines, but concerns have emerged over transmission of diseases. Recombinant collagens would be preferable, but are difficult to produce. Recently, studies have shown that 'collagens' from bacteria, including Streptococcus pyogenes, can be produced in the laboratory as recombinant products, and that these are biocompatible. In the present study we have established that examples of bacterial collagens can be produced in a bioreactor with high yields providing proof of manufacture of this important group of proteins. RESULTS: Production trials in shake flask cultures gave low yields of recombinant product, < 1 g/L. Increased yields, of around 1 g/L, were obtained when the shake flask process was transferred to a stirred tank bioreactor, and the yield was further enhanced to around 10 g/L by implementation of a high cell density fed-batch process and the use of suitably formulated fully defined media. Similar yields were obtained with 2 different constructs, one containing an introduced heparin binding domain. The best yields, of up to 19 g/L were obtained using this high cell density strategy, with an extended 24 h production time. CONCLUSIONS: These data have shown that recombinant bacterial collagen from S. pyogenes, can be produced in sufficient yield by a scalable microbial production process to give commercially acceptable yields for broad use in biomedical applications.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno/metabolismo , Streptococcus pyogenes/metabolismo , Proteínas de Bactérias/genética , Materiais Biocompatíveis/metabolismo , Reatores Biológicos/microbiologia , Colágeno/genética , Meios de Cultura/metabolismo , Escherichia coli/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Temperatura
10.
J Pers Med ; 11(9)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34575617

RESUMO

Cellular therapy is an emerging field in clinical and personalised medicine. Many adult mesenchymal stem/progenitor cells (MSC) or pluripotent derivatives are being assessed simultaneously in preclinical trials for their potential treatment applications in chronic and degenerative human diseases. Endometrial mesenchymal stem/progenitor cells (eMSC) have been identified as clonogenic cells that exist in unique perivascular niches within the uterine endometrium. Compared with MSC isolated from other tissue sources, such as bone marrow and adipose tissue, eMSC can be extracted through less invasive methods of tissue sampling, and they exhibit improvements in potency, proliferative capacity, and control of culture-induced differentiation. In this review, we summarize the potential cell therapy and tissue engineering applications of eMSC in pelvic organ prolapse (POP), emphasising their ability to exert angiogenic and strong immunomodulatory responses that improve tissue integration of novel surgical constructs for POP and promote vaginal tissue healing.

11.
J Heart Valve Dis ; 19(6): 766-71, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21214102

RESUMO

BACKGROUND AND AIM OF THE STUDY: Tissue engineering is an emerging strategy for the development of replacement heart valves where the properties of native tissues are to be replicated. The complexity of the distribution of various collagens in the aortic, mitral, and pulmonary valve leaflets of porcine, bovine, and ovine origin, has been examined. METHODS: Immunohistological and transmission electron microscopy analyses using monoclonal antibodies to types I, III, IV, V and VI collagens were performed. RESULTS: The results indicated that each collagen type has its own distinct distribution, with minimal variation between heart valve anatomic sites and species. Of particular interest was type VI collagen, which had an asymmetric distribution that was principally localized along the outflow surface of the valve. CONCLUSION: Successful tissue engineering constructs of heart valves may need to replicate the complex distribution of different collagens found in heart valve tissues.


Assuntos
Colágeno/análise , Valvas Cardíacas/química , Imuno-Histoquímica , Animais , Valva Aórtica/química , Bovinos , Valvas Cardíacas/ultraestrutura , Microscopia Eletrônica de Transmissão , Valva Mitral/química , Valva Pulmonar/química , Ovinos , Especificidade da Espécie , Suínos
12.
J Mater Sci Mater Med ; 21(4): 1081-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20012163

RESUMO

Porous polyurethane networks containing covalently attached zwitterionic compounds dihydroxypolycaprolactone phosphorylcholine and 1,2-dihydroxy-N,N-dimethylamino-propane sulfonate have been prepared and characterised. Three polymers were prepared by reacting methyl 2,6-diisocyanato hexanoate functionalised D: -glucose as prepolymer A with either polycaprolactone triol alone or with addition of 10 mol% zwitterion as prepolymer B. All polymer compositions were mixed with 10 wt% hydrated gelatin beads. The cured polymers with the gelatin beads showed compression strengths that were still suitable for use in articular cartilage repair. The incorporation of zwitterions yielded more hydrophilic polymers that showed increased water absorption and increased porosity. After four months degradation in phosphate buffered saline, the polymers containing zwitterions had approximately 50% mass loss compared with 30% mass loss for that with polycaprolactone triol alone. All polymers were non-toxic in chondrocyte-based assays. Subcutaneous implantation of these polymers into rats confirmed that the polymers degraded slowly. Only a very mild inflammatory response was observed and the polymers were able to support new, well vascularised tissue formation.


Assuntos
Implantes Absorvíveis , Cartilagem Articular/metabolismo , Poliuretanos/química , Poliuretanos/farmacocinética , Animais , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacocinética , Cartilagem Articular/cirurgia , Células Cultivadas , Força Compressiva/fisiologia , Feminino , Regeneração Tecidual Guiada , Humanos , Interações Hidrofóbicas e Hidrofílicas , Implantes Experimentais , Teste de Materiais , Modelos Biológicos , Poliuretanos/síntese química , Ratos , Ratos Sprague-Dawley , Eletricidade Estática , Propriedades de Superfície , Alicerces Teciduais/química
13.
Nanomaterials (Basel) ; 10(6)2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32517067

RESUMO

Pelvic organ prolapse (POP) is a hidden women's health disorder that impacts 1 in 4 women across all age groups. Surgical intervention has been the only treatment option, often involving non-degradable meshes, with variable results. However, recent reports have highlighted the adverse effects of meshes in the long term, which involve unacceptable rates of erosion, chronic infection and severe pain related to mesh shrinkage. Therefore, there is an urgent unmet need to fabricate of new class of biocompatible meshes for the treatment of POP. This review focuses on the causes for the downfall of commercial meshes, and discusses the use of emerging technologies such as electrospinning and 3D printing to design new meshes. Furthermore, we discuss the impact and advantage of nano-/microstructured alternative meshes over commercial meshes with respect to their tissue integration performance. Considering the key challenges of current meshes, we discuss the potential of cell-based tissue engineering strategies to augment the new class of meshes to improve biocompatibility and immunomodulation. Finally, this review highlights the future direction in designing the new class of mesh to overcome the hurdles of foreign body rejection faced by the traditional meshes, in order to have safe and effective treatment for women in the long term.

14.
J Pers Med ; 10(4)2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271899

RESUMO

Rare perivascular mesenchymal stromal cells (MSCs) with therapeutic properties have been identified in many tissues. Their rarity necessitates extensive in vitro expansion, resulting in spontaneous differentiation, cellular senescence and apoptosis, producing therapeutic products with variable quality and decreased potency. We previously demonstrated that A83-01, a transforming growth factor beta (TGF-ß) receptor inhibitor, maintained clonogenicity and promoted the potency of culture-expanded premenopausal endometrial MSCs using functional assays and whole-transcriptome sequencing. Here, we compared the effects of A83-01 on MSCs derived from postmenopausal endometrium, menstrual blood, placenta decidua-basalis, bone marrow and adipose tissue. Sushi-domain-containing-2 (SUSD2+) and CD34+CD31-CD45- MSCs were isolated. Expanded MSCs were cultured with or without A83-01 for 7 days and assessed for MSC properties. SUSD2 identified perivascular cells in the placental decidua-basalis, and their maternal origin was validated. A83-01 promoted MSC proliferation from all sources except bone marrow and only increased SUSD2 expression and prevented apoptosis in MSCs from endometrial-derived tissues. A83-01 only improved the cloning efficiency of postmenopausal endometrial MSCs (eMSCs), and expanded adipose tissue MSCs (adMSCs) underwent significant senescence, which was mitigated by A83-01. MSCs derived from bone marrow (bmMSCs) were highly apoptotic, but A83-01 was without effect. A83-01 maintained the function and phenotype in MSCs cultured from endometrial, but not other, tissues. Our results also demonstrated that cellular SUSD2 expression directly correlates with the functional phenotype.

15.
Front Pharmacol ; 11: 353, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32265721

RESUMO

PURPOSE: Transvaginal meshes for the treatment of Pelvic Organ Prolapse (POP) have been associated with severe adverse events and have been banned for clinical use in many countries. We recently reported the design of degradable poly L-lactic acid-co-poly ε-caprolactone nanofibrous mesh (P nanomesh) bioengineered with endometrial mesenchymal stem/stromal cells (eMSC) for POP repair. We showed that such bioengineered meshes had high tissue integration as well as immunomodulatory effects in vivo. This study aimed to determine the key molecular players enabling eMSC-based foreign body response modulation. METHODS: SUSD2+ eMSC were purified from single cell suspensions obtained from endometrial biopsies from cycling women by magnetic bead sorting. Electrospun P nanomeshes with and without eMSC were implanted in a NSG mouse skin wound repair model for 1 and 6 weeks. Quantitative PCR was used to assess the expression of extracellular matrix (ECM), cell adhesion, angiogenesis and inflammation genes as log2 fold changes compared to sham controls. Histology and immunostaining were used to visualize the ECM, blood vessels, and multinucleated foreign body giant cells around implants. RESULTS: Bioengineered P nanomesh/eMSC constructs explanted after 6 weeks showed significant increase in 35 genes associated with ECM, ECM regulation, cell adhesion angiogenesis, and immune response in comparison to P nanomesh alone. In the absence of eMSC, acute inflammatory genes were significantly elevated at 1 week. However, in the presence of eMSC, there was an increased expression of anti-inflammatory genes including Mrc1 and Arg1 by 6 weeks. There was formation of multinucleated foreign body giant cells around both implants at 6 weeks that expressed CD206, a M2 macrophage marker. CONCLUSION: This study reveals that eMSC modulate the foreign body response to degradable P nanomeshes in vivo by altering the expression profile of mouse genes. eMSC reduce acute inflammatory and increase ECM synthesis, angiogenesis and anti-inflammatory gene expression at 6 weeks while forming newly synthesized collagen within the nanomeshes and neo-vasculature in close proximity. From a tissue engineering perspective, this is a hallmark of a highly successful implant, suggesting significant potential as alternative surgical constructs for the treatment of POP.

16.
J Mater Sci Mater Med ; 20 Suppl 1: S3-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18379858

RESUMO

This paper reviews the structure, function and applications of collagens as biomaterials. The various formats for collagens, either as tissue-based devices or as reconstituted soluble collagens are discussed. The major emphasis is on the new technologies that are emerging that will lead to new and improved collagen-based medical devices. In particular, the development of recombinant collagens, especially using microorganism systems, is allowing the development of safe and reproducible collagen products. These systems also allow for the development of novel, non-natural structures, for example collagen like structures containing repeats of key functional domains or as chimeric structures where a collagen domain is covalently linked to another biologically active component.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Colágeno/uso terapêutico , Animais , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/uso terapêutico , Colágeno/química , Colágeno/genética , Variação Genética/fisiologia , Humanos , Proteínas Recombinantes/uso terapêutico , Engenharia Tecidual/tendências
17.
Interface Focus ; 9(4): 20180089, 2019 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-31263531

RESUMO

An excessive foreign body response (FBR) has contributed to the adverse events associated with polypropylene mesh usage for augmenting pelvic organ prolapse surgery. Consequently, current biomaterial research considers the critical role of the FBR and now focuses on developing better biocompatible biomaterials rather than using inert implants to improve the clinical outcomes of their use. Tissue engineering approaches using mesenchymal stem cells (MSCs) have improved outcomes over traditional implants in other biological systems through their interaction with macrophages, the main cellular player in the FBR. The unique angiogenic, immunomodulatory and regenerative properties of MSCs have a direct impact on the FBR following biomaterial implantation. In this review, we focus on key aspects of the FBR to tissue-engineered MSC-based implants for supporting pelvic organs and beyond. We also discuss the immunomodulatory effects of the recently discovered endometrial MSCs on the macrophage response to new biomaterials designed for use in pelvic floor reconstructive surgery. We conclude with a focus on considerations in biomaterial design that take into account the FBR and will likely influence the development of the next generation of biomaterials for gynaecological applications.

18.
Acta Biomater ; 97: 162-176, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31386931

RESUMO

Endometrial mesenchymal stem/stromal cells (eMSCs) exhibit excellent regenerative capacity in the endometrial lining of the uterus following menstruation and high proliferative capacity in vitro. Bioprinting eMSCs onto a mesh could be a potential therapy for Pelvic Organ Prolapse (POP). This study reports an alternative treatment strategy targeting vaginal wall repair using bioprinting of eMSCs encapsulated in a hydrogel and 3D melt electrospun mesh to generate a tissue engineering construct. Following a CAD, 3D printed poly ε-caprolactone (PCL) meshes were fabricated using melt electrospinning (MES) at different temperatures using a GMP clinical grade GESIM Bioscaffolder. Electron and atomic force microscopies revealed that MES meshes fabricated at 100 °C and with a speed 20 mm/s had the largest open pore diameter (47.2 ±â€¯11.4 µm) and the lowest strand thickness (121.4 ±â€¯46 µm) that promoted optimal eMSC attachment. An Aloe Vera-Sodium Alginate (AV-ALG) composite based hydrogel was optimised to a 1:1 mixture (1%AV-1%ALG) and eMSCs, purified from human endometrial biopsies, were then bioprinted in this hydrogel onto the MES printed meshes. Acute in vivo foreign body response assessment in NSG mice revealed that eMSC printed on MES constructs promoted tissue integration, eMSC retention and an anti-inflammatory M2 macrophage phenotype characterised by F4/80+CD206+ colocalization. Our results address an unmet medical need highlighting the potential of 3D bioprinted eMSC-MES meshes as an alternative approach to overcome the current challenges with non-degradable knitted meshes in POP treatment. STATEMENT OF SIGNIFICANCE: This study presents the first report of bioprinting mesenchymal stem cells derived from woman endometrium (eMSCs) to boost Pelvic Organ Prolapse (POP) treatment. It impacts over 50% of elderly women with no optimal treatment at present. The overall study is conducted in three stages as fabricating a melt electrospun (MES) mesh, bioprinting eMSCs into a Ca2+ free Aloe Vera-Alginate (AV-Alg) based hydrogel and in vivo study. Our data showed that AV-ALG hydrogel potentially suppresses the foreign body response and further addition of eMSCs triggered a high influx of anti-inflammatory CD206+ M2 macrophages. Our final construct demonstrates a favourable foreign body response to predict expected tissue integration, therefore, provides a potential for developing an alternative treatment for POP.


Assuntos
Células Imobilizadas/transplante , Endométrio/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Prolapso de Órgão Pélvico/terapia , Poliésteres/química , Impressão Tridimensional , Telas Cirúrgicas , Animais , Células Imobilizadas/metabolismo , Células Imobilizadas/patologia , Endométrio/patologia , Feminino , Humanos , Células-Tronco Mesenquimais/patologia , Camundongos , Prolapso de Órgão Pélvico/metabolismo , Prolapso de Órgão Pélvico/patologia
19.
J Biomed Mater Res A ; 107(8): 1763-1770, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30983124

RESUMO

If tolerated in biological environments, recombinant structural proteins offer the advantage that biological cues dictating cell attachment and material degradation can be modified as required for clinical application using genetic engineering. In this study, we investigate the biological response to materials generated from the recombinant honeybee silk protein, AmelF3, a structural protein that can be produced at high levels by fermentation in Escherichia coli. The protein can be readily purified from E. coli host cell proteins after transgenic production and fabricated into various material formats. When implanted subcutaneously according to International Standard ISO 10993 tests, materials generated from the purified recombinant protein were found to be noncytotoxic, inducing a transient weak immunogenic response and a chronic inflammatory response that resolved over time. While preliminary, this study supports the ongoing development of materials generated from this protein for biomedical applications. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1763-1770, 2019.


Assuntos
Abelhas/química , Materiais Biocompatíveis/farmacologia , Proteínas Recombinantes/imunologia , Seda/imunologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Inflamação/patologia , Camundongos , Implantação de Prótese , Ratos Sprague-Dawley , Tela Subcutânea/efeitos dos fármacos , Fatores de Tempo
20.
ACS Appl Mater Interfaces ; 11(49): 45511-45519, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31713411

RESUMO

The development of antifibrotic materials and coatings that can resist the foreign body response (FBR) continues to present a major hurdle in the advancement of current and next-generation implantable medical devices, biosensors, and cell therapies. From an implant perspective, the most important issue associated with the FBR is the prolonged inflammatory response leading to a collagenous capsule that ultimately blocks mass transport and communication between the implant and the surrounding tissue. Up to now, most attempts to reduce the capsule thickness have focused on providing surface coatings that reduce protein fouling and cell attachment. Here, we present an approach that is based on the sustained release of a peptide drug interfering with the FBR. In this study, the biodegradable polymer poly(lactic-co-glycolic) acid (PLGA) was used as a coating releasing the relaxin peptide analogue B7-33, which has been demonstrated to reduce organ fibrosis in animal models. While in vitro protein quantification was used to demonstrate controlled release of the antifibrotic peptide B7-33 from PLGA coatings, an in vitro reporter cell assay was used to demonstrate that B7-33 retains activity against the relaxin family peptide receptor 1 (RXFP1). Subcutaneous implantation of PLGA-coated polypropylene samples in mice with and without the peptide demonstrated a marked reduction in capsule thickness (49.2%) over a 6 week period. It is expected that this novel approach will open the door to a range of new and improved implantable medical devices.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Fibrose/prevenção & controle , Reação a Corpo Estranho/prevenção & controle , Fragmentos de Peptídeos/farmacologia , Relaxina/farmacologia , Animais , Materiais Revestidos Biocompatíveis/química , Humanos , Camundongos , Fragmentos de Peptídeos/química , Peptídeos/química , Peptídeos/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Próteses e Implantes/efeitos adversos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Relaxina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA