Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
Biomater Adv ; 155: 213669, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37980818

RESUMO

Pelvic Organ Prolapse (POP) is a common gynaecological disorder where pelvic organs protrude into the vagina. While transvaginal mesh surgery using non-degradable polymers was a commonly accepted treatment for POP, it has been associated with high rates of adverse events such as mesh erosion, exposure and inflammation due to serious foreign body response and therefore banned from clinical use after regulatory mandates. This study proposes a tissue engineering strategy using uterine endometrium-derived mesenchymal stem/stromal cells (eMSC) delivered with degradable poly L-lactic acid-co-poly ε-caprolactone (PLACL) and gelatin (G) in form of a composite electrospun nanofibrous mesh (P + G nanomesh) and evaluates the immunomodulatory mechanism at the material interfaces. The study highlights the critical acute and chronic inflammatory markers along with remodelling factors that determine the mesh surgery outcome. We hypothesise that such a bioengineered construct enhances mesh integration and mitigates the Foreign Body Response (FBR) at the host interface associated with mesh complications. Our results show that eMSC-based nanomesh significantly increased 7 genes associated with ECM synthesis and cell adhesion including, Itgb1, Itgb2, Vcam1, Cd44, Cdh2, Tgfb1, Tgfbr1, 6 genes related to angiogenesis including Ang1, Ang2, Vegfa, Pdgfa, Serpin1, Cxcl12, and 5 genes associated with collagen remodelling Col1a1, Col3a1, Col6a1, Col6a2, Col4a5 at six weeks post-implantation. Our findings suggest that cell-based tissue-engineered constructs potentially mitigate the FBR response elicited by biomaterial implants. From a clinical perspective, this construct provides an alternative to current inadequacies in surgical outcomes by modulating the immune response, inducing angiogenesis and ECM synthesis during the acute and chronic phases of the FBR.


Assuntos
Corpos Estranhos , Células-Tronco Mesenquimais , Nanofibras , Feminino , Camundongos , Animais , Gelatina , Engenharia Tecidual/métodos , Diafragma da Pelve , Células-Tronco Mesenquimais/metabolismo , Inflamação/metabolismo , Corpos Estranhos/metabolismo , Regeneração
6.
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.

7.
Front Cell Dev Biol ; 8: 567610, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32984350

RESUMO

Endometrial mesenchymal stem cells (eMSC) drive the extraordinary regenerative capacity of the human endometrium. Clinical application of eMSC for therapeutic purposes is hampered by spontaneous differentiation and cellular senescence upon large-scale expansion in vitro. A83-01, a selective transforming growth factor-ß receptor (TGFß-R) inhibitor, promotes expansion of eMSC in culture by blocking differentiation and senescence, but the underlying mechanisms are incompletely understood. In this study, we combined RNA-seq and ATAC-seq to study the impact of sustained TGFß-R inhibition on gene expression and chromatin architecture of eMSC. Treatment of primary eMSC with A83-01 for 5 weeks resulted in differential expression of 1,463 genes. Gene ontology analysis showed enrichment of genes implicated in cell growth whereas extracellular matrix genes and genes involved in cell fate commitment were downregulated. ATAC-seq analysis demonstrated that sustained TGFß-R inhibition results in opening and closure of 3,555 and 2,412 chromatin loci, respectively. Motif analysis revealed marked enrichment of retinoic acid receptor (RAR) binding sites, which was paralleled by the induction of RARB, encoding retinoic acid receptor beta (RARß). Selective RARß inhibition attenuated proliferation and clonogenicity of A83-01 treated eMSC. Taken together, our study provides new insights into the gene networks and genome-wide chromatin changes that underpin maintenance of an undifferentiated phenotype of eMSC in prolonged culture.

8.
Front Cell Dev Biol ; 8: 497, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32742977

RESUMO

A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFß receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.

9.
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.

10.
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.

11.
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
12.
Stem Cells Dev ; 27(1): 35-43, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29105567

RESUMO

Human endometrial mesenchymal stem cells (eMSCs) are a well-characterized adult stem cell type with potential for use in regenerative medicine or cell therapy. As a proof of principle, we demonstrated that eMSCs promoted wound healing by reducing the inflammatory response through a paracrine action in a subcutaneous rat model of wound repair. However, an efficient protocol for culturing eMSCs in the undifferentiated state and a reliable method of labeling them for cell tracking were lacking. In this study, we investigated the use of a lentiviral vector containing the mCherry fluorescent reporter gene to transduce and label eMSCs following in vitro culturing in A83-01 containing medium, and different methods of tracing the labeled cells following transplantation under the kidney capsule of immunocompromised NSG mice. Perivascular SUSD2+ eMSCs were isolated from human endometrium. Passage 1 eMSCs were transduced by lentiviruses with mCherry fluorescent reporter gene; mCherry+ cells were isolated by fluorescence-activated cell sorting and cultured until passage 6 in 5% O2 in serum-free medium with fibroblast growth factor 2 (FGF2) and epidermal growth factor (EGF). The cells were subsequently divided into two flasks and treated with either dimethyl sulfoxide (0.01%) or A83-01 (1 µM) for 7 days. 5 × 105 control or A83-01 pretreated cells were encapsulated into a fibrin gel and transplanted under the subrenal capsules of NSG mice. Tissues were analyzed at 7, 14, and 30 days posttransplantation. Human eMSCs were efficiently transduced with mCherry gene. They proliferated and maintained high mCherry expression over five passages. Analyzing transplanted kidneys using polymerase chain reaction, flow cytometry, and immunofluorescence showed that both cell types survived at least 30 days. Efficient labeling of eMSCs using a lentiviral vector and culturing them in an environment maintaining them in an undifferentiated state enable reliable detection in preclinical animal models and highlight the need for generating a pure population of undifferentiated MSCs for long-term survival in vivo to prolong their treatment effect.


Assuntos
Endométrio/fisiologia , Rim/fisiologia , Células-Tronco Mesenquimais/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Endométrio/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Rim/metabolismo , Lentivirus/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Cicatrização/fisiologia
13.
Stem Cells Transl Med ; 5(9): 1127-32, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27245365

RESUMO

UNLABELLED: SummaryHuman endometrium is a highly regenerative tissue, undergoing more than 400 cycles of proliferation, differentiation, and shedding during a woman's reproductive life. Adult stem cells, including mesenchymal stem/stromal cells (MSCs), are likely responsible for the immense cellular turnover in human endometrium. The unique properties of MSCs, including high proliferative ability, self-renewal, differentiation to mesodermal lineages, secretion of angiogenic factors, and many other growth-promoting factors make them useful candidates for cellular therapy and tissue engineering. In this review, we summarize the identification and characterization of newly discovered MSCs from the human endometrium: their properties, the surface markers used for their prospective isolation, their perivascular location in the endometrium, and their potential application in cellular therapies. SIGNIFICANCE: The endometrium, or the lining of uterus, has recently been identified as a new and accessible source of mesenchymal stem cells, which can be obtained without anesthesia. Endometrial mesenchymal stem cells have comparable properties to bone marrow and adipose tissue mesenchymal stem cells. Endometrial mesenchymal stem cells are purified with known and novel perivascular surface markers and are currently under investigation for their potential use in cellular therapy for several clinical conditions with significant burden of disease.


Assuntos
Endométrio/citologia , Células-Tronco Mesenquimais , Feminino , Humanos , Transplante de Células-Tronco Mesenquimais/métodos
14.
Acta Biomater ; 39: 114-123, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27163402

RESUMO

UNLABELLED: Pelvic Organ Prolapse (POP) is the herniation of pelvic organs into the vagina. Despite broad acceptance of mesh use in POP surgical repair, the complication rate is unacceptable. We hypothesized that collagen-containing polypropylene (PP) mesh types could modulate mesh-tissue integration and reduce long-term inflammation, thereby reducing mesh-associated complications. This study compared the long-term tissue response to an unmodified PP mesh and two collagen containing meshes in an ovine model which has similar pelvic anatomy and vaginal size to human. Three commercially available macroporous PP meshes, uncoated PP mesh (Avaulta Solo) (PP), the same textile PP mesh layered with a sheet of cross-linked porcine acellular matrix (Avaulta Plus) (PP-ACM) and a different yet also macroporous PP (Sofradim) mesh coated with solubilized atelocollagen (Ugytex) (PP-sCOL) were implanted in the ovine vagina and tissue explanted after 60 and 180days. The macrophage phenotype and response to implanted meshes, and vascularity were quantified by immunostaining and morphometry. We quantified changes in extracellular matrix composition biochemically and collagen organisation and percentage area around the interface of the mesh implants by Sirius Red birefringence and morphometry. PP-ACM induced a more sustained inflammatory response, indicated by similar CD45(+) leukocytes but reduced CD163(+) M2 macrophages at 60days (P<0.05). PP-sCOL increased Von Willebrand Factor (vWF)-immunoreactive vessel profiles after 60days. At the micro-molecular level, collagen birefringence quantification revealed significantly fewer mature collagen fibrils (red, thick fibrils) at the mesh-tissue interface than control tissue for all mesh types (P<0.001) but still significantly greater than the proportion of immature (green thin fibrils) at 60days (P<0.05). The proportion of mature collagen fibrils increased with time around the mesh filaments, particularly those containing collagen. The total collagen percent area at the mesh interface was greatest around the PP-ACM mesh at 60days (P<0.05). By 180days the total mature and immature collagen fibres at the interface of the mesh filaments resembled that of native tissue. In particular, these results suggest that both meshes containing collagen evoke different types of tissue responses at different times during the healing response yet both ultimately lead to physiological tissue formation approaching that of normal tissue. STATEMENT OF SIGNIFICANCE: Pelvic organ prolapse (POP) is the descent of the pelvic organs to the vagina. POP affects more than 25% of all women and the lifetime risk of undergoing POP surgery is 19%. Although synthetic polypropylene (PP) meshes have improved the outcome of the surgical treatment for POP, there was an unacceptable rate of adverse events including mesh exposure and contracture. It is hypothesized that coating the PP meshes with collagen would provide a protective effect by preventing severe mesh adhesions to the wound, resulting in a better controlled initial inflammatory response, and diminished risk of exposure. In this study we assessed the effect of two collagen-containing PP meshes on the long-term vaginal tissue response using new techniques to quantify these tissue responses.


Assuntos
Materiais Revestidos Biocompatíveis/química , Colágeno/química , Matriz Extracelular/química , Teste de Materiais , Polipropilenos/química , Telas Cirúrgicas , Vagina/cirurgia , Animais , Feminino , Ovinos , Suínos , Vagina/lesões , Vagina/metabolismo
15.
Tissue Eng Part A ; 18(15-16): 1720-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22571521

RESUMO

In recent decades, stem cell therapy has been introduced as a novel therapeutic approach for patients suffering from bone disorders. Recently, menstrual blood has been identified as an easily accessible and recycled stem cell source. However, the osteogenic differentiation capacity of menstrual blood-derived stem cells (MenSCs) compared with other adult stem cells remained unsolved. The aim of this study was to investigate the osteogenic differentiation capacity of MenSCs compared to bone marrow-derived mesenchymal stem cells (BMSCs) in the presence of human platelet releasate (HPR). Our results showed that MenSCs were strongly positive for mesenchymal and negative for hematopoietic stem cell markers in a similar manner to BMSCs. In contrary to BMSCs, MenSCs exhibited marked expression of OCT-4 and a significantly higher proliferative capacity. Mineralization, as judged by alizarin red staining, was more pronounced in differentiated BMSCs than in differentiated MenSCs in an osteogenic medium fortified with fetal bovine serum (FBS). However, FBS substitution with HPR in a differentiation medium resulted in typical impact on intensity of MenSC mineralization. The results of semiquantitative reverse transcription-polymerase chain reaction showed comparable levels of parathyroid hormone receptor and osteocalcin transcripts in both types of differentiated stem cells in an HPR medium supplemented with osteogenic inducers. However, the upregulation level of alkaline phosphatase was relatively lower in differentiated MenSCs than that in differentiated BMSCs. We concluded that despite lower osteogenic differentiation capacity of MenSCs compared to BMSCs, substitution of FBS with HPR could equalize the osteogenic differentiation of MenSCs. Therefore, by taking advantage of osteogenic driving potential of HPR, MenSCs could be introduced as an apt and safe alternative to BMSCs for bone tissue-engineering purposes.


Assuntos
Células Sanguíneas/citologia , Plaquetas/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Menstruação , Osteogênese , Células-Tronco/citologia , Adulto , Proliferação de Células , Separação Celular , Forma Celular , Feminino , Antígenos HLA/metabolismo , Humanos , Imunofenotipagem , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Osteoblastos/citologia , Adulto Jovem
16.
Int J Artif Organs ; 35(1): 55-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22307334

RESUMO

INTRODUCTION: The recent identification of menstrual blood-derived stem cells (MenSCs) as a unique population of stem cells has created enormous promise for tissue engineering. In this study, after characterization of MenSCs in comparison with bone marrow-derived stem cells (BMSCs), the potential of MenSCs seeded into electrospun, biodegradable, nanofibrous scaffolds in order to engineer cartilage was evaluated. METHODS: MenSCs and BMSCs were isolated by discontinuous density gradient centrifugation and plastic adherence. After characterization of MenSCs compared with BMSCs, MenSC differentiation into chondrocytes was investigated on a nanofibrous scaffold with specific growth and differentiation factors. The scaffold was prepared from polycaprolactone (PCL) and its surface was modified by plasma treatment. RESULTS: Flow cytometric analysis of expanded cells showed that MenSCs typically express some surface and intracellular markers associated with BMSCs. But marked expression of OCT-4 and the absence of STRO1 distinguished them from mesenchymal stem cells obtained from bone marrow. Based on scanning electron microscope images, the MenSCs were strongly anchored to the highly porous scaffold, which they penetrated and proliferated on. The scaffold contained an extensive cartilage-like extracellular matrix with about 50% greater glycosaminoglycan content than control MenSCs differentiated in a two-dimensional (2D) culture system (p<0.05). Considerable amounts of proteoglycan were produced by the cells differentiated on the scaffold, as demonstrated by Alcian blue staining. Unlike undifferentiated MenSCs, cells differentiated on the scaffold had strong immunoreactivity with monoclonal antibody against collagen type II. CONCLUSIONS: The evidence presented in this study introduces MenSCs as a suitable stem cell population candidate for cartilage tissue engineering.


Assuntos
Diferenciação Celular , Condrócitos/fisiologia , Condrogênese , Menstruação/sangue , Nanofibras , Poliésteres/química , Células-Tronco/fisiologia , Engenharia Tecidual , Alicerces Teciduais , Biomarcadores/metabolismo , Células da Medula Óssea/fisiologia , Adesão Celular , Técnicas de Cultura de Células , Proliferação de Células , Separação Celular , Células Cultivadas , Condrócitos/imunologia , Condrócitos/metabolismo , Condrócitos/ultraestrutura , Colágeno Tipo II/metabolismo , Matriz Extracelular/metabolismo , Feminino , Citometria de Fluxo , Glicosaminoglicanos/metabolismo , Humanos , Imunofenotipagem , Microscopia Eletrônica de Varredura , Nanotecnologia , Fenótipo , Porosidade , Proteoglicanas/metabolismo , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura , Propriedades de Superfície , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA