Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Biotechnol Bioeng ; 120(6): 1657-1666, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36810698

RESUMO

Diabetic foot ulcers are a major complication of diabetes that occurs following minor trauma. Diabetes-induced hyperglycemia is a leading factor inducing ulcer formation and manifests notably through the accumulation of advanced glycation end-products (AGEs) such as N-carboxymethyl-lysin. AGEs have a negative impact on angiogenesis, innervation, and reepithelialization causing minor wounds to evolve into chronic ulcers which increases the risks of lower limb amputation. However, the impact of AGEs on wound healing is difficult to model (both in vitro on cells, and in vivo in animals) because it involves a long-term toxic effect. We have developed a tissue-engineered wound healing model made of human keratinocytes, fibroblasts, and endothelial cells cultured in a collagen sponge biomaterial. To mimic the deleterious effects induced by glycation on skin wound healing, the model was treated with 300 µM of glyoxal for 15 days to promote AGEs formation. Glyoxal treatment induced carboxymethyl-lysin accumulation and delayed wound closure in the skin mimicking diabetic ulcers. Moreover, this effect was reversed by the addition of aminoguanidine, an inhibitor of AGEs formation. This in vitro diabetic wound healing model could be a great tool for the screening of new molecules to improve the treatment of diabetic ulcers by preventing glycation.


Assuntos
Diabetes Mellitus , Pé Diabético , Animais , Humanos , Reação de Maillard , Células Endoteliais , Cicatrização , Produtos Finais de Glicação Avançada/farmacologia , Glioxal/farmacologia
2.
Stroke ; 53(4): 1263-1275, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34991336

RESUMO

BACKGROUND: Variants in the ring finger protein 213 (RNF213) gene are known to be associated with increased predisposition to cerebrovascular diseases development. Genomic studies have identified RNF213 as a major risk factor of Moyamoya disease in East Asian descendants. However, little is known about the RNF213 (ring finger protein 213) biological functions or its associated pathogenic mechanisms underlying Moyamoya disease. METHODS: To investigate RNF213 loss-of-function effect in endothelial cell, stable RNF213-deficient human cerebral endothelial cells were generated using the CRISPR-Cas9 genome editing technology. RESULTS: In vitro assays, using RNF213 knockout brain endothelial cells, showed clear morphological changes and increased blood-brain barrier permeability. Downregulation and delocalization of essential interendothelial junction proteins involved in the blood-brain barrier maintenance, such as PECAM-1 (platelet endothelial cell adhesion molecule-1), was also observed. Brain endothelial RNF213-deficient cells also showed an abnormal potential to transmigration of leukocytes and secreted high amounts of proinflammatory cytokines. CONCLUSIONS: Taken together, these results indicate that RNF213 could be a key regulator of cerebral endothelium integrity, whose disruption could be an early pathological mechanism leading to Moyamoya disease. This study also further reinforces the importance of blood-brain barrier integrity in the development of Moyamoya disease and other RNF213-associated diseases.


Assuntos
Adenosina Trifosfatases , Doença de Moyamoya , Ubiquitina-Proteína Ligases , Adenosina Trifosfatases/genética , Células Endoteliais/metabolismo , Endotélio , Predisposição Genética para Doença , Humanos , Doença de Moyamoya/patologia , Fatores de Transcrição , Ubiquitina-Proteína Ligases/genética
3.
Biotechnol Bioeng ; 119(7): 1938-1948, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35289393

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease affecting upper and lower motor neurons (MNs). To investigate whether Schwann cells could be involved in the disease pathogenesis, we developed a tissue-engineered three-dimensional (3D) in vitro model that combined MNs cocultured with astrocytes and microglia seeded on top of a collagen sponge populated with epineurium fibroblasts to enable 3D axonal migration. C2C12 myoblasts were seeded underneath the sponge in the presence or absence of Schwann cells. To reproduce an ALS cellular microenvironment, MNs, astrocytes, and microglia were extracted from SOD1G93A mice recapitulating many aspects of the human disease. This 3D ALS in vitro model was compared with a 3D control made of cells isolated from SOD1WT mice. We showed that normal Schwann cells strongly enhanced MN axonal migration in the 3D control model but had no effect in the ALS model. However, ALS-derived Schwann cells isolated from SOD1G93A mice failed to significantly improve axonal migration in both models. These results suggest that a cell therapy using healthy Schwann cells may not be effective in promoting axonal regeneration in ALS. In addition, this 3D ALS model could be used to study the impact of other cell types on ALS by various combinations of normal and diseased cells.


Assuntos
Esclerose Lateral Amiotrófica , Doenças Neurodegenerativas , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Camundongos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Células de Schwann/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
4.
Exp Dermatol ; 28(12): 1466-1469, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31125475

RESUMO

Recent years have brought an enhanced understanding of keratinocyte contribution to cutaneous nociception. While intra-epidermal nerve endings were classically considered as the exclusive transducers of cutaneous noxious stimuli, it has now been demonstrated that epidermal keratinocytes can initiate nociceptive responses, like Merkel cells do for the innocuous mechanotransduction. In the light of recent in vivo findings, this article outlines this paradigm shift that points to a not yet considered population of sensory epidermal cells.


Assuntos
Queratinócitos/fisiologia , Nociceptividade , Animais , Humanos , Terminações Nervosas/fisiologia
5.
Acta Biomater ; 182: 1-13, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38750917

RESUMO

Skin denervation has been shown to cause remission of psoriatic lesions in patients, which can reappear if reinnervation occurs. This effect can be induced by the activation of dendritic cells through sensory innervation. However, a direct effect of nerves on the proliferation of keratinocytes involved in the formation of psoriatic plaques has not been investigated. We developed, by tissue engineering, a model of psoriatic skin made of patient skin cells that showed increased keratinocyte proliferation and epidermal thickness compared to healthy controls. When this model was treated with CGRP, a neuropeptide released by sensory neurons, an increased keratinocyte proliferation was observed in the psoriatic skin model, but not in the control. When a sensory nerve network was incorporated in the psoriatic model and treated with capsaicin to induce neuropeptide release, an increase of keratinocyte proliferation was confirmed, which was blocked by a CGRP antagonist while no difference was noticed in the innervated healthy control. We showed that sensory neurons can participate directly to keratinocyte hyperproliferation in the formation of psoriatic lesions through the release of CGRP, independently of the immune system. Our unique tissue-engineered innervated psoriatic skin model could be a valuable tool to better understand the mechanism by which nerves may modulate psoriatic lesion formation in humans. STATEMENT OF SIGNIFICANCE: This study shows that keratinocytes extracted from patients' psoriatic skin retain, at least in part, the disease phenotype. Indeed, when combined in a 3D model of tissue-engineered psoriatic skin, keratinocytes exhibited a higher proliferation rate, and produced a thicker epidermis than a healthy skin control. In addition, their hyperproliferation was aggravated by a treatment with CGRP, a neuropeptide released by sensory nerves. In a innervated model of tissue-engineered psoriatic skin, an increase in keratinocyte hyperproliferation was also observed after inducing neurons to release neuropeptides. This effect was prevented by concomitant treatment with an antagonist to CGRP. Thus, this study shows that sensory nerves can directly participate to affect keratinocyte hyperproliferation in psoriasis through CGRP release.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Proliferação de Células , Queratinócitos , Psoríase , Células Receptoras Sensoriais , Engenharia Tecidual , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Psoríase/patologia , Psoríase/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Modelos Biológicos , Feminino , Adulto , Masculino , Pele/inervação , Pele/patologia , Pele/metabolismo
6.
Bioengineering (Basel) ; 10(1)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36671665

RESUMO

The study of neurodegenerative diseases (such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis) is very complex due to the difficulty in investigating the cellular dynamics within nervous tissue. Despite numerous advances in the in vivo study of these diseases, the use of in vitro analyses is proving to be a valuable tool to better understand the mechanisms implicated in these diseases. Although neural cells remain difficult to obtain from patient tissues, access to induced multipotent stem cell production now makes it possible to generate virtually all neural cells involved in these diseases (from neurons to glial cells). Many original 3D culture model approaches are currently being developed (using these different cell types together) to closely mimic degenerative nervous tissue environments. The aim of these approaches is to allow an interaction between glial cells and neurons, which reproduces pathophysiological reality by co-culturing them in structures that recapitulate embryonic development or facilitate axonal migration, local molecule exchange, and myelination (to name a few). This review details the advantages and disadvantages of techniques using scaffolds, spheroids, organoids, 3D bioprinting, microfluidic systems, and organ-on-a-chip strategies to model neurodegenerative diseases.

7.
Tissue Eng Part C Methods ; 29(4): 134-143, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36792923

RESUMO

In the peripheral nervous system, Schwann cells (SCs) play a crucial role in axonal growth, metabolic support of neurons, and the production of myelin sheaths. Expansion of SCs after extraction from human or animal nerves is a long and often low-yielding process. We established a rapid cell culture method using a defined serum-free medium to differentiate human induced pluripotent stem cells (iPSCs) into SCs in only 21 days. The SC identity was characterized by expression of SRY-Box Transcription factor 10 (SOX10), S100b, glial fibrillary acidic protein (GFAP), P75, growth-associated protein 43 (GAP43), and early growth response 2 (EGR2) markers. The SC purity reached 87% as assessed by flow cytometry using the specific SOX10 marker, and 69% based on S100b expression. When SCs were cocultured with iPSC-derived motor neurons two-dimensionally or three-dimensionally (3D), they also expressed the markers of myelin MBP, MPZ, and gliomedin. Likewise, when they were seeded on the opposite side of a porous collagen sponge from motor neurons in the 3D model, they were able to migrate through it and colocalize with motor axons after 8 weeks of maturation. Moreover, they were shown by transmission electron microscopy to form myelin sheaths around motor axons. These results suggest that the use of autologous iPSC-derived SCs for clinical applications such as the repair of peripheral nerve damage, the treatment of spinal cord injuries, or for demyelinating diseases could be a valuable option. Impact Statement Peripheral nerve injuries can cause the complete paralysis of the upper or lower limbs, which considerably reduces the quality of life of patients. To repair this injury, many approaches have been developed by tissue engineering. Combining biomaterials with Schwann cells (SCs) has been shown to be an effective solution for stimulating nerve regeneration. However, the challenge faced concerns the strategy for obtaining autologous SCs to treat patients. A promising approach is to differentiate them from the patient's own cells, previously induced into pluripotent stem cells. We propose a fast culture method to generate functional SCs differentiated from induced pluripotent stem cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Qualidade de Vida , Células de Schwann , Bainha de Mielina/metabolismo , Diferenciação Celular , Regeneração Nervosa/fisiologia , Células Cultivadas
8.
Stem Cells Int ; 2023: 1496597, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37096129

RESUMO

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is an early-onset neurodegenerative disease mainly characterized by spasticity in the lower limbs and poor muscle control. The disease is caused by mutations in the SACS gene leading in most cases to a loss of function of the sacsin protein, which is highly expressed in motor neurons and Purkinje cells. To investigate the impact of the mutated sacsin protein in these cells in vitro, induced pluripotent stem cell- (iPSC-) derived motor neurons and iPSC-derived Purkinje cells were generated from three ARSACS patients. Both types of iPSC-derived neurons expressed the characteristic neuronal markers ß3-tubulin, neurofilaments M and H, as well as specific markers like Islet-1 for motor neurons, and parvalbumin or calbindin for Purkinje cells. Compared to controls, iPSC-derived mutated SACS neurons expressed lower amounts of sacsin. In addition, characteristic neurofilament aggregates were detected along the neurites of both iPSC-derived neurons. These results indicate that it is possible to recapitulate in vitro, at least in part, the ARSACS pathological signature in vitro using patient-derived motor neurons and Purkinje cells differentiated from iPSCs. Such an in vitro personalized model of the disease could be useful for the screening of new drugs for the treatment of ARSACS.

9.
J Cell Physiol ; 227(5): 2130-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21769871

RESUMO

Cooperation between endothelial cells and pericytes is essential to the stabilization and maturation of blood microvessels. We developed a unique in vitro tissue-engineered model to study angiogenesis. The human endothelialized reconstructed connective tissue model promotes the formation of a three-dimensional branching network of capillary-like tubes (CLT) with closed lumens. The purpose of this work was to investigate whether pericytes were spontaneously recruited around CLT in the model. We demonstrated that smooth muscle α-actin (SMA)-positive cells were found closely associated with PECAM-1-positive capillaries in the model. Twelve percent (±2.6) of SMA-positive cells were detected along with 15% (±1.64) von Willebrand factor-positive endothelial cells in the culture system after 31 days of in vitro maturation. Conversely, no SMA-positive cells were detected in reconstructed connective tissues made solely of fibroblasts. Knowing that PDGF is a major factor in the recruitment of pericytes, we showed that blockade of the PDGFB receptor using the inhibitor AG1296 induced an overall 5, 2.6, and 2.4-fold decrease in the SMA-positive cells, von Willebrand factor-positive cells, and number of capillaries, respectively. Using combinations of human GFP-positive fibroblasts and endothelial cells, we demonstrated that pericytes were recruited from the fibroblast population in the model. In conclusion, our tissue-engineered culture system promotes the spontaneous formation of a network of capillaries and the recruitment of pericytes derived from fibroblasts. Since pericytes are essential components of the blood microvasculature, this culture system is a powerful model to study angiogenesis and endothelial cell/pericyte interactions in vitro.


Assuntos
Fibroblastos/citologia , Fibroblastos/fisiologia , Neovascularização Fisiológica/fisiologia , Pericitos/citologia , Pericitos/fisiologia , Engenharia Tecidual/métodos , Actinas/metabolismo , Becaplermina , Capilares/citologia , Capilares/fisiologia , Técnicas de Cultura de Células , Células Endoteliais da Veia Umbilical Humana , Humanos , Recém-Nascido , Modelos Cardiovasculares , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de von Willebrand/metabolismo
10.
Biomaterials ; 280: 121269, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34847434

RESUMO

One of the major challenges in the development of a larger and longer nerve conduit for peripheral nerve repair is the limitation in oxygen and nutrient diffusion within the tissue after transplantation preventing Schwann cell and axonal migration. This restriction is due to the slow neovascularization process of the graft starting from both nerve endings. To overcome this limitation, we propose the design of a living tissue-engineered nerve conduit made of an internal tube with a three-dimensional structure supporting axonal migration, which is inserted inside a hollow external tube that plays the role of an epineurium and is strong enough to be stitched to the severed nerve stumps. The internal tube is made of a rolled living fibroblast sheet and can be seeded with endothelial cells to promote the formation of a network containing capillary-like structures which allow rapid inosculation with the host nerve microvasculature after grafting. Human nerve conduits were grafted in immunodeficient rats to bridge a 15 mm sciatic nerve gap. Human capillaries within the pre-vascularized nerve conduit successfully connected to the host circulation 2 weeks after grafting. Twenty-two weeks after surgery, rats transplanted with the nerve conduits had a similar motor function recovery compared to the autograft group. By promoting rapid vascularization of the internal nerve tube from both ends of the nerve stumps, this endothelialized nerve conduit model displays a favorable environment to enhance axonal migration in both larger caliber and longer nerve grafts.


Assuntos
Traumatismos dos Nervos Periféricos , Animais , Células Endoteliais , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/terapia , Ratos , Células de Schwann , Nervo Isquiático/fisiologia , Engenharia Tecidual/métodos
11.
Sci Rep ; 12(1): 19786, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396670

RESUMO

Extracellular matrix (ECM) secretion, deposition and assembly are part of a whole complex biological process influencing the microenvironment and other cellular behaviors. Emerging evidence is attributing a significant role to extracellular vesicles (EVs) and exosomes in a plethora of ECM-associated functions, but the role of dermal fibroblast-derived EVs in paracrine signalling is yet unclear. Herein, we investigated the effect of exosomes isolated from stimulated human dermal fibroblasts. We report that tridimensional (3D) cell culture of dermal fibroblasts promotes secretion of exosomes carrying a large quantity of proteins involved in the formation, organisation and remodelling of the ECM. In our 3D model, gene expression was highly modulated and linked to ECM, cellular migration and proliferation, as well as inflammatory response. Mass spectrometry analysis of exosomal proteins, isolated from 3D cultured fibroblast-conditioned media, revealed ECM protein enrichment, of which many were associated with the matrisome. We also show that the cytokine interleukin 6 (IL-6) is predicted to be central to the signalling pathways related to ECM formation and contributing to cell migration and proliferation. Overall, our data suggest that dermal fibroblast-derived EVs participate in many steps of the establishment of dermis's ECM.


Assuntos
Exossomos , Humanos , Exossomos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Técnicas de Cultura de Células , Fibroblastos/metabolismo , Matriz Extracelular/metabolismo
12.
Med Sci (Paris) ; 37(1): 68-76, 2021 Jan.
Artigo em Francês | MEDLINE | ID: mdl-33492221

RESUMO

The immune system and the sensory nervous system are responsible for perceiving danger under distinct yet complementary forms. In the last few years, neuroimmune interactions have become an important topic of dermatological research for conditions including wound healing, atopic dermatitis and psoriasis. We present here a selection of tridimensional in vitro models that reproduce skin structure and integrate an immune or a sensory function. Future evolutions of such models are expected to greatly contribute in a better understanding of reciprocal influences between sensory nervous system and immune system.


TITLE: Modélisation tridimensionnelle in vitro des systèmes nerveux et immunitaire de la peau. ABSTRACT: Le système immunitaire et le système nerveux sensoriel sont responsables de la perception du danger, sous des formes distinctes mais complémentaires. Ces dernières années, les interactions neuro-immunes se sont imposées comme un axe de recherche important en dermatologie pour comprendre la cicatrisation, la dermatite atopique ou le psoriasis. Nous présentons ici une sélection de modèles tridimensionnels in vitro reproduisant la structure de la peau et intégrant une fonction immunitaire ou sensorielle. Les évolutions futures de ces modèles permettront d'obtenir une vision aussi complète que possible des influences réciproques entre système immunitaire et système nerveux sensoriel.


Assuntos
Modelos Biológicos , Pele/imunologia , Pele/inervação , Técnicas de Cultura de Tecidos , Animais , Células Cultivadas , Humanos , Neuroimunomodulação/fisiologia , Técnicas de Cultura de Órgãos , Pele/patologia , Técnicas de Cultura de Tecidos/métodos , Técnicas de Cultura de Tecidos/tendências , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências , Alicerces Teciduais , Cicatrização/fisiologia
13.
J Neurosci ; 29(13): 4172-88, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19339612

RESUMO

Adult neuronal precursors retain the remarkable capacity to migrate long distances from the posterior (subventricular zone) to the most anterior [olfactory bulb (OB)] parts of the brain. The knowledge about the mechanisms that keep neuronal precursors in the migratory stream and organize this long-distance migration is incomplete. Here we show that blood vessels precisely outline the migratory stream for new neurons in the adult mammalian forebrain. Real-time video imaging of cell migration in the acute slices demonstrate that neuronal precursors are retained in the migratory stream and guided into the OB by blood vessels that serve as a physical substrate for migrating neuroblasts. Our data suggest that endothelial cells of blood vessels synthesize brain-derived neurotrophic factor (BDNF) that fosters neuronal migration via p75NTR expressed on neuroblasts. Interestingly, GABA released from neuroblasts induces Ca(2+)-dependent insertion of high-affinity TrkB receptors on the plasma membrane of astrocytes that trap extracellular BDNF. We hypothesize that this renders BDNF unavailable for p75NTR-expressing migrating cells and leads to their entrance into the stationary period. Our findings provide new insights into the functional organization of substrates that facilitate the long-distance journey of adult neuronal precursors.


Assuntos
Células-Tronco Adultas/fisiologia , Vasos Sanguíneos/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Movimento Celular/fisiologia , Neurônios/fisiologia , Prosencéfalo/fisiologia , Transdução de Sinais/fisiologia , Animais , Astrócitos , Bicuculina/farmacologia , Compostos de Boro/farmacologia , Fator Neurotrófico Derivado do Encéfalo/genética , Bromodesoxiuridina/metabolismo , Cálcio/metabolismo , Movimento Celular/genética , Células Cultivadas , Células Endoteliais/fisiologia , Transportador 1 de Aminoácido Excitatório/genética , Citometria de Fluxo/métodos , Antagonistas GABAérgicos/farmacologia , Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/deficiência , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Vídeo/métodos , Neurônios/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Prosencéfalo/citologia , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Receptor trkB/metabolismo , Receptores de Fator de Crescimento Neural/deficiência , Transdução de Sinais/genética , Técnicas de Cultura de Tecidos , Ácido gama-Aminobutírico/farmacologia
14.
Protein Cell ; 11(4): 239-250, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31907794

RESUMO

Cutaneous nociception is essential to prevent individuals from sustaining injuries. According to the conventional point of view, the responses to noxious stimuli are thought to be exclusively initiated by sensory neurons, whose activity would be at most modulated by keratinocytes. However recent studies have demonstrated that epidermal keratinocytes can also act as primary nociceptive transducers as a supplement to sensory neurons. To enlighten our understanding of cutaneous nociception, this review highlights recent and relevant findings on the cellular and molecular elements that underlie the contribution of epidermal keratinocytes as nociceptive modulators and noxious sensors, both under healthy and pathological conditions.


Assuntos
Queratinócitos/metabolismo , Nociceptividade , Células Receptoras Sensoriais/metabolismo , Animais , Humanos
15.
Tissue Eng Part A ; 26(13-14): 811-822, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32354258

RESUMO

Tissue engineering offers novel therapies for vaginal reconstruction in patients with congenital vaginal agenesis such as Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome. This study aims to reconstruct a prevascularized tissue-engineered model of human vaginal mucosa (HVM) using the self-assembly approach, free of exogenous materials. In this study, a new cell culture method was used to enhance microcapillary network formation while maintaining sufficient biomechanical properties for surgical manipulation. Human vaginal fibroblasts were coseeded with human umbilical vein endothelial cells (HUVECs). Transduction of HUVEC with a vector that allows the expression of both green fluorescent protein (GFP) and luciferase allowed the monitoring of the formation of a microvascular network in vitro and the assessment of the viability and stability of HUVEC in vivo. Two reconstructed vaginal mucosa grafts, a prevascularized, and a nonvascularized control were implanted subcutaneously on the back of 12 female nude mice and monitored for up to 21 days. Prevascularized grafts demonstrated signs of earlier vascularization compared with controls. However, there were no differences in graft survival outcomes in both groups. The finding of mouse red blood cells within GFP-positive capillaries 1 week after implantation demonstrates the capacity of the reconstructed capillary-like network to connect to the host circulation and sustain blood perfusion in vivo. Furthermore, sites of inosculation between GFP-positive HUVEC and mouse endothelial cells were observed within prevascularized grafts. Our results demonstrate that the addition of endothelial cells using a hybrid approach of self-assembly and reseeding generates a mature capillary-like network that has the potential to become functional in vivo, offering an optimized prevascularized HVM model for further translational research. Impact statement This study introduces a prevascularized tissue-engineered model of human vaginal mucosa (HVM), which is adapted for surgical applications. The prevascularization of tissue-engineered grafts aims to enhance graft survival and is an interesting feature for sexual function. Various scaffold-free cell culture methods were tested to reconstruct a mature microcapillary network within HVM grafts while meeting biomechanical needs for surgery. Moreover, this animal study assesses the vascular functionality of prevascularized grafts in vivo, serving as a proof of concept for further translational applications. This research underlines the continuous efforts to optimize current models to closely mimic native tissues and further improve surgical outcomes.


Assuntos
Mucosa/irrigação sanguínea , Mucosa/citologia , Engenharia Tecidual/métodos , Vagina/irrigação sanguínea , Vagina/citologia , Animais , Técnicas de Cultura de Células , Feminino , Humanos , Camundongos , Camundongos Nus , Alicerces Teciduais/química
16.
Med Sci (Paris) ; 25(3): 288-92, 2009 Mar.
Artigo em Francês | MEDLINE | ID: mdl-19361393

RESUMO

The nervous system is extraordinarily complex and exposed to various trauma and degenerative diseases that remain difficult to treat. To facilitate its study, in vitro models were developed by culturing neurons and glial cells in monolayer cultures, or through organotypic cultures of brain or spinal cord slices. These in vitro models were, and are still very helpful for the advancement of neurosciences. However, they are for some studies, either overly simplified, or too complex. The application of tissue engineering to neurosciences offers a new and highly versatile approach to develop accurate models of the nervous system. These models can be engineered in three-dimensions while choosing for each individual component, cellular and molecular, that will compose it. The level of complexity of the model can be adjusted from the simplest to the more complete as needed. For example, through the use of a three-dimensional tissue-engineered model of the spinal cord, it was possible to reproduce the process of myelin sheath formation around motor neuron axons for the first time in vitro. This breakthrough shows the promising potential of tissue engineering in the development of powerful in vitro models of the nervous system. The combination of these models with the use of human adult neurons and glial cells obtained from the differentiation of neural precursor cells isolated from accessible tissues from patients (skin, fat, bone marrow), opens promising perspectives to better understand -neurodegenerative diseases.


Assuntos
Encéfalo/fisiologia , Fenômenos Fisiológicos do Sistema Nervoso/fisiologia , Neurônios/fisiologia , Adulto , Animais , Humanos , Modelos Neurológicos , Neuroglia/fisiologia , Neurônios/citologia , Engenharia Tecidual/métodos
17.
Acta Biomater ; 82: 93-101, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30316025

RESUMO

Cutaneous innervation is increasingly recognized as a major element of skin physiopathology through the neurogenic inflammation driven by neuropeptides that are sensed by endothelial cells and the immune system. To investigate this process in vitro, models of innervated tissue-engineered skin (TES) were developed, yet exclusively with murine sensory neurons extracted from dorsal root ganglions. In order to build a fully human model of innervated TES, we used induced pluripotent stem cells (iPSC) generated from human skin fibroblasts. Nearly 100% of the iPSC differentiated into sensory neurons were shown to express the neuronal markers BRN3A and ß3-tubulin after 19 days of maturation. In addition, these cells were also positive to TRPV1 and neurofilament M, and some of them expressed Substance P, TrkA and TRPA1. When stimulated with molecules inducing neuropeptide release, iPSC-derived neurons released Substance P and CGRP, both in conventional monolayer culture and after seeding in a 3D fibroblast-populated collagen sponge model. Schwann cells, the essential partners of neurons for function and axonal migration, were also successfully differentiated from human iPSC as shown by their expression of the markers S100, GFAP, p75 and SOX10. When cultured for one additional month in the TES model, iPSC-derived neurons seeded at the bottom of the sponge formed a network of neurites spanning the whole TES up to the epidermis, but only when combined with mouse or iPSC-derived Schwann cells. This unique model of human innervated TES should be highly useful for the study of cutaneous neuroinflammation. STATEMENT OF SIGNIFICANCE: The purpose of this work was to develop in vitro an innovative fully human tissue-engineered skin enabling the investigation of the influence of cutaneous innervation on skin pathophysiology. To reach that aim, neurons were differentiated from human induced pluripotent stem cells (iPSCs) generated from normal human skin fibroblasts. This innervated tissue-engineered skin model will be the first one to show iPSC-derived neurons can be successfully used to build a 3D nerve network in vitro. Since innervation has been recently recognized to play a central role in many human skin diseases, such as psoriasis and atopic dermatitis, this construct promises to be at the forefront to model these diseases while using patient-derived cells.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Células de Schwann/metabolismo , Células Receptoras Sensoriais/metabolismo , Pele/inervação , Pele/metabolismo , Engenharia Tecidual , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células de Schwann/citologia , Células Receptoras Sensoriais/citologia , Pele/citologia
18.
J Neurosci Methods ; 163(1): 111-8, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17445905

RESUMO

Neuron-glial interactions are important in development of the nervous system and pathogenesis of disease. Primary cell cultures prepared from nervous tissue are often used to study the properties of individual cell types and how they interact with each other. Isolation of pure populations of cells and their culture is challenging, particularly from murine spinal cord. The purpose of this study was to optimize various protocols to achieve efficient, parallel isolation and purification of primary motor neurons, microglia and astrocytes from the same mouse embryonic spinal cord sample. Following dissociation of E12 embryonic spinal cords, motor neurons were isolated at 97% purity by a single step centrifugation of the cell suspension through multiple discontinuous density gradients of NycoPrep. The residual mixed cell pellet was resuspended and cultured for 2 weeks. Mixed cultures were then shaken to release microglia, which were then harvested from the medium and subjected to another round of differential adhesion to achieve 99% purity. The astrocytes remaining in the mixed cultures were culled to 98% purity by treatment with leucine methyl ester and a subsequent vigorous shaking step to remove any remaining microglia and neurons. Furthermore, no cross contamination was observed in the glial cultures. This technique provides a simple, convenient, and reliable method of obtaining highly purified preparations of motor neurons, microglia and astrocytes from embryonic spinal cord for the study of spinal cord cell biology and motor neuron diseases.


Assuntos
Astrócitos/fisiologia , Técnicas de Cultura de Células , Microglia/fisiologia , Neurônios Motores/fisiologia , Medula Espinal/citologia , Animais , Antígeno CD11b/metabolismo , Células Cultivadas , Colina O-Acetiltransferase/metabolismo , Embrião de Mamíferos , Citometria de Fluxo/métodos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica/métodos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Fatores de Transcrição/metabolismo
19.
Tissue Eng Part B Rev ; 23(1): 59-82, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27609352

RESUMO

Many wound management protocols have been developed to improve wound healing after burn with the primordial aim to restore the barrier function of the skin and also provide a better esthetic outcome. Autologous skin grafts remain the gold standard in the treatment of skin burn, but this treatment has its limitation especially for patients presenting limited donor sites due to extensive burn areas. Deep burn injuries also alter the integrity of skin-sensitive innervation and have an impact on patient's quality of life by compromising perceptions of touch, temperature, and pain. Thus, patients can suffer from long-term disabilities ranging from cutaneous sensibility loss to chronic pain. The cellular mechanisms involved in skin reinnervation following injury are not elucidated yet. Depending on the depth of the burn, nerve sprouting can occur from the wound bed or the surrounding healthy tissue, but somehow this process fails to provide correct reinnervation of the wound during scarring. In addition, several clinical observations indicate that damage to the peripheral nervous system influences wound healing, resulting in delayed wound healing or chronic wounds, underlining the role of innervation and neuromediators for normal cutaneous tissue repair development. Promising tissue engineering strategies, including the use of biomaterials, skin substitutes, and stem cells, could provide novel alternative treatments in wound healing and help in improving patient's sensory recovery.


Assuntos
Queimaduras , Humanos , Qualidade de Vida , Pele , Transplante de Pele , Cicatrização
20.
Biomaterials ; 27(15): 2988-93, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16448695

RESUMO

The use of autologous reconstructed skin appears to be a promising treatment for the permanent coverage of deep and extensive burns. However, the capability of reconstructed skin transplanted on wounds to promote recovery of sensory perception is a major concern. Our aim was to assess the effect of laminin on cutaneous nerve regeneration. We prepared collagen-chitosan sponges enriched with 0, 1, 10 or 50 microg of laminin/sponge to produce tissue-engineered reconstructed skins by culture of human fibroblasts and keratinocytes, then grafted on the back of athymic mice for 120 days. Immunohistochemical studies demonstrated that there were 7 times more neurofilament 150 kD-positive nerve fibers migrating in the graft in the samples enriched with 10 microg laminin/sponge, compared to reconstructed skin without laminin, 120 days after graft. A significant improvement in the current perception threshold of the Abeta and Adelta nerve fibers was measured using a Neurometer in all grafts enriched with laminin. In addition, the type C nerve fibers reached an identical current perception threshold than mouse skin, in all reconstructed skins enriched or not with laminin. We conclude that the use of a tissue-engineered autologous skin graft enriched with laminin has the potential to efficiently optimize cutaneous sensory nerve regeneration in vivo.


Assuntos
Procedimentos Cirúrgicos Dermatológicos , Fibroblastos/transplante , Laminina/administração & dosagem , Regeneração Nervosa/efeitos dos fármacos , Limiar da Dor/fisiologia , Pele/efeitos dos fármacos , Engenharia Tecidual/métodos , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Nus , Pele/lesões , Pele/inervação , Pele/fisiopatologia , Pele Artificial , Resultado do Tratamento , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA