Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Front Bioeng Biotechnol ; 10: 954682, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35935504

RESUMO

Stem cells have been introduced as a promising therapy for acute and chronic wounds, including burn injuries. The effects of stem cell-based wound therapies are believed to result from the secreted bioactive molecules produced by stem cells. Therefore, treatments using stem cell-derived conditioned medium (CM) (referred to as secretome) have been proposed as an alternative option for wound care. However, safety and regulatory concerns exist due to the uncharacterized biochemical content and variability across different batches of CM samples. This study presents an alternative treatment strategy to mitigate these concerns by using fully characterized recombinant proteins identified by the CM analysis to promote pro-regenerative healing. This study analyzed the secretome profile generated from human placental stem cell (hPSC) cultures and identified nine predominantly expressed proteins (ANG-1, FGF-7, Follistatin, HGF, IL-6, Insulin, TGFß-1, uPAR, and VEGF) that are known to contribute to wound healing and angiogenesis. These proteins, referred to as s (CMFs), were used in combination to test the effects on human dermal fibroblasts (HDFs). Our results showed that CMF treatment increased the HDF growth and accelerated cell migration and wound closure, similar to stem cell and CM treatments. In addition, the CMF treatment promoted angiogenesis by enhancing new vessel formation. These findings suggest that the defined CMF identified by the CM proteomic analysis could be an effective therapeutic solution for wound healing applications. Our strategy eliminates the regulatory concerns present with stem cell-derived secretomes and could be developed as an off-the-shelf product for immediate wound care and accelerating healing.

2.
Cell Tissue Res ; 386(1): 145-156, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34415395

RESUMO

Alternative methods to obtain mature oocytes are still needed for women with premature ovarian failure (POF). Oogonial stem cells (OSCs), found in adult ovaries, have provided insight into potential paths to treating infertility. Previously, the DDX4 antibody marker alone was utilized to isolate OSCs; however, extensive debate over its location in OSCs versus resulting oocytes (transmembrane or intracytoplasmic) has raised doubt about the identity of these cells. Separate groups, however, have efficiently isolated OSCs using another antibody marker Ifitm3 which is consistently recognized to be transmembrane in location. We hypothesized that by using anti-DDX4 and anti-IFITM3 antibodies, in combination, with MACS, we would improve the yield of isolated OSCs versus using anti-DDX4 antibodies alone. Our study supports earlier findings of OSCs in ovaries during the entire female lifespan: from reproductive age through post-menopausal age. MACS sorting ovarian cells using a the two-marker combination yielded a ~ twofold higher percentage of OSCs from a given mass of ovarian tissue compared to existing single marker methods while minimizing the debate surrounding germline marker selection. During in vitro culture, isolated cells retained the germline phenotype expression of DDX4 and IFITM3 as confirmed by gene expression analysis, demonstrated characteristic germline stem cell self-assembly into embryoid bodies, and formed > 40 µm "oocyte-like" structures that expressed the early oocyte markers GDF9, DAZL, and ZP1. This enhanced and novel method is clinically significant as it could be utilized in the future to more efficiently produce mature, secondary oocytes, for use with IVF/ICSI to treat POF patients.


Assuntos
Pesquisa Biomédica/métodos , Fertilidade/fisiologia , Células-Tronco de Oogônios/metabolismo , Feminino , Humanos
3.
Reprod Sci ; 28(6): 1573-1595, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33877644

RESUMO

Diseases, disorders, and dysfunctions of the female reproductive tract tissues can result in either infertility and/or hormonal imbalance. Current treatment options are limited and often do not result in tissue function restoration, requiring alternative therapeutic approaches. Regenerative medicine offers potential new therapies through the bioengineering of female reproductive tissues. This review focuses on some of the current technologies that could address the restoration of functional female reproductive tissues, including the use of stem cells, biomaterial scaffolds, bio-printing, and bio-fabrication of tissues or organoids. The use of these approaches could also be used to address issues in infertility. Strategies such as cell-based hormone replacement therapy could provide a more natural means of restoring normal ovarian physiology. Engineering of reproductive tissues and organs could serve as a powerful tool for correcting developmental anomalies. Organ-on-a-chip technologies could be used to perform drug screening for personalized medicine approaches and scientific investigations of the complex physiological interactions between the female reproductive tissues and other organ systems. While some of these technologies have already been developed, others have not been translated for clinical application. The continuous evolution of biomaterials and techniques, advances in bioprinting, along with emerging ideas for new approaches, shows a promising future for treating female reproductive tract-related disorders and dysfunctions.


Assuntos
Doenças dos Genitais Femininos/terapia , Genitália Feminina , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Células-Tronco Adultas , Técnicas de Cultura de Células em Três Dimensões , Células-Tronco Embrionárias , Tubas Uterinas , Feminino , Doenças dos Genitais Femininos/fisiopatologia , Genitália Feminina/fisiopatologia , Hormônios Esteroides Gonadais , Humanos , Técnicas de Maturação in Vitro de Oócitos , Oogênese , Ovário , Gravidez , Útero
4.
Cell Tissue Res ; 385(1): 161-171, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33582866

RESUMO

Ovaries are the primary physiological source of female sex hormones, which play a crucial role in maintaining ovarian cycle, determining secondary sexual characteristics and preparing the endometrium for implantation. In vitro follicle engineering has been used to investigate follicle development, including ovarian hormone production and gamete maturation. To engineer functional follicles, culture and expansion of the primary ovarian cells are essential. However, the phenotypic and functional characteristics of primary ovarian cells are often lost during culture. The objective of this study is to develop an optimized culture system for maintaining ovarian cell growth and functionality. Granulosa cells (GCs) and theca cells (TCs) were isolated from female rats. The addition of follicle-stimulating hormone (FSH) or luteinizing hormone (LH) to the basal culture media significantly enhanced the secretion of estradiol from GCs and androstenedione from TCs. Serum concentrations of 5% and 10% had a similar role in promoting ovarian cell expansion and secretion of estradiol and androstenedione hormones from both types of cells. Growth differentiation factor 9 (GDF9), bone morphogenic protein 15 (BMP15), BMP7 and basic fibroblast growth factor (bFGF) enhanced GC proliferation and estradiol production, respectively. Among them, the effect of bFGF was most significant. bFGF also enhanced TC proliferation. When GCs and TCs were cultured in 5% serum, gonadotropin and bFGF-containing medium, they proliferated exponentially throughout the culture period of up to 40 days while maintaining their functional characteristics. Taken together, these results indicate that our medium formula is optimal for maximizing proliferation of functionally differentiated ovarian cells.


Assuntos
Técnicas de Cultura de Células/métodos , Ovário/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Feminino , Técnicas In Vitro , Ratos
5.
Reprod Sci ; 28(6): 1697-1708, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33511540

RESUMO

Although menopausal hormone therapy (MHT) is the most effective approach to managing the loss of ovarian activity, serious side effects have been reported. Cell-based therapy is a promising alternative for MHT. This study constructed engineered ovarian cell spheroids and investigated their endocrine function. Theca and granulosa cells were isolated from ovaries of 10-week-old rats. Two types of engineered ovarian cell spheroids were fabricated through forced aggregation in microwells, multilayered spheroids with centralized granulosa aggregates surrounded by an outer layer of theca cells and mixed ovarian spheroids lacking spatial rearrangement. The ovarian cell spheroids were encapsulated into a collagen gel. Non-aggregated ovarian cells served as controls. The endocrine function of the engineered ovarian spheroids was assessed over 30 days. The structure of the spheroids was well maintained during culture. The secretion of 17ß-estradiol from both types of engineered ovarian cell spheroids was higher than in the control group and increased continuously in a time-dependent manner. Secretion of 17ß-estradiol in the multi-layered ovarian cell spheroids was higher than in the non-layered constructs. Increased secretion of progesterone was detected in the multi-layered ovarian cell spheroids at day 5 of culture and was sustained during the culture period. The initial secretion level of progesterone in the non-layered ovarian cell spheroids was similar to those from the controls and increased significantly from days 21 to 30. An in vitro rat model of engineered ovarian cell spheroids was developed that was capable of secreting sex steroid hormones, indicating that the hormone secreting function of ovaries can be recapitulated ex vivo and potentially adapted for MHT.


Assuntos
Encapsulamento de Células/métodos , Células da Granulosa/citologia , Esferoides Celulares/metabolismo , Células Tecais/citologia , Animais , Técnicas de Cultura de Células em Três Dimensões , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados/análise , Estradiol/metabolismo , Terapia de Reposição de Estrogênios/métodos , Feminino , Menopausa , Progesterona/metabolismo , Ratos , Ratos Endogâmicos F344
7.
J Tissue Eng Regen Med ; 14(10): 1394-1402, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32652851

RESUMO

Human placental stem cells (PSCs) enhance histological and functional recovery in a rodent erectile dysfunction (ED) model. We tested the hypothesis that bioactive factors secreted by PSC (i.e., the secretome) mediate functional recovery and that acellular-conditioned media (CM) from PSC culture (PSC-CM) could be used independently to facilitate functional and histological recovery. To identify factors relative to efficacy of PSC, a comparison of CM from PSC and three additional human stem cell populations was performed. CM from human PSC, amniotic fluid stem cells (AFSCs), adipose-derived stem cells (ADSC), and human umbilical vein endothelial cells (HUVECs) was assayed using a semi-quantitative human cytokine antibody array. Male rats, after surgically created ED by neurovascular injury, were randomly divided into four groups: vehicle control (phosphate-buffered saline [PBS]), PSC, PSC-CM, and serum-free media control (SFM) as control. Functional data on intracorporal and mean arterial pressure were obtained, and histological architecture was examined 6 weeks after single injection. PSCs were found to secrete at least 27 cytokines and growth factors at a significantly higher level than the other three cell types. Either single injection of PSC-CM or PSC significantly improved erectile functional recovery and histological architecture compared with SFM or PBS. Injection of the secretome isolated from human PSC improves erectile functional recovery and histological structure in a rat model of neurovascular injury-induced ED. Further characterization of the unique protein expression within the PSC-CM may help to identify the potential for a novel injectable cell-free therapeutic for applicable patients.


Assuntos
Meios de Cultivo Condicionados/química , Metaboloma , Pelve/irrigação sanguínea , Pelve/lesões , Ereção Peniana , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Líquido Amniótico/citologia , Animais , Biomarcadores/metabolismo , Análise por Conglomerados , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Músculo Liso/metabolismo , Pelve/inervação , Ratos , Recuperação de Função Fisiológica , Regeneração
8.
Sci Rep ; 10(1): 9766, 2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32555384

RESUMO

The blood-brain barrier (BBB) is a dynamic component of the brain-vascular interface that maintains brain homeostasis and regulates solute permeability into brain tissue. The expression of tight junction proteins between adjacent endothelial cells and the presence of efflux proteins prevents entry of foreign substances into the brain parenchyma. BBB dysfunction, however, is evident in many neurological disorders including ischemic stroke, trauma, and chronic neurodegenerative diseases. Currently, major contributors to BBB dysfunction are not well understood. Here, we employed a multicellular 3D neurovascular unit organoid containing human brain microvascular endothelial cells, pericytes, astrocytes, microglia, oligodendrocytes and neurons to model the effects of hypoxia and neuroinflammation on BBB function. Organoids were cultured in hypoxic chamber with 0.1% O2 for 24 hours. Organoids cultured under this hypoxic condition showed increased permeability, pro-inflammatory cytokine production, and increased oxidative stress. The anti-inflammatory agents, secoisolariciresinol diglucoside and 2-arachidonoyl glycerol, demonstrated protection by reducing inflammatory cytokine levels in the organoids under hypoxic conditions. Through the assessment of a free radical scavenger and an anti-inflammatory endocannabinoid, we hereby report the utility of the model in drug development for drug candidates that may reduce the effects of ROS and inflammation under disease conditions. This 3D organoid model recapitulates characteristics of BBB dysfunction under hypoxic physiological conditions and when exposed to exogenous neuroinflammatory mediators and hence may have potential in disease modeling and therapeutic development.


Assuntos
Barreira Hematoencefálica/patologia , Endotélio Vascular/patologia , Hipóxia/fisiopatologia , Inflamação/fisiopatologia , Modelos Biológicos , Neurônios/patologia , Organoides/patologia , Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Permeabilidade da Membrana Celular , Citocinas/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Organoides/efeitos dos fármacos , Organoides/metabolismo , Estresse Oxidativo
9.
J Sex Med ; 17(3): 400-411, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32001204

RESUMO

INTRODUCTION: The human placenta provides a bountiful and noncontroversial source of stem cells which have the potential for regeneration of injured tissue. These cells may restore erectile function after neurovascular tissue injury such as that seen in radical pelvic surgeries and pelvic trauma. AIM: To determine the effect of human placenta-derived stem cells on erectile function recovery and histological changes at various time points in a cavernous nerve injury rat model and to study the fate of injected stem cells throughout the regenerative process. METHODS: Human placental stem cells (PSCs) were dual labeled with monomeric Katushka far red fluorescent protein (mKATE)-renLUC using a lentivirus vector. A pelvic neurovascular injury-induced erectile dysfunction model was established in male, athymic rats by crushing the cavernous nerves and ligating the internal pudendal neurovascular bundles, bilaterally. At the time of defect creation, nonlabeled PSCs were injected into the corpus cavernosum at a concentration of 2.5 × 106 cells/0.2 mL. The phosphate-buffered saline-treated group served as the negative control group, and age-matched rats (age-matched controls) were used as the control group. Erectile function, histomorphological analyses, and Western blot were assessed at 1, 6, and 12 weeks after model creation. The distribution of implanted, dual-labeled PSCs was monitored using an in vivo imaging system (IVIS). Implanted cells were further tracked by detection of mKATE fluorescence in histological sections. MAIN OUTCOME MEASURE: The main outcome measure includes intracavernous pressure/mean arterial pressure ratio, neural, endothelial, smooth muscle cell regeneration, mKATE fluorescence, and IVIS imaging. RESULTS: The ratio of intracavernous pressure to mean arterial pressure significantly increased in PSC-injected rats compared with phosphate-buffered saline controls (P < 0.05) at the 6- and 12-week time points, reaching 72% and 68% of the age-matched control group, respectively. Immunofluorescence staining and Western blot analysis showed significant increases in markers of neurons (84.3%), endothelial cells (70.2%), and smooth muscle cells (70.3%) by 6 weeks in treatment groups compared with negative controls. These results were maintained through 12 weeks. IVIS analysis showed luminescence of implanted PSCs in the injected corpora immediately after injection and migration of cells to the sites of injury, including the incision site and periprostatic vasculature by day 1. mKATE fluorescence data revealed the presence of PSCs in the penile corpora and major pelvic ganglion at 1 and 3 days postoperatively. At 7 days, immunofluorescence of penile PSCs had disappeared and was diminished in the major pelvic ganglion. CLINICAL IMPLICATIONS: Placenta-derived stem cells may represent a future "off-the-shelf" treatment to mitigate against development of erectile dysfunction after radical prostatectomy or other forms of pelvic injury. STRENGTH & LIMITATIONS: Single dose injection of PSCs after injury resulted in maximal functional recovery and tissue regeneration at 6 weeks, and the results were maintained through 12 weeks. Strategies to optimize adult stem cell therapy might achieve more effective outcomes for human clinical trials. CONCLUSION: Human PSC therapy effectively restores the erectile tissue and function in this animal model. Thus, PSC therapy may provide an attractive modality to lessen the incidence of erectile dysfunction after pelvic neurovascular injury. Further improvement in tissue regeneration and functional recovery may be possible using multiple injections or systemic introduction of stem cells. Gu X, Thakker PU, Matz EL, et al. Dynamic Changes in Erectile Function and Histological Architecture After Intracorporal Injection of Human Placental Stem Cells in a Pelvic Neurovascular Injury Rat Model. J Sex Med 2020;17:400-411.


Assuntos
Disfunção Erétil/fisiopatologia , Placenta/citologia , Transplante de Células-Tronco/métodos , Traumatismos do Sistema Nervoso/complicações , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Feminino , Humanos , Plexo Hipogástrico/metabolismo , Masculino , Pelve/patologia , Ereção Peniana/fisiologia , Gravidez , Prostatectomia/efeitos adversos , Ratos , Ratos Nus , Recuperação de Função Fisiológica
10.
Tissue Eng Part A ; 25(21-22): 1493-1503, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30829146

RESUMO

Kidney disease is a major medical problem globally. Chronic kidney disease (CKD) is a progressive loss of kidney function. It causes accumulation of waste and fluid in the body, eventually resulting in kidney failure as well as damaging other organs. Although dialysis and kidney transplantation have been used as primary treatments for renal disease, dialysis does not restore full renal function, and there is a shortage of donor kidneys for transplantation. Recent advances in cell-based therapies have offered a means to augment and restore renal function. Various types of cells have been tested to evaluate their therapeutic effects on injured kidneys. Among various types of cells, amniotic fluid stem cells (AFSCs) share advantages of both embryonic and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects, which may allow their future therapeutic use as an "off-the-shelf" cell source. AFSC presents advantages of both conventional pluripotent and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects. This study demonstrates that administration of human-derived AFSC facilitates functional and structural improvement in a rat model of CKD, and suggests that cell therapy with AFSC has potential as a therapeutic strategy to recover renal function in patients with CKD. Impact Statement Patients with chronic kidney disease (CKD) have limited treatment options, and renal transplantation is the only definitive treatment method that restores kidney function. However, challenges associated with transplantation, including donor organ shortage, rejection, and life-long immunosuppression, remain a problem. Recently, stem cell-based therapies have been proposed as an alternative approach to augment and restore renal function. In this study, we used human-derived amniotic fluid stem cells (AFSCs) to treat CKD in a rat model and demonstrated that AFSC treatment facilitated positive effects in terms of improvements of renal function.


Assuntos
Líquido Amniótico/citologia , Testes de Função Renal , Rim/fisiopatologia , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/terapia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Modelos Animais de Doenças , Humanos , Rim/patologia , Masculino , Podócitos/ultraestrutura , Ratos Nus
11.
Sci Rep ; 9(1): 1856, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755653

RESUMO

The early treatment and rapid closure of acute or chronic wounds is essential for normal healing and prevention of hypertrophic scarring. The use of split thickness autografts is often limited by the availability of a suitable area of healthy donor skin to harvest. Cellular and non-cellular biological skin-equivalents are commonly used as an alternative treatment option for these patients, however these treatments usually involve multiple surgical procedures and associated with high costs of production and repeated wound treatment. Here we describe a novel design and a proof-of-concept validation of a mobile skin bioprinting system that provides rapid on-site management of extensive wounds. Integrated imaging technology facilitated the precise delivery of either autologous or allogeneic dermal fibroblasts and epidermal keratinocytes directly into an injured area, replicating the layered skin structure. Excisional wounds bioprinted with layered autologous dermal fibroblasts and epidermal keratinocytes in a hydrogel carrier showed rapid wound closure, reduced contraction and accelerated re-epithelialization. These regenerated tissues had a dermal structure and composition similar to healthy skin, with extensive collagen deposition arranged in large, organized fibers, extensive mature vascular formation and proliferating keratinocytes.


Assuntos
Bioimpressão/métodos , Pele/citologia , Cicatrização , Animais , Proliferação de Células , Colágeno/química , Células Epidérmicas/citologia , Desenho de Equipamento , Feminino , Fibroblastos/citologia , Humanos , Hidrogéis/química , Queratinócitos/citologia , Camundongos , Camundongos Nus , Estudo de Prova de Conceito , Reepitelização , Pele Artificial , Suínos , Engenharia Tecidual/métodos
12.
Curr Opin Organ Transplant ; 23(6): 649-656, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30234736

RESUMO

PURPOSE OF REVIEW: Loss of organ function is a critical issue that threatens a patient's life. Currently, the only available treatment is organ transplantation; however, shortage of donor organs, histocompatibility, and life-long immunosuppression present major challenges. Three-dimensional bioprinting technology holds a promising solution for treating organ failure by fabricating autologous tissues and organs for transplantation. To biofabricate a functional tissue, target-cell types are combined with an appropriate biomaterial for structural support and a bioink that supports cell function and maturation. Bioprinted structures can mimic the native tissue shape and functionality. RECENT FINDINGS: The main goal of three-dimensional bioprinting is to produce functional tissues/organs; however, whole organ printing has not been achieved. There have been recent advances in the successful three-dimensional bioprinting of numerous tissues. This review will discuss the types of bioprinters, biomaterials, bioinks, and the fabrication of various constructs for repair of vascular, cartilage, skin, cardiac, and liver tissues. These bioprinted tissue constructs have the potential to be used to treat tissues and organs that have been damaged by injury or disease. SUMMARY: Three-dimensional bioprinting technology offers the ability to fabricate three-dimensional tissue structures with high precision, fidelity, and stability at human clinical scale. The creation of complex tissue architectures with heterogeneous compositions has the potential to revolutionize transplantation of tissues and organs.


Assuntos
Bioimpressão/métodos , Impressão Tridimensional/tendências , Engenharia Tecidual/métodos , Humanos
13.
Stem Cells Transl Med ; 7(2): 241-250, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29380564

RESUMO

Host stem/progenitor cells can be mobilized and recruited to a target location using biomaterials, and these cells may be used for in situ tissue regeneration. The objective of this study was to investigate whether host biologic resources could be used to regenerate renal tissue in situ. Collagen hydrogel was injected into the kidneys of normal mice, and rat kidneys that had sustained ischemia/reperfusion injury. After injection, the kidneys of both animal models were examined up to 4 weeks for host tissue response. The infiltrating host cells present within the injection regions expressed renal stem/progenitor cell markers, PAX-2, CD24, and CD133, as well as mesenchymal stem cell marker, CD44. The regenerated renal structures were identified by immunohistochemistry for renal cell specific markers, including synaptopodin and CD31 for glomeruli and cytokeratin and neprilysin for tubules. Quantitatively, the number of glomeruli found in the injected regions was significantly higher when compared to normal regions of renal cortex. This phenomenon occurred in normal and ischemic injured kidneys. Furthermore, the renal function after ischemia/reperfusion injury was recovered after collagen hydrogel injection. These results demonstrate that introduction of biomaterials into the kidney is able to facilitate the regeneration of glomerular and tubular structures in normal and injured kidneys. Such an approach has the potential to become a simple and effective treatment for patients with renal failure. Stem Cells Translational Medicine 2018;7:241-250.


Assuntos
Colágeno/farmacologia , Hidrogéis/farmacologia , Nefropatias/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Antígenos CD/metabolismo , Materiais Biocompatíveis/farmacologia , Biomarcadores/metabolismo , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Traumatismo por Reperfusão/metabolismo
14.
Sci Rep ; 7(1): 8837, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28821762

RESUMO

Many drugs have progressed through preclinical and clinical trials and have been available - for years in some cases - before being recalled by the FDA for unanticipated toxicity in humans. One reason for such poor translation from drug candidate to successful use is a lack of model systems that accurately recapitulate normal tissue function of human organs and their response to drug compounds. Moreover, tissues in the body do not exist in isolation, but reside in a highly integrated and dynamically interactive environment, in which actions in one tissue can affect other downstream tissues. Few engineered model systems, including the growing variety of organoid and organ-on-a-chip platforms, have so far reflected the interactive nature of the human body. To address this challenge, we have developed an assortment of bioengineered tissue organoids and tissue constructs that are integrated in a closed circulatory perfusion system, facilitating inter-organ responses. We describe a three-tissue organ-on-a-chip system, comprised of liver, heart, and lung, and highlight examples of inter-organ responses to drug administration. We observe drug responses that depend on inter-tissue interaction, illustrating the value of multiple tissue integration for in vitro study of both the efficacy of and side effects associated with candidate drugs.


Assuntos
Dispositivos Lab-On-A-Chip , Análise Serial de Tecidos , Descoberta de Drogas/métodos , Desenho de Equipamento , Coração , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Microfluídica/instrumentação , Microfluídica/métodos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Análise Serial de Tecidos/instrumentação , Análise Serial de Tecidos/métodos
15.
Wound Repair Regen ; 25(3): 398-407, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28544322

RESUMO

For treatments requiring split-thickness skin grafts, it is preferable to mesh the grafts. This reduces the amount of excised skin and covers more wound area. The mesh technique, however, destroys surface continuity, which results in scarring. Strain-based bioreactors, on the other hand, have successfully expanded split-thickness skin grafts in vitro within a 7-day period, increasing graft coverage. After in vitro expansion, the expanded skin grafts were tested in a porcine full-thickness excisional wound model. Expanded graft take rate was 100%. Volumetric, histologic, and mechanical assessments indicated that expanded grafts were comparable to unexpanded grafts (positive control). While there was considerable variation in expansion (31% to -3.1%), this technique has the potential to enhance the coverage area of skin grafts while reducing or eliminating scarring.


Assuntos
Queimaduras/patologia , Queimaduras/terapia , Transplante de Pele/métodos , Cicatrização/fisiologia , Animais , Cicatriz/patologia , Cicatriz/prevenção & controle , Modelos Animais de Doenças , Estudos de Viabilidade , Sobrevivência de Enxerto/fisiologia , Imuno-Histoquímica , Transplante de Pele/instrumentação , Suínos , Resistência à Tração/fisiologia , Dispositivos para Expansão de Tecidos
16.
Biomed Mater ; 11(6): 065009, 2016 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-27834314

RESUMO

The in vivo function and phenotype of ovarian follicle cells are determined by many factors. When these cells are removed from the in vivo microenvironment and grown in a 2D in vitro environment, the function of the follicular cells is difficult to preserve. A collagen hydrogel was used to examine the hormone and oocyte maturation of ovary follicles in a 3D culture system. Ovarian follicles from rats were isolated and cultured in various concentration of type I collagen hydrogels ranging from 1% to 7% (weight/volume). Differences in cell survival, follicle growth and development, sex hormone production, and oocyte maturation were seen with the modifications in the collagen hydrogel density and elasticity. The results show the significance of the collagen hydrogel properties on phenotype and function maintenance of the ovarian follicles in a 3D culture system.


Assuntos
Técnicas de Cultura de Células/métodos , Colágeno/química , Hidrogéis/química , Folículo Ovariano/fisiologia , Animais , Sobrevivência Celular , Células Cultivadas , Elasticidade , Estradiol/química , Feminino , Humanos , Meiose , Oócitos/citologia , Fenótipo , Progesterona/química , Ratos , Ratos Endogâmicos F344 , Reologia , Viscosidade
17.
PLoS One ; 11(10): e0164997, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27776163

RESUMO

Chronic kidney disease (CKD) occurs when certain conditions cause the kidneys to gradually lose function. For patients with CKD, renal transplantation is the only treatment option that restores kidney function. In this study, we evaluated primary renal cells obtained from diseased kidneys to determine whether their normal phenotypic and functional characteristics are retained, and could be used for cell therapy. Primary renal cells isolated from both normal kidneys (NK) and diseased kidneys (CKD) showed similar phenotypic characteristics and growth kinetics. The expression levels of renal tubular cell markers, Aquaporin-1 and E-Cadherin, and podocyte-specific markers, WT-1 and Nephrin, were similar in both NK and CKD kidney derived cells. Using fluorescence- activated cell sorting (FACS), specific renal cell populations were identified and included proximal tubular cells (83.1% from NK and 80.3% from CKD kidneys); distal tubular cells (11.03% from NK and 10.9% from CKD kidneys); and podocytes (1.91% from NK and 1.78% from CKD kidneys). Ultra-structural analysis using scanning electron microscopy (SEM) revealed microvilli on the apical surface of cultured cells from NK and CKD samples. Moreover, transmission electron microscopy (TEM) analysis showed a similar organization of tight junctions, desmosomes, and other intracellular structures. The Na+ uptake characteristics of NK and CKD derived renal cells were also similar (24.4 mmol/L and 25 mmol/L, respectively) and no significant differences were observed in the protein uptake and transport characteristics of these two cell isolates. These results show that primary renal cells derived from diseased kidneys such as CKD have similar structural and functional characteristics to their counterparts from a normal healthy kidney (NK) when grown in vitro. This study suggests that cells derived from diseased kidney may be used as an autologous cell source for renal cell therapy, particularly in patients with CKD or end-stage renal disease (ESRD).


Assuntos
Biomarcadores/metabolismo , Terapia Baseada em Transplante de Células e Tecidos/métodos , Rim/citologia , Insuficiência Renal Crônica/terapia , Adolescente , Adulto , Idoso , Separação Celular , Feminino , Citometria de Fluxo , Humanos , Rim/metabolismo , Rim/ultraestrutura , Masculino , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Medicina Regenerativa , Transplante Autólogo
18.
Acta Biomater ; 29: 52-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26596567

RESUMO

The only definitive treatment for end stage renal disease is renal transplantation, however the current shortage of organ donors has resulted in a long list of patients awaiting transplant. Whole organ engineering based on decellularization/recellularization techniques has provided the possibility of creating engineered kidney constructs as an alternative to donor organ transplantation. Previous studies have demonstrated that small units of engineered kidney are able to maintain function in vivo. However, an engineered kidney with sufficient functional capacity to replace normal renal function has not yet been developed. One obstacle in the generation of such an organ is the development of effective cell seeding methods for robust colonization of engineered kidney scaffolds. We have developed cell culture methods that allow primary porcine renal cells to be efficiently expanded while maintaining normal renal phenotype. We have also established an effective cell seeding method for the repopulation of acellular porcine renal scaffolds. Histological and immunohistochemical analyses demonstrate that a majority of the expanded cells are proximal tubular cells, and the seeded cells formed tubule-like structures that express normal renal tubule phenotypic markers. Functional analysis revealed that cells within the kidney construct demonstrated normal renal functions such as re-adsorption of sodium and protein, hydrolase activity, and production of erythropoietin. These structural and functional outcomes suggest that engineered kidney scaffolds may offer an alternative to donor organ transplant. STATEMENT OF SIGNIFICANCE: Kidney transplantation is the only definitive treatment for end stage renal disease, however the current shortage of organ donors has limited the treatment. Whole organ engineering based on decellularization/recellularization techniques has provided the possibility of creating engineered kidney constructs as an alternative to donor organ transplantation. While previous studies have shown that small units of engineered kidneys are able to maintain function in animal studies, engineering of kidneys with sufficient functional capacity to replace normal renal function is still challenging due to inefficient cell seeding methods. This study aims to establish an effective cell seeding method using pig kidney cells for the repopulation of acellular porcine kidney scaffolds, suggesting that engineered kidneys may offer an alternative to donor organ transplant.


Assuntos
Rim/metabolismo , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Antígenos de Diferenciação/metabolismo , Falência Renal Crônica/metabolismo , Falência Renal Crônica/terapia , Suínos
19.
Tissue Eng Part A ; 21(9-10): 1520-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25603924

RESUMO

The reconstruction of large craniofacial defects remains a significant clinical challenge. The complex geometry of facial bone and the lack of suitable donor tissue often hinders successful repair. One strategy to address both of these difficulties is the development of an in vivo bioreactor, where a tissue flap of suitable geometry can be orthotopically grown within the same patient requiring reconstruction. Our group has previously designed such an approach using tissue chambers filled with morcellized bone autograft as a scaffold to autologously generate tissue with a predefined geometry. However, this approach still required donor tissue for filling the tissue chamber. With the recent advances in biodegradable synthetic bone graft materials, it may be possible to minimize this donor tissue by replacing it with synthetic ceramic particles. In addition, these flaps have not previously been transferred to a mandibular defect. In this study, we demonstrate the feasibility of transferring an autologously generated tissue-engineered vascularized bone flap to a mandibular defect in an ovine model, using either morcellized autograft or synthetic bone graft as scaffold material.


Assuntos
Mandíbula/cirurgia , Procedimentos de Cirurgia Plástica , Retalhos Cirúrgicos , Engenharia Tecidual/métodos , Animais , Modelos Animais de Doenças , Feminino , Mandíbula/diagnóstico por imagem , Mandíbula/patologia , Polimetil Metacrilato/química , Implantação de Prótese , Ovinos , Transplante Autólogo , Microtomografia por Raio-X
20.
Cell Transplant ; 24(9): 1887-900, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25197964

RESUMO

X-linked myotubular myopathy (XLMTM) is an isogenic muscle disease characterized by progressive wasting of skeletal muscle, weakness, and premature death of affected male offspring. Recently, the XLMTM gene knock-in mouse, Mtm1 p.R69C, was found to have a similar phenotype as the Mtm1 gene mutation in humans (e.g., central nucleation of small myofibers, attenuated muscle strength, and motor unit potentials). Using this rodent model, we investigated whether syngeneic cell therapy could mitigate muscle weakness. Donor skeletal muscle-derived myoblasts were isolated from C57BL6 wild-type (WT) and Mtm1 p.R69C (KI) mice for transplantation into the gastrocnemius muscle of recipient KI mice. Initial experiments demonstrated that donor skeletal muscle-derived myoblasts from WT and KI mice remained in the gastrocnemius muscle of the recipient KI mouse for up to 4 weeks posttransplantation. KI mice receiving syngeneic skeletal muscle-derived myoblasts displayed an increase in skeletal muscle mass, augmented force generation, and increased nerve-evoked skeletal muscle action potential amplitude. Taken together, these results support our hypothesis that syngeneic cell therapy may potentially be used to ameliorate muscle weakness and delay the progression of XLMTM, as application expands to other muscles.


Assuntos
Mioblastos/transplante , Miopatias Congênitas Estruturais/terapia , Potenciais de Ação , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Genótipo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Proteína MyoD/metabolismo , Mioblastos/citologia , Fator Regulador Miogênico 5/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA