co-transplantation

共同移植
  • 文章类型: Journal Article
    糖尿病是一种常见的慢性疾病,传统上需要严重依赖药物治疗。口服药物和外源性胰岛素只能暂时维持血糖水平,不能治愈疾病。大多数患者需要终身注射外源性胰岛素。近年来,胰岛移植的进步显著推进了糖尿病的治疗,允许患者停止外源性胰岛素并避免并发症。最近关于胰岛移植的报道的长期随访结果表明,尽管患者仍然需要免疫疗法,但它们提供了显着的治疗益处。表明未来移植策略的重要性。尽管器官短缺仍然是胰岛移植发展的主要障碍,胰岛细胞的新来源,如干细胞和猪胰岛细胞,已经被提议,并逐步纳入临床研究。进一步研究新的移植部位,例如皮下空间和肠系膜脂肪,可能最终取代传统的门静脉内胰岛细胞输注。此外,胰岛移植中的免疫排斥反应将通过联合应用免疫抑制剂来解决,胰岛封装技术,以及最有前途的间充质干细胞/调节性T细胞和胰岛细胞联合移植细胞治疗。本文综述了胰岛移植的研究进展,并讨论了所面临挑战的研究进展和潜在解决方案。
    Diabetes is a prevalent chronic disease that traditionally requires severe reliance on medication for treatment. Oral medication and exogenous insulin can only temporarily maintain blood glucose levels and do not cure the disease. Most patients need life-long injections of exogenous insulin. In recent years, advances in islet transplantation have significantly advanced the treatment of diabetes, allowing patients to discontinue exogenous insulin and avoid complications.Long-term follow-up results from recent reports on islet transplantation suggest that they provide significant therapeutic benefit although patients still require immunotherapy, suggesting the importance of future transplantation strategies. Although organ shortage remains the primary obstacle for the development of islet transplantation, new sources of islet cells, such as stem cells and porcine islet cells, have been proposed, and are gradually being incorporated into clinical research. Further research on new transplantation sites, such as the subcutaneous space and mesenteric fat, may eventually replace the traditional portal vein intra-islet cell infusion. Additionally, the immunological rejection reaction in islet transplantation will be resolved through the combined application of immunosuppressant agents, islet encapsulation technology, and the most promising mesenchymal stem cells/regulatory T cell and islet cell combined transplantation cell therapy. This review summarizes the progress achieved in islet transplantation, and discusses the research progress and potential solutions to the challenges faced.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰岛移植可能是1型糖尿病(T1DM)患者最有效的治疗技术。然而,这种方法的临床应用面临着许多限制,包括孤立的胰岛细胞凋亡,收件人拒绝,和移植血管重建。间充质干细胞(MSCs)具有抗凋亡,免疫调节,和血管生成特性。这里,我们回顾了最近关于胰岛与MSCs共培养和共移植的研究。我们总结了共同移植的制备方法,特别是共同文化的优点,以及共同移植的效果。积累的实验证据表明,胰岛与MSCs共培养促进胰岛存活,增强胰岛分泌功能,并通过各种移植前的准备来促进胰岛的生长。本文旨在为探索MSCs在临床胰岛共移植中的应用提供参考。
    Islet transplantation may be the most efficient therapeutic technique for patients with type 1 diabetes mellitus (T1DM). However, the clinical application of this method is faced with numerous limitations, including isolated islet apoptosis, recipient rejection, and graft vascular reconstruction. Mesenchymal stem cells (MSCs) possess anti-apoptotic, immunomodulatory, and angiogenic properties. Here, we review recent studies on co-culture and co-transplantation of islets with MSCs. We have summarized the methods of preparation of co-transplantation, especially the merits of co-culture, and the effects of co-transplantation. Accumulating experimental evidence shows that co-culture of islets with MSCs promotes islet survival, enhances islet secretory function, and prevascularizes islets through various pretransplant preparations. This review is expected to provide a reference for exploring the use of MSCs for clinical islet co-transplantation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:骨髓移植(BMT)可以应用于造血和非造血疾病;尽管如此,它仍然伴随着许多挑战和限制,导致治疗失败.记住这一点,增加BMT成功率的可能方法是间充质干细胞(MSCs)和造血干细胞(HSCs)的共移植,以改善骨髓生态位并分泌增强造血植入的分子。
    目的:分析小鼠模型中HSC和MSC的特性及其相互作用。
    方法:我们搜索了过去十年在PubMed和Scopus中索引的原始文章,这些文章使用HSC和MSC共移植以及在动物模型中进行体内BMT,同时评估细胞移植。我们排除了体外研究或涉及移植物抗宿主病或其他血液系统疾病的研究以及英语以外语言的出版物。在PubMed中,我们最初确定了555篇文章,经过选择,只有12人被选中。在Scopus,2010年被确定,筛选和资格程序后剩下6人。
    结果:在数据库中找到的2565篇文章中,只有18项原始研究符合资格标准.按来源划分的HSC分布显示出相似的比率,人脐带血或动物骨髓主要通过静脉内或体内途径以1×107个细胞的剂量给药。然而,MSCs在人类捐献者中的患病率很高,来源多种多样(脐带血,骨髓,扁桃体,脂肪组织或胎儿肺),使用较低的剂量,主要是106个细胞,范围为104到1.5×107个细胞,使用相同的路线。在几乎每个实验中,在施用之前表征MSC。使用的受体主要是接受低剂量照射或化疗的免疫缺陷小鼠。移植用于HSC和MSC共移植评估的主要技术是嵌合体,然后进行造血重建和生存分析。除了雕刻,归巢和细胞性也在一些研究中进行了评估。
    结论:临床前研究结果验证了MSCs在异种和异基因造血细胞移植动物模型中体内移植HSC的潜力,在没有毒性的情况下。
    BACKGROUND: Bone marrow transplantation (BMT) can be applied to both hematopoietic and nonhematopoietic diseases; nonetheless, it still comes with a number of challenges and limitations that contribute to treatment failure. Bearing this in mind, a possible way to increase the success rate of BMT would be cotransplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) to improve the bone marrow niche and secrete molecules that enhance the hematopoietic engraftment.
    OBJECTIVE: To analyze HSC and MSC characteristics and their interactions through cotransplantation in murine models.
    METHODS: We searched for original articles indexed in PubMed and Scopus during the last decade that used HSC and MSC cotransplantation and in vivo BMT in animal models while evaluating cell engraftment. We excluded in vitro studies or studies that involved graft versus host disease or other hematological diseases and publications in languages other than English. In PubMed, we initially identified 555 articles and after selection, only 12 were chosen. In Scopus, 2010 were identified, and six were left after the screening and eligibility process.
    RESULTS: Of the 2565 articles found in the databases, only 18 original studies met the eligibility criteria. HSC distribution by source showed similar ratios, with human umbilical cord blood or animal bone marrow being administered mainly with a dose of 1 × 107 cells by intravenous or intrabone routes. However, MSCs had a high prevalence of human donors with a variety of sources (umbilical cord blood, bone marrow, tonsil, adipose tissue or fetal lung), using a lower dose, mainly 106 cells and ranging 104 to 1.5 × 107 cells, utilizing the same routes. MSCs were characterized prior to administration in almost every experiment. The recipient used was mostly immunodeficient mice submitted to low-dose irradiation or chemotherapy. The main technique of engraftment for HSC and MSC cotransplantation evaluation was chimerism, followed by hematopoietic reconstitution and survival analysis. Besides the engraftment, homing and cellularity were also evaluated in some studies.
    CONCLUSIONS: The preclinical findings validate the potential of MSCs to enable HSC engraftment in vivo in both xenogeneic and allogeneic hematopoietic cell transplantation animal models, in the absence of toxicity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在临床前实验和临床试验中,间充质干细胞(MSC)已被证明对股骨头坏死(ONFH)产生积极影响。股骨头缺血性坏死后,移植的MSCs在这种微环境中经历了大量的应激诱导的凋亡和衰老。所以,在ONFH中,骨髓间充质干细胞在坏死区的存活和分化尤为重要。虽然MSCs和内皮细胞(ECs)共培养增强了MSCs的增殖和成骨分化,并在体内形成更成熟的脉管系统,目前尚不清楚共培养细胞是否能够修复ONFH。在这项研究中,我们探讨了血管紧张素II(AngII)-MSCs和ECs共同移植在修复早期ONFH中的作用和机制。体外,当MSCs和ECs以5:1的比例共培养时,两种类型的细胞都设法增殖并诱导成骨和血管生成。然后,我们建立了兔激素诱导的ONFH模型,并通过核减压通道将AngII-MSCs和ECs共同移植。四周后,组织学和蛋白质印迹分析显示,用AngII-MSCs和ECs处理的ONFH可能通过增加I型胶原的表达来促进骨化和血运重建。矮小相关转录因子2,骨钙蛋白,股骨头血管内皮生长因子。我们的数据表明AngII-MSCs和ECs的共同移植能够通过促进成骨和血管生成来挽救早期类固醇诱导的ONFH。这可能被认为是一种在临床上治疗ONFH的新疗法。
    Mesenchymal stem cells (MSCs) have been shown to exert a positive impact on osteonecrosis of the femoral head (ONFH) in preclinical experiments and clinical trials. After the femoral head suffers avascular necrosis, the transplanted MSCs undergo a great deal of stress-induced apoptosis and senescence in this microenvironment. So, survival and differentiation of MSCs in osteonecrotic areas are especially important in ONFH. Although MSCs and endothelial cells (ECs) co-culture enhancing proliferation and osteogenic differentiation of MSCs and form more mature vasculature in vivo, it remains unknown whether the co-culture cells are able to repair ONFH. In this study, we explored the roles and mechanisms of co-transplantation of angiotensin II (Ang II)-MSCs and ECs in repairing early ONFH. In vitro, when MSCs and ECs were co-cultured in a ratio of 5:1, both types of cells managed to proliferate and induce both osteogenesis and angiogenesis. Then, we established a rabbit model of steroid-induced ONFH and co-transplantation of Ang II-MSCs and ECs through the tunnel of core decompression. Four weeks later, histological and Western blot analyses revealed that ONFH treated with Ang II-MSCs and ECs may promote ossification and revascularization by increasing the expression of collagen type I, runt-related transcription factor 2, osteocalcin, and vascular endothelial growth factor in the femoral head. Our data suggest that co-transplantation of Ang II-MSCs and ECs was able to rescue the early steroid-induced ONFH via promoting osteogenesis and angiogenesis, which may be regarded as a novel therapy for the treatment of ONFH in a clinical setting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    我们研究了使用脱氧核糖核酸钠(Derinat)提高间充质(MSC)和造血干细胞(HSC)共移植到雌性F1(CBA×C57BL/6)小鼠骨髓增生的效率的可能性暴露于γ辐射。发现免疫调节剂Derinat增强了共移植的效果,特别是,从外周血参数判断Derinat的三重照射后加速造血恢复,骨髓和脾脏的细胞总数,动物的生存在照射前单次或两次施用Derinat无效。当应用以下方案时,获得最佳结果:MSC→HSC,在辐射和Derinat的三重给药后的第一个小时内开始,间隔为48小时(在10-15分钟内,照射后3和7天),剂量为3mg/小鼠。
    We studied the possibility of using sodium deoxyribonucleate (Derinat) for improving the efficiency of co-transplantation of mesenchymal (MSC) and hematopoietic stem cells (HSC) to female F1(CBA×C57BL/6) mice with bone marrow aplasia caused by exposure to γ-radiation. It was found that immunomodulator Derinat enhanced the effect of co-transplantation, in particular, triple post-irradiation administration of Derinat accelerated hematopoiesis recovery judging from the parameters of peripheral blood, total cellularity of the bone marrow and spleen, and animal survival. Single or double administration of Derinat prior to irradiation was ineffective. The optimal result was obtained when the following scheme was applied: MSC→HSC with an interval of 48 h starting during the first hours after irradiation and triple administration of Derinat (in 10-15 min, 3 and 7 days after irradiation) in a dose of 3 mg/mouse.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    脂肪来源的间充质干细胞/基质细胞(ASC)是能够自我更新并分化成多种细胞系的成体干细胞群。ASC由于其促进血管形成的能力而为治疗性血管生成提供了有希望的未来。具体来说,它们分化成内皮细胞(ECs)和周细胞样细胞,分泌促血管生成生长因子和细胞外囊泡(EVs)的能力,使其成为包括组织缺血在内的细胞治疗和再生医学的理想选择.在最近的血管生成研究中,ASC通常与内皮细胞(EC)类型共培养,以便在特定培养条件下形成成熟的血管样网络。在这次审查中,我们介绍了ASCs和ECs之间的共培养系统和共移植研究。在共同文化中,细胞通过直接的细胞-细胞接触或通过旁分泌信号进行通信。大多数情况下,ASC在血管周围的小生境中发现,它们稳定血管结构并表达常见的周细胞表面蛋白。在共同文化中,ASCs通过促进血管形成来调节内皮细胞和诱导血管生成,部分是通过电动汽车的分泌。与单细胞类型移植相比,ASC和EC的体内共移植显示出改善的功能性血管形成。脂肪组织作为间充质干细胞和用于共移植的EC的细胞来源是体内治疗性血管生成和血液灌注的突出选择。
    Adipose-derived mesenchymal stem/stromal cells (ASCs) are an adult stem cell population able to self-renew and differentiate into numerous cell lineages. ASCs provide a promising future for therapeutic angiogenesis due to their ability to promote blood vessel formation. Specifically, their ability to differentiate into endothelial cells (ECs) and pericyte-like cells and to secrete angiogenesis-promoting growth factors and extracellular vesicles (EVs) makes them an ideal option in cell therapy and in regenerative medicine in conditions including tissue ischemia. In recent angiogenesis research, ASCs have often been co-cultured with an endothelial cell (EC) type in order to form mature vessel-like networks in specific culture conditions. In this review, we introduce co-culture systems and co-transplantation studies between ASCs and ECs. In co-cultures, the cells communicate via direct cell-cell contact or via paracrine signaling. Most often, ASCs are found in the perivascular niche lining the vessels, where they stabilize the vascular structures and express common pericyte surface proteins. In co-cultures, ASCs modulate endothelial cells and induce angiogenesis by promoting tube formation, partly via secretion of EVs. In vivo co-transplantation of ASCs and ECs showed improved formation of functional vessels over a single cell type transplantation. Adipose tissue as a cell source for both mesenchymal stem cells and ECs for co-transplantation serves as a prominent option for therapeutic angiogenesis and blood perfusion in vivo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    急性肺损伤是患者死亡的严重形式和主要原因,仍然需要有效的治疗。本研究表明,间充质干细胞(MSCs)和端粒细胞(TCs)的共移植改善了实验性肺组织炎症的严重程度,水肿,和伤害,其中TC增加了MSCs迁移到肺中以及MSCs增殖和运动的能力。分子机制,骨桥蛋白显性网络在MSCs和TCs中是活跃的,并可能在MSCs和TC之间的相互作用中发挥支持和营养作用,特别是通过脂多糖激活的TC。表皮生长因子及其受体在MSCs与TCs之间的相互作用可能在MSCs与TCs之间的通讯中起关键作用,负责MSCs的增殖和运动,尤其是炎症激活后。我们的研究提供了证据,表明TC在植入的MSCs中具有营养和支持作用,MSCs和TCs联合移植可作为治疗急性肺损伤的新选择。
    Acute lung injury is a serious form and major cause of patient death and still needs efficient therapies. The present study evidenced that co-transplantation of mesenchymal stem cells (MSCs) and telocytes (TCs) improved the severity of experimental lung tissue inflammation, edema, and injury, where TCs increased MSCs migration into the lung and the capacity of MSCs proliferation and movement. Of molecular mechanisms, Osteopontin-dominant networks were active in MSCs and TCs, and might play supportive and nutrimental roles in the interaction between MSCs and TCs, especially activated TCs by lipopolysaccharide. The interaction between epidermal growth factor and its receptor from MSCs and TCs could play critical roles in communications between MSCs and TCs, responsible for MSCs proliferation and movement, especially after inflammatory activation. Our studies provide the evidence that TCs possess nutrimental and supportive roles in implanted MSCs, and co-transplantation of MSCs and TCs can be a new alternative in the therapy of acute lung injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    据报道,在单臂试验中,使用间充质干细胞(MSC)输注的单倍体造血干细胞移植(HSCT)作为重度再生障碍性贫血(SAA)的治疗方案是有效的。然而,如果不将结果与一线的结果进行比较,就很难进行评估,匹配的兄弟HSCT。在这里,我们回顾性分析了91例获得性SAA患者.他们接受来自单倍体供体的HSCT结合MSC转移(HID组)。我们将这些患者与其他103名接受一线配对同胞HSCT(MSD组)的患者进行了比较,以评估相对治疗效果。与MSD组患者相比,HID组中II-IV级和III-IV级急性移植物抗宿主病(aGvHD)和慢性移植物抗宿主病(cGvHD)的发病率较高(p<0.05).然而,骨髓和血小板植入的发生率,移植失败,移植物功能差,和广泛的cGvHD在两组中具有可比性。中位随访时间为36.6个月,两组的3年总生存率相似(83.5%对79.1%)。单变量和多变量分析显示,从诊断到移植的时间间隔大于4个月,经历过移植失败,移植物功能差,或III-IV级aGvHD与不良结局显著相关.所有HID患者均接受MSC与造血干细胞的共移植。然而,输注的MSCs来自脐带(UC-MSC组;43例患者)或骨髓(BM-MSC组;48例患者),并在不同的医疗中心给药.我们首先比较了两组之间的结果,并发现BM-MSC组表现出III-IV级aGvHD和cGvHD的发生率较低(p<0.05)。这项研究表明,造血和MSC的共移植显着降低了移植物排斥的风险和发生率,并且即使在没有密切相关的组织相容性供体材料的情况下,也可以有效地改善SAA患者的总体生存率。
    The application of haploidentical hematopoietic stem cell transplantation (HSCT) with mesenchymal stem cell (MSC) infusion as a treatment regimen for severe aplastic anemia (SAA) has been reported to be efficacious in single-arm trials. However, it is difficult to assess without comparing the results with those from a first-line, matched-sibling HSCT. Herein, we retrospectively reviewed 91 patients with acquired SAA. They received HSCT from haploidentical donors combined with MSC transfer (HID group). We compared these patients with 103 others who received first-line matched-sibling HSCT (MSD group) to evaluate relative treatment efficacy. Compared with the patients in the MSD group, those in the HID group presented with higher incidences of grades II-IV and III-IV acute graft versus host disease (aGvHD) and chronic graft versus host disease (cGvHD) (p < 0.05). However, the incidence of myeloid and platelet engraftment, graft failure, poor graft function, and extensive cGvHD were comparable for both groups. The median follow-up was 36.6 months and the 3-year overall survival rate was similar for both groups (83.5% versus 79.1%). Univariate and multivariate analyses revealed that time intervals greater than 4 months from diagnosis to transplantation, experienced graft failure, poor graft function, or grade III-IV aGvHD were significantly associated with adverse outcomes. All HID patients received MSC co-transplantation with hematopoietic stem cells. However, the infused MSCs were derived from umbilical cord (UC-MSC group; 43 patients) or bone marrow (BM-MSC group; 48 patients) and were administered at different medical centers. We first compared the outcomes between the two groups and detected that the BM-MSC group exhibited lower incidences of grade III-IV aGvHD and cGvHD (p < 0.05). This study suggests that co-transplantation of hematopoietic and MSCs significantly reduces the risk and incidence of graft rejection and may effectively improve overall survival in patients with SAA even in the absence of closely related histocompatible donor material.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Radiation therapy is crucial in the therapeutic arsenal to cure cancers; however, non-neoplastic tissues around an abdominopelvic tumor can be damaged by ionizing radiation. In particular, the radio-induced death of highly proliferative stem/progenitor cells of the colonic mucosa could induce severe ulcers. The importance of sequelae for patients with gastrointestinal complications after radiotherapy and the absence of satisfactory management has opened the field to the testing of innovative treatments. The aim of this study was to use adult epithelial cells from the colon, to reduce colonic injuries in an animal model reproducing radiation damage observed in patients. We demonstrated that transplanted in vitro-amplified epithelial cells from colonic organoids (ECO) of C57/Bl6 mice expressing green fluorescent protein implant, proliferate, and differentiate in irradiated mucosa and reduce ulcer size. To improve the therapeutic benefit of ECO-based treatment with clinical translatability, we performed co-injection of ECO with mesenchymal stromal cells (MSCs), cells involved in niche function and widely used in clinical trials. We observed in vivo an improvement of the therapeutic benefit and in vitro analysis highlighted that co-culture of MSCs with ECO increases the number, proliferation, and size of colonic organoids. We also demonstrated, using gene expression analysis and siRNA inhibition, the involvement of bone morphogenetic protein antagonists in MSC-induced organoid formation. This study provides evidence of the potential of ECO to limit late radiation effects on the colon and opens perspectives on combined strategies to improve their amplification abilities and therapeutic effects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    We demonstrate a novel approach to reverse advanced stages of blindness using hydrogel-mediated delivery of retinal pigmented epithelium (RPE) and photoreceptors directly to the degenerated retina of blind mice. With sodium iodate (NaIO3) injections in mice, both RPE and photoreceptors degenerate, resulting in complete blindness and recapitulating the advanced retinal degeneration that is often observed in humans. We observed vision restoration only with co-transplantation of RPE and photoreceptors in a hyaluronic acid-based hydrogel, and not with transplantation of each cell type alone as determined with optokinetic head tracking and light avoidance assays. Both RPE and photoreceptors survived significantly better when co-transplanted than in their respective single cell type controls. While others have pursued transplantation of one of either RPE or photoreceptors, we demonstrate the importance of transplanting both cell types with a minimally-invasive hydrogel for vision repair in a degenerative disease model of the retina.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号