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
    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
    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
    胰腺祖细胞(PPCs)是所有胰腺细胞的主要来源,包括β细胞,因此,PPC的增殖和分化为胰岛样细胞簇(ICC)为糖尿病患者提供可移植的胰岛开辟了途径。同时,间充质干细胞(MSCs)可以增强不同类型感兴趣的细胞的发育和功能,但是它们在PPC上的作用仍然未知。我们旨在通过我们建立的人PPC与人胎儿骨髓来源的MSC的共培养系统,探索MSC诱导体外和体内PPC/ICC发育的作用机制。我们检查了MSC条件培养基对PPC增殖和存活的影响。同时,我们研究了MSC共培养在体外和体内共/移植中增强PPC/ICC功能的作用。此外,我们确定IGF1是MSC分别通过IGF1-PI3K/Akt和IGF1-MEK/ERK1/2影响PPC分化和增殖的关键因素。总之,我们的数据表明,MSCs通过IGF1信号刺激人PPCs的分化和增殖,更重要的是,促进了ICC的体内移植功能。一起来看,我们的方案可能为PPC增殖和分化为临床可移植胰岛提供机制驱动的基础.
    Pancreatic progenitor cells (PPCs) are the primary source for all pancreatic cells, including beta-cells, and thus the proliferation and differentiation of PPCs into islet-like cell clusters (ICCs) opens an avenue to providing transplantable islets for diabetic patients. Meanwhile, mesenchymal stem cells (MSCs) can enhance the development and function of different cell types of interest, but their role on PPCs remains unknown. We aimed to explore the mechanism-of-action whereby MSCs induce the in vitro and in vivo PPC/ICC development by means of our established co-culture system of human PPCs with human fetal bone marrow-derived MSCs. We examined the effect of MSC-conditioned medium on PPC proliferation and survival. Meanwhile, we studied the effect of MSC co-culture enhanced PPC/ICC function in vitro and in vivo co-/transplantation. Furthermore, we identified IGF1 as a critical factor responsible for the MSC effects on PPC differentiation and proliferation via IGF1-PI3K/Akt and IGF1-MEK/ERK1/2, respectively. In conclusion, our data indicate that MSCs stimulated the differentiation and proliferation of human PPCs via IGF1 signaling, and more importantly, promoted the in vivo engraftment function of ICCs. Taken together, our protocol may provide a mechanism-driven basis for the proliferation and differentiation of PPCs into clinically transplantable islets.
    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
    Cell-based therapy is an alternative strategy to improve outcomes of peripheral nerve injury (PNI). Epidermal neural crest stem cell (EPI-NCSC) is obtained from autologous tissue without immunological rejection, which could expand quickly in vitro and is suitable candidate for cell-based therapy. Olfactory ensheathing cell (OEC) could secrete multiple neurotrophic factors (NTFs), which is often used to repair PNI individually. However, whether the combination of EPI-NCSC and OEC have better effects on PNI repair remains unclear. Here we use EPI-NCSC and OEC co-transplantation in a rat sciatic nerve defect model to ascertain the effects and potential mechanisms of cells co-transplantation on PNI. The effect of EPI-NCSC and OEC co-transplantation on PNI is assessed by using a combination of immunohistochemistry (IHC), electrophysiological recording and neural function test. Co-transplantation of EPI-NCSC and OEC exerts a beneficial effect upon PNI such as better organized structure, nerve function recovery, and lower motoneuron apoptosis. IHC and enzyme-linked immuno sorbent assay (ELISA) further demonstrate that cells co-transplantation may improve PNI via the expression of brain derived growth factor (BDNF) and nerve growth factor (NGF) up-regulated by EPI-NCSC and OEC synergistically. Eventually, the results from this study reveal that EPI-NCSC and OEC co-transplantation effectively repairs PNI through enhancing the level of BDNF and NGF, indicating that cells co-transplantation may serve as a fruitful avenue for PNI in clinic treatment.
    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
    Hemophilia A (HA) is an X-linked recessive disorder caused by mutations in the Factor VIII (FVIII) gene leading to deficient blood coagulation. As a monogenic disorder, HA is an ideal target for cell-based gene therapy, but successful treatment has been hampered by insufficient engraftment of potential therapeutic cells.
    In this study, we sought to determine whether co-transplantation of endothelial colony-forming cells (ECFCs) and placenta-derived mesenchymal stromal cells (PMSCs) can achieve long-term engraftment and FVIII expression. ECFCs and PMSCs were transduced with a B domain deleted factor VIII (BDD-FVIII) expressing lentiviral vector and luciferase, green fluorescent protein or Td-Tomato containing lentiviral tracking vectors. They were transplanted intramuscularly into neonatal or adult immunodeficient mice.
    In vivo bioluminescence imaging showed that the ECFC only and the co-transplantation groups but not the PMSCs only group achieved long-term engraftment for at least 26 weeks, and the co-transplantation group showed a higher engraftment than the ECFC only group at 16 and 20 weeks post-transplantation. In addition, cell transplantation at the neonatal age achieved higher engraftment than at the adult age. Immunohistochemical analyses further showed that the engrafted ECFCs expressed FVIII, maintained endothelial phenotype, and generated functional vasculature. Next, co-transplantation of ECFCs and PMSCs into F8 knock-out HA mice reduced the blood loss volume from 562.13 ± 19.84 μl to 155.78 ± 44.93 μl in a tail-clip assay.
    This work demonstrated that co-transplantation of ECFCs with PMSCs at the neonatal age is a potential strategy to achieve stable, long-term engraftment, and thus holds great promise for cell-based treatment of HA.
    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
    内皮细胞(ECs)和间充质干细胞(MSCs)的共移植是修复复杂和大型骨缺损的重要策略。然而,ECs和MSCs相互作用的方式还有待完全阐明.我们发现从hBMSCs到hUVECs的前向ephrinB2/Ephs信号通过激活PI3K/AKT/mTOR通路促进hUVECs的管形成。从hUVECs到hBMSCs的反向ephrinB2/Ephs信号通过上调OCT4、SOX2和YAP1促进hBMSCs自我更新的增殖和维持。ECs和MSCs在裸鼠皮下共移植证实,正向ephrinB2/Ephs信号可以增加移植区血管的横截面积,逆转ephrinB2/Ephs信号传导可以维持体内移植的hBMSCs的自我更新。基于这些结果,EphrinB2/Ephs在ECs和MSCs之间的双向并分泌调节通过促进血管生成和获得足够数量的MSCs在改善骨缺损愈合中起关键作用。
    Co-transplantation of endothelial cells (ECs) and mesenchymal stem cells (MSCs) is an important strategy for repairing complex and large bone defects. However, the ways in which ECs and MSCs interact remain to be fully clarified. We found that forward ephrinB2/Ephs signaling from hBMSCs to hUVECs promoted the tube formation of hUVECs by activating the PI3K/AKT/mTOR pathway. Reverse ephrinB2/Ephs signaling from hUVECs to hBMSCs promoted the proliferation and maintenance of hBMSCs self-renewal via upregulation of OCT4, SOX2, and YAP1. Subcutaneous co-transplantation of ECs and MSCs in nude mice confirmed that forward ephrinB2/Ephs signaling could increase the cross-sectional area of blood vessels in the transplanted area, and reverse ephrinB2/Ephs signaling could maintain the self-renewal of transplanted hBMSCs in vivo. Based on these results, ephrinB2/Ephs bidirectional juxtacrine regulation between ECs and MSCs plays a pivotal role in improving the healing of bone defects by promoting angiogenesis and achieving a sufficient number of MSCs.
    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
    Spinal cord injury (SCI) is a common and serious disease which often induces catastrophic consequence in patients. Part of them exhibit neuropathic pain which presents unique challenges to clinicians, and there is no effective approach for the treatment up to now. Neural stem cells (NSCs) transplantation, as a promising and an effective method, could be considered for the treatment of SCI, whereas a main problem is the low survival of NSCs in traumatic milieu in host spinal cords, and the effect of NSCs on sensory function remains elusive. In this study, we investigated the effect and underlying molecular mechanism of co-transplantation of NSCs with olfactory ensheathing cells (OECs) on sensory functional improvement. In the measurement of thermal and mechanical stimuli, NSCs grafts recovered sensory function in SCI rats, while OECs led to hyperalgesia, indicated by the tail flick latency (TFL) and paw withdraw latency (PWL) (p<0.05). Co-transplantation could promote NSCs survival, and reverses the hyperalgesia triggered by OECs. This was corresponding to a significant improvement in sensory function. Moreover, NGF expression was substantial downregulated in the spinal cord of co-transplantation rats. The present findings suggested that co-transplantation of NSCs with OECs could improve sensory function and the possible mechanism is involved in NGF downregulation in rats with SCI. This may give some new indications for the treatment of SCI in future clinic cell therapy trial.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号