anterior chamber of the eye

眼前房
  • 文章类型: Journal Article
    将生物细胞与人造传感器杂交能够以高特异性检测宽范围的弱生理反应。眼前房(ACE)由于其眼免疫特权和光学透明性而成为理想的移植部位。这使得能够对细胞和微组织进行优越的非侵入性纵向分析。ACE中生物混合微结构的参与可能,然而,受瞳孔反应和动力学的影响。这里,生物混合微结构的无缝线移植,IP-Visio光敏树脂3D打印,提出了含有精确定位的胰岛的小鼠ACE。生物混合微结构允许机械固定在ACE中,与虹膜动力学无关。移植后,尽管与血管源(虹膜)物理分离并浸入具有连续循环和清除的低粘度液体(房水)中,但微结构中的胰岛仍成功维持其功能超过20周,并血管化。这种方法为ACE中的生物混合微组织移植开辟了新的前景,推进微组织-宿主相互作用的监测,疾病建模,治疗结果,和工程组织中的血管化。本文受版权保护。保留所有权利。
    Hybridizing biological cells with man-made sensors enable the detection of a wide range of weak physiological responses with high specificity. The anterior chamber of the eye (ACE) is an ideal transplantation site due to its ocular immune privilege and optical transparency, which enable superior noninvasive longitudinal analyses of cells and microtissues. Engraftment of biohybrid microstructures in the ACE may, however, be affected by the pupillary response and dynamics. Here, sutureless transplantation of biohybrid microstructures, 3D printed in IP-Visio photoresin, containing a precisely localized pancreatic islet to the ACE of mice is presented. The biohybrid microstructures allow mechanical fixation in the ACE, independent of iris dynamics. After transplantation, islets in the microstructures successfully sustain their functionality for over 20 weeks and become vascularized despite physical separation from the vessel source (iris) and immersion in a low-viscous liquid (aqueous humor) with continuous circulation and clearance. This approach opens new perspectives in biohybrid microtissue transplantation in the ACE, advancing monitoring of microtissue-host interactions, disease modeling, treatment outcomes, and vascularization in engineered tissues.
    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
    已提出通过重新激活其复制来扩大产生胰岛素的β细胞的数量作为预防或延迟糖尿病出现的疗法。使用抗体阵列,我们确定CCN4/Wisp1是一种富含断奶前小鼠的循环因子,β细胞数量急剧增加的时期。这一发现使我们研究了CCN4在β细胞增殖中的参与。我们证明了CCN4在体外培养的分离胰岛中促进成年β细胞增殖,并在体内移植到眼睛前房的胰岛中。在这一章中,我们介绍了在这两种情况下用于研究增殖的方法.
    Expanding the number of insulin-producing beta cells through reactivation of their replication has been proposed as a therapy to prevent or delay the appearance of diabetes. Using antibody arrays, we identified CCN4/Wisp1 as a circulating factor enriched in preweaning mice, a period in which beta cells exhibit a dramatic increase in number. This finding led us to investigate the involvement of CCN4 in beta cell proliferation. We demonstrated that CCN4 promotes adult beta cell proliferation in vitro in cultured isolated islets, and in vivo in islets transplanted into the anterior chamber of the eye. In this chapter, we present the methodology that was used to study proliferation in both settings.
    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
    在小鼠和灵长类动物模型中,胰岛移植到眼睛前房(ACE)中已显示出改善糖尿病的血糖控制和代谢参数。这种新的移植部位还允许递送治疗剂,比如免疫抑制药物,局部防止胰岛移植物排斥反应和避免不必要的全身副作用。在同种异体胰岛移植到非人灵长类动物的ACE中之前,进行微粉化地塞米松植入物的局部玻璃体内给药。观察到两个研究组,即无免疫抑制的同种异体移植物(n=4眼)和具有局部免疫抑制的同种异体移植物(n=8眼)。在24周内平行评估胰岛移植物的存活率和ACE中的地塞米松浓度。接受局部地塞米松治疗的同种异体胰岛移植物的生存率明显优于无免疫抑制者(中位生存期为15周vs3周,对数秩检验p<0.0001)。大约73%的移植物在第10周用单一的局部地塞米松植入物仍然存活。对照组没有移植物存活。地塞米松处理的胰岛移植物显示出对高葡萄糖刺激的良好功能反应,尽管在第8周至第12周存在胰岛素分泌的短暂抑制。我们的发现表明,在局部地塞米松治疗的ACE中,同种异体移植物的存活率显着提高。这些结果强调了在ACE中局部施用药理化合物以改善胰岛移植物存活和功能的可行性。通过消除全身免疫抑制的需要,这些发现可能会影响临床胰岛移植治疗糖尿病,ACE可以作为一种新型的治疗性胰岛移植部位,具有很高的局部药物干预潜力。
    Pancreatic islet transplantation into the anterior chamber of the eye (ACE) has been shown to improve glycemic control and metabolic parameters of diabetes in both murine and primate models. This novel transplantation site also allows the delivery of therapeutic agents, such as immunosuppressive drugs, locally to prevent islet graft rejection and circumvent unwanted systemic side effects. Local intravitreal administration of micronized dexamethasone implant was performed prior to allogeneic islet transplantation into the ACEs of non-human primates. Two study groups were observed namely allogeneic graft without immunosuppression (n = 4 eyes) and allogeneic graft with local immunosuppression (n = 8 eyes). Survival of islet grafts and dexamethasone concentration in the ACE were assessed in parallel for 24 weeks. Allogeneic islet grafts with local dexamethasone treatment showed significantly better survival than those with no immunosuppression (median survival time- 15 weeks vs 3 weeks, log-rank test p<0.0001). Around 73% of the grafts still survived at week 10 with a single local dexamethasone implant, where the control group showed no graft survival. Dexamethasone treated islet grafts revealed a good functional response to high glucose stimulation despite there was a transient suppression of insulin secretion from week 8 to 12. Our findings show a significant improvement of allografts survival in the ACE with local dexamethasone treatment. These results highlight the feasibility of local administration of pharmacological compounds in the ACE to improve islet graft survival and function. By eliminating the need for systemic immunosuppression, these findings may impact clinical islet transplantation in the treatment of diabetes, and the ACE may serve as a novel therapeutic islet transplantation site with high potential for local pharmacological intervention.
    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
    在这篇评论中,我们回应了最近一篇题为“干细胞衍生胰岛替代疗法的承诺”的评论的结论,其中强调了从干细胞产生葡萄糖反应性“胰岛”的最新进展,以及它们在1型糖尿病(T1D)的胰岛移植治疗中的应用。该综述还概述了临床胰岛移植的现状以及阻碍其实现全部治疗前景的挑战。我们同意该评论的结论,并建议通过使用眼前房作为胰岛移植部位可以克服已确定的挑战。我们预计,干细胞衍生的胰岛和眼内移植的结合可以帮助这种有前途的T1D治疗取得圆满成功。
    In this Commentary, we echo the conclusions of a recent review titled \"The promise of stem cell-derived islet replacement therapy,\" which highlighted recent advances in producing glucose responsive \"islets\" from stem cells and the benefits of their use in islet transplant therapy in type 1 diabetes (T1D). The review also outlined the status of clinical islet transplantation and the challenges that have prevented it from reaching its full therapeutic promise. We agree with the conclusions of the review and suggest that the identified challenges may be overcome by using the eye anterior chamber as an islet transplant site. We anticipate that the combination of stem cell-derived islets and intraocular transplant could help this promising T1D therapy reach full fruition.
    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
    An understanding of the immune mechanisms that lead to rejection versus tolerance of allogeneic pancreatic islet grafts is of paramount importance, as it facilitates the development of innovative methods to improve the transplant outcome. Here, we used our established intraocular islet transplant model to gain novel insight into changes in the local metabolome and proteome within the islet allograft\'s immediate microenvironment in association with immune-mediated rejection or tolerance. We performed integrated metabolomics and proteomics analyses in aqueous humor samples representative of the graft\'s microenvironment under each transplant outcome. The results showed that several free amino acids, small primary amines, and soluble proteins related to the Warburg effect were upregulated or downregulated in association with either outcome. In general, the observed shifts in the local metabolite and protein profiles in association with rejection were consistent with established pro-inflammatory metabolic pathways and those observed in association with tolerance were immune regulatory. Taken together, the current findings further support the potential of metabolic reprogramming of immune cells towards immune regulation through targeted pharmacological and dietary interventions against specific metabolic pathways that promote the Warburg effect to prevent the rejection of transplanted islets and promote their immune tolerance.
    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
    限制在胰岛中的内分泌细胞负责维持血糖稳态。特别是,β细胞产生和分泌胰岛素,一种调节葡萄糖摄取和代谢的必需激素。β细胞数量不足或导致葡萄糖诱导的胰岛素分泌的分子机制缺陷会引发糖尿病的发展,一种流行病在全世界蔓延的严重疾病。因此,全面了解调节β细胞质量和功能的各种适应性程序对于理解糖尿病的发病机理和开发有效的治疗策略至关重要。虽然通过使用从胰腺分离的胰岛获得了重要的发现,体外研究本身是有限的,因为它们缺乏影响体内胰岛细胞功能的许多因素,并且不允许对活体中胰岛细胞的可塑性进行纵向监测。在这方面,多年来已经开发了许多成像方法来研究胰腺中的胰岛。由于胰岛的尺寸和位置相对较小,这是一项具有挑战性的任务,散布在整个器官中。为了提高成像分辨率并允许在单个胰岛中进行纵向研究,另一种策略是将胰岛移植到其他更容易成像的部位.在这篇综述中,我们介绍了眼前房作为移植和成像部位的胰岛细胞可塑性研究,并总结了该技术平台推动的主要研究成果。
    The endocrine cells confined in the islets of Langerhans are responsible for the maintenance of blood glucose homeostasis. In particular, beta cells produce and secrete insulin, an essential hormone regulating glucose uptake and metabolism. An insufficient amount of beta cells or defects in the molecular mechanisms leading to glucose-induced insulin secretion trigger the development of diabetes, a severe disease with epidemic spreading throughout the world. A comprehensive appreciation of the diverse adaptive procedures regulating beta cell mass and function is thus of paramount importance for the understanding of diabetes pathogenesis and for the development of effective therapeutic strategies. While significant findings were obtained by the use of islets isolated from the pancreas, in vitro studies are inherently limited since they lack the many factors influencing pancreatic islet cell function in vivo and do not allow for longitudinal monitoring of islet cell plasticity in the living organism. In this respect a number of imaging methodologies have been developed over the years for the study of islets in situ in the pancreas, a challenging task due to the relatively small size of the islets and their location, scattered throughout the organ. To increase imaging resolution and allow for longitudinal studies in individual islets, another strategy is based on the transplantation of islets into other sites that are more accessible for imaging. In this review we present the anterior chamber of the eye as a transplantation and imaging site for the study of pancreatic islet cell plasticity, and summarize the major research outcomes facilitated by this technological platform.
    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
    The review presents the main pathogenetic mechanisms of cataract development in an avitic eye as well as anatomical and functional changes of the eye in the state of avitria. The authors have also analyzed distinctive behavior of such eyes during phacoemulsification and surgical techniques used to answer specific avitreal phenomena.
    В обзоре представлены основные патогенетические механизмы развития катаракты в авитреальном глазу и анатомо-функциональные изменения глаза в состоянии авитрии. Проанализированы и обобщены особенности поведения авитреального глаза при факоэмульсификации катаракты и оперативно технические приемы, используемые при возникновении специфических авитреальных феноменов.
    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
    Cytotoxic T lymphocytes (CTL) are an essential part of our immune system by killing infected and malignant cells. To fully understand this process, it is necessary to study CTL function in the physiological setting of a living organism to account for their interplay with other immune cells like CD4+ T helper cells and macrophages. The anterior chamber of the eye (ACE), originally developed for diabetes research, is ideally suited for non-invasive and longitudinal in vivo imaging. We take advantage of the ACE window to observe immune responses, particularly allorejection of islets of Langerhans cells by CTLs. We follow the onset of the rejection after vascularization on islets until the end of the rejection process for about a month by repetitive two-photon microscopy. We find that CTLs show reduced migration on allogeneic islets in vivo compared to in vitro data, indicating CTL activation. Interestingly, the temporal infiltration pattern of T cells during rejection is precisely regulated, showing enrichment of CD4+ T helper cells on the islets before arrival of CD8+ CTLs. The adaptation of the ACE to immune responses enables the examination of the mechanism and regulation of CTL-mediated killing in vivo and to further investigate the killing in gene-deficient mice that resemble severe human immune diseases.
    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
    The applicability and benefits of pancreatic islet transplantation are limited due to various issues including the need to avoid immune-mediated rejection. Here, we used our experimental platform of allogeneic islet transplant in the anterior chamber of the eye (ACE-platform) to longitudinally monitor the progress of rejection in mice and obtain aqueous humor samples representative of the microenvironment of the graft for accurately-timed proteomic analyses. LC-MS/MS-based proteomics performed on such mass-limited samples (~5 μL) identified a total of 1296 proteins. Various analyses revealed distinct protein patterns associated with the mounting of the inflammatory and immune responses and their evolution with the progression of the rejection. Pathway analyses indicated predominant changes in cytotoxic functions, cell movement, and innate and adaptive immune responses. Network prediction analyses revealed transition from humoral to cellular immune response and exacerbation of pro-inflammatory signaling. One of the proteins identified by this localized proteomics as a candidate biomarker of islet rejection, Cystatin 3, was further validated by ELISA in the aqueous humor. This study provides (1) experimental evidence demonstrating the feasibility of longitudinal localized proteomics using small aqueous humor samples and (2) proof-of-concept for the discovery of biomarkers of impending immune attack from the immediate local microenvironment of ACE-transplanted islets. SIGNIFICANCE: The combination of the ACE-platform and longitudinal localized proteomics offers a powerful approach to biomarker discovery during the various stages of immune reactions mounted against transplanted tissues including pancreatic islets. It also supports proteomics-assisted drug discovery and development efforts aimed at preventing rejection through efficacy assessment of new agents by noninvasive and longitudinal graft monitoring.
    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
    Replacement of the insulin-secreting beta cells through transplantation of pancreatic islets to the liver is a promising treatment for type-1 diabetes. However, low oxygen tension, shear stress, and the induction of inflammation lead to significant islet dysfunction and loss. The anterior chamber of the eye (ACE) has gained considerable interest and represents an alternative therapeutic islet transplantation site because of its accessibility, high oxygen tension, and immune-privileged milieu. We have previously demonstrated the feasibility of intraocular islet transplant in mouse and nonhuman primate models of type-1 diabetes and are now assessing its efficacy on glucose homeostasis in a nonhuman primate model of type-2 diabetes. We transplanted allogeneic donor islets (1,500 islet equivalents/kg) into the anterior chamber of one eye in a cynomolgus monkey with high-fat-diet-induced type-2 diabetes. Repeated examinations of the anterior and posterior segments of both eyes were done to monitor the engrafted islets and assess the overall ocular health. Fasting blood glucose level, blood biochemistry, and other metabolic parameters were routinely evaluated to determine the function of the islet graft and diabetes status. The transplanted islets were rapidly engrafted onto the iris and became vascularized 1 month after transplantation. We did not detect changes in intraocular pressure, cataract formation, ophthalmitis, or retinal vessel deformation. A significant lower fasting blood glucose level was observed while the graft was in place, and the transplantation reverts the progression of diabetes. The metabolic markers, hemoglobin A1C and fructosamine, demonstrated improvement following islet transplantation. As a conclusion, intraocular islet transplantation in one eye of a cynomolgus monkey with type-2 diabetes improved its overall plasma glucose homeostasis, as evidenced by short-term measures and long-term metabolic markers. These results further support the future application of the ACE as an alternative site for clinical islet transplants in the context of type-2 diabetes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号