cavernous nerve injury

海绵状神经损伤
  • 文章类型: Journal Article
    背景:海绵状神经(CN)损伤,由前列腺切除术和糖尿病引起的,在患者和动物模型的阴茎海绵体中启动重塑过程(平滑肌细胞凋亡和胶原蛋白增加),这是勃起功能障碍(ED)的根本原因,Sonichedgehog(SHH)通路在阴茎对神经支配的反应中起着至关重要的作用,胶原蛋白随着SHH抑制而增加,随着SHH处理而减少。
    目的:我们检查了SHH如何防止阴茎重塑和CN损伤引起的胶原蛋白增加的部分机制是否涉及骨形态发生蛋白4(BMP4)和gremlin1(GREM1),并检查了SHH之间的关系。ED患者和CN损伤大鼠模型阴茎中的BMP4、GREM1和胶原,SHH抑制,SHH,BMP4和GREM1治疗。
    方法:佩罗尼病的海绵体(对照),前列腺切除术,获得糖尿病性ED患者(N=30)。成年SpragueDawley大鼠(n=90)接受(1)CN挤压(1-7天)或假手术;(2)CN损伤和BMP4,GREM1或小鼠血清白蛋白(对照)通过Affi-Gel珠或肽两亲物(PA)治疗14天;(3)5E1SHH抑制剂,IgG,或磷酸盐缓冲盐水(对照)处理2至4天;或(4)用小鼠血清白蛋白或SHH压碎CN9天。
    结果:进行了BMP4和GREM1的免疫组织化学和Western分析,以及通过羟脯氨酸和三色染色进行的胶原蛋白分析。
    结果:在前列腺切除术的海绵体平滑肌中发现了BMP4和GREM1蛋白,糖尿病,和佩罗尼的病人,在大鼠平滑肌中,交感神经纤维,神经束,血管,还有尿道.在CN损伤和BMP4治疗的大鼠中,胶原蛋白减少了25.4%(P=.02),在CN损伤和GREM1治疗的大鼠中,胶原蛋白增加了61.3%(P=.005)。三色染色显示用GREM1处理的大鼠中胶原蛋白增加。西方分析发现,前列腺切除术和糖尿病患者的海绵体中BMP4和GREM1增加,和CN损伤后(1-2天)在我们的大鼠模型。BMP4和GREM1的定位随SHH抑制而改变。SHH处理增加了BMP4和GREM1的单体形式,改变了它们的信号传导范围。
    结论:更好地了解阴茎重塑以及神经支配丧失是如何发生纤维化的,对于开发新的ED疗法至关重要。
    SHH,BMP4,GREM1和胶原蛋白在阴茎中是复杂的。
    结论:BMP4和GREM1是影响胶原蛋白的SHH下游靶标,可能与SHH合作预防阴茎重塑和ED。
    BACKGROUND: Cavernous nerve (CN) injury, caused by prostatectomy and diabetes, initiates a remodeling process (smooth muscle apoptosis and increased collagen) in the corpora cavernosa of the penis of patients and animal models that is an underlying cause of erectile dysfunction (ED), and the Sonic hedgehog (SHH) pathway plays an essential role in the response of the penis to denervation, as collagen increases with SHH inhibition and decreases with SHH treatment.
    OBJECTIVE: We examined if part of the mechanism of how SHH prevents penile remodeling and increased collagen with CN injury involves bone morphogenetic protein 4 (BMP4) and gremlin1 (GREM1) and examined the relationship between SHH, BMP4, GREM1, and collagen in penis of ED patients and rat models of CN injury, SHH inhibition, and SHH, BMP4, and GREM1 treatment.
    METHODS: Corpora cavernosa of Peyronie\'s disease (control), prostatectomy, and diabetic ED patients were obtained (N = 30). Adult Sprague Dawley rats (n = 90) underwent (1) CN crush (1-7 days) or sham surgery; (2) CN injury and BMP4, GREM1, or mouse serum albumin (control) treatment via Affi-Gel beads or peptide amphiphile (PA) for 14 days; (3) 5E1 SHH inhibitor, IgG, or phosphate-buffered saline (control) treatment for 2 to 4 days; or (4) CN crush with mouse serum albumin or SHH for 9 days.
    RESULTS: Immunohistochemical and Western analysis for BMP4 and GREM1, and collagen analysis by hydroxyproline and trichrome stain were performed.
    RESULTS: BMP4 and GREM1 proteins were identified in corpora cavernosa smooth muscle of prostatectomy, diabetic, and Peyronie\'s patients, and in rat smooth muscle, sympathetic nerve fibers, perineurium, blood vessels, and urethra. Collagen decreased 25.4% in rats with CN injury and BMP4 treatment (P = .02) and increased 61.3% with CN injury and GREM1 treatment (P = .005). Trichrome stain showed increased collagen in rats treated with GREM1. Western analysis identified increased BMP4 and GREM1 in corpora cavernosa of prostatectomy and diabetic patients, and after CN injury (1-2 days) in our rat model. Localization of BMP4 and GREM1 changed with SHH inhibition. SHH treatment increased the monomer form of BMP4 and GREM1, altering their range of signaling.
    CONCLUSIONS: A better understanding of penile remodeling and how fibrosis occurs with loss of innervation is essential for development of novel ED therapies.
    UNASSIGNED: The relationship between SHH, BMP4, GREM1, and collagen is complex in the penis.
    CONCLUSIONS: BMP4 and GREM1 are downstream targets of SHH that impact collagen and may be useful in collaboration with SHH to prevent penile remodeling and ED.
    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
    目的:探讨富马酸二甲酯(DMF)改善双侧海绵体神经损伤(BCNI)大鼠勃起功能的治疗作用。同时阐明其潜在机制。
    方法:采用双侧海绵体神经(CN)夹闭方法建立BCNI大鼠模型。通过以低(20mg/kg/天)和高(40mg/kg/天)剂量管饲法给予DMF,持续4周。通过CN的电刺激评估勃起功能。收集阴茎和CN组织用于后续分析。此外,PC-12细胞系用于验证DMF的体外作用机制。通过慢病毒转染构建Nfe2l2或Ho-1基因敲低的PC-12细胞系,分别。使用H2O2诱导受损的细胞模型。然后采用分子生物学方法分析细胞分子和蛋白质。
    结果:给予DMF4周导致勃起功能改善,BCNI大鼠阴茎海绵体纤维化减少。CN形态得到改善,神经纤维数量增加。此外,nNOS的水平,NO,cGMP增加了,而阴茎海绵体中Ca2+减少。值得注意的是,ROS的水平,3-NT和NLRP3炎性体产生减少,Nrf2和HO-1蛋白在阴茎背神经(DPN)和CN中的表达增加。体外,DMF增加细胞活力,降低ROS水平,促进的SOD,减少3-NT,MDA和DNA损伤标记,并抑制H2O2诱导的PC-12细胞中NLRP3炎性体的活化。NFe2l2敲低和Ho-1敲低可显著减弱DMF的保护作用,分别。此外,N-乙酰半胱氨酸抑制ROS产生导致H2O2诱导的PC-12细胞中NLRP3炎性体活化减少。
    结论:DMF通过激活Nrf2/HO-1通路,抑制氧化应激和NLRP3炎性体介导的焦亡,保护神经,从而改善BCNI大鼠的勃起功能。
    OBJECTIVE: To investigate the therapeutic potential of dimethyl fumarate (DMF) in improving erectile function of bilateral cavernous nerve injury (BCNI) rats, along with elucidating its underlying mechanisms.
    METHODS: A BCNI rat model was established by clamping bilateral cavernous nerve (CN). DMF was given by gavage at low (20 mg/kg/day) and high (40 mg/kg/day) dosages for a duration of 4 weeks. Erectile function was assessed by electrical stimulation of CN. Penis and CN tissues were collected for subsequent analysis. Additionally, PC-12 cell line was used to verify the mechanism of DMF in vitro. Nfe2l2 or Ho-1 gene knockdown PC-12 cell lines were constructed by lentiviral transfection, respectively. A damaged cell model was induced using H2O2. And then molecular biological methods were employed to analyze cellular molecules and proteins.
    RESULTS: DMF administration for 4 weeks led to improvements in erectile function, reduced fibrosis of penis corpus cavernosum in BCNI rats. The morphology of CN was improved and the number of nerve fibers increased. Furthermore, the levels of nNOS, NO, and cGMP were increased, while Ca2+ was decreased in penis corpus cavernosum. Notably, the levels of ROS, 3-NT and NLRP3 inflammasomes production were reduced, alongside increased expression of Nrf2 and HO-1 proteins in the dorsal penile nerve (DPN) and CN. In vitro, DMF increased cell viability, reduced ROS level, promoted SOD, diminished 3-NT, MDA and DNA damage markers, and inhibited the activation of NLRP3 inflammasomes in H2O2 induced PC-12 cells. Nfe2l2 knockdown and Ho-1 knockdown significantly attenuated the protective effect of DMF, respectively. Furthermore, inhibition of ROS production by N-acetylcysteine led to a reduction in NLRP3 inflammasome activation in H2O2 induced PC-12 cells.
    CONCLUSIONS: DMF improved erectile function of BCNI rats by protecting nerves through inhibiting oxidative stress and the activation of NLRP3 inflammasome-mediated pyroptosis via activation of Nrf2/HO-1 pathway.
    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
    在根治性前列腺切除术和膀胱切除术后,勃起功能障碍(ED)发生在越来越多的患者中,阴茎海绵体平滑肌细胞的表型调节与ED密切相关。
    确定内质网应激(ERS)是否与双侧海绵状神经损伤(BCNI)引起的ED的表型调节有关。
    总共,36只Sprague-Dawley大鼠随机分为3组:假,其中大鼠接受假手术,双侧海绵体神经暴露加磷酸盐缓冲盐水;对照组,其中大鼠接受BCNI加磷酸盐缓冲盐水;和实验,其中大鼠接受BCNI加4-苯基丁酸。方差分析和Bonferroni多重比较检验用于评估组间差异。
    勃起功能,平滑肌/胶原蛋白比例,测量表型调节和ERS的表达水平。
    与假手术组相比,对照组的两个比率-最大海绵体内压/平均动脉压和平滑肌/胶原-降低。在阴茎组织中,GRP78(78kDa葡萄糖调节蛋白)的表达增加,p-PERK/PERK(磷酸化蛋白激酶R样内质网激酶/蛋白激酶R样内质网激酶),caspase3,CHOP(C/EBP同源蛋白),和OPN(骨桥蛋白),但nNOS(神经元一氧化氮合酶)和α-SMA(α-平滑肌肌动蛋白)的表达降低。与对照组相比,实验组勃起功能得到改善,病理变化部分恢复。
    本研究表明ERS与海绵状神经损伤引起的ED有关,从而为临床治疗提供新的靶点和理论依据。
    本研究首次证明ERS与海绵状神经损伤引起的ED有关。ERS的抑制逆转了BCNI大鼠的表型调节并改善了勃起功能。应进行其他体外研究以验证这些结论并探索表型调节的具体机制。
    本研究表明,抑制ERS可以逆转BCNI大鼠的表型调节并增强勃起功能。
    UNASSIGNED: Erectile dysfunction (ED) occurs in an increasing number of patients after radical prostatectomy and cystectomy, and the phenotypic modulation of corpus cavernosum smooth muscle cells is closely related to ED.
    UNASSIGNED: To determine whether endoplasmic reticulum stress (ERS) is implicated in the phenotypic modulation of ED induced by bilateral cavernous nerve injury (BCNI).
    UNASSIGNED: In total, 36 Sprague-Dawley rats were randomly divided into 3 groups: sham, in which rats received sham surgery with bilateral cavernous nerve exposure plus phosphate-buffered saline; control, in which rats received BCNI plus phosphate-buffered saline; and experimental, in which rats received BCNI plus 4-phenylbutyric acid. Analysis of variance and a Bonferroni multiple-comparison test were utilized to evaluate differences among groups.
    UNASSIGNED: Erectile function, smooth muscle/collagen ratios, and the expression levels of phenotypic modulation and ERS were measured.
    UNASSIGNED: Two ratios-maximum intracavernosal pressure/mean arterial pressure and smooth muscle/collagen-were decreased in the control group as compared with the sham group. In penile tissue, there was increased expression of GRP78 (78-kDa glucose-regulated protein), p-PERK/PERK (phosphorylated protein kinase R-like endoplasmic reticulum kinase/protein kinase R-like endoplasmic reticulum kinase), caspase 3, CHOP (C/EBP homologous protein), and OPN (osteopontin) but decreased expression of nNOS (neuronal nitric oxide synthase) and α-SMA (α-smooth muscle actin). As compared with the control group, erectile function was improved and pathologic changes were partially recovered in the experimental group.
    UNASSIGNED: The present study demonstrated that ERS is involved in ED caused by cavernous nerve injury, thereby providing a new target and theoretical basis for clinical treatment.
    UNASSIGNED: The present study demonstrated for the first time that ERS is related to ED caused by cavernous nerve injury. Inhibition of ERS reverses phenotypic modulation and improves erectile function in rats with BCNI. Additional in vitro studies should be performed to verify these conclusions and explore the specific mechanism of phenotypic modulation.
    UNASSIGNED: The present study demonstrated that inhibiting ERS reverses phenotypic modulation and enhances erectile function in rats with BCNI.
    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
    作为一种周围神经损伤疾病,由前列腺癌手术和其他盆腔手术引起的海绵状神经损伤(CNI)会对海绵状血管和神经造成器质性损害,从而显著减弱对磷酸二酯酶-5抑制剂的反应。这里,我们使用双侧CNI小鼠模型研究血红素结合蛋白1(Hebp1)在勃起功能中的作用,已知它能促进血管生成和改善糖尿病小鼠的勃起。我们发现Hebp1在CNI小鼠中具有强大的神经血管再生作用,证明外源递送的Hebp1通过促进海绵状内皮壁细胞和神经元的存活来改善勃起功能。我们进一步发现,由小鼠海绵状周细胞(MCP)衍生的细胞外囊泡递送的内源性Hebp1促进CNI小鼠的神经血管再生。此外,Hebp1通过调节claudin家族蛋白降低血管通透性来实现这些作用。我们的发现为Hebp1作为神经血管再生因子提供了新的见解,并证明了其在各种周围神经损伤中的潜在治疗应用。
    As a peripheral nerve injury disease, cavernous nerve injury (CNI) caused by prostate cancer surgery and other pelvic surgery causes organic damage to cavernous blood vessels and nerves, thereby significantly attenuating the response to phosphodiesterase-5 inhibitors. Here, we investigated the role of heme-binding protein 1 (Hebp1) in erectile function using a mouse model of bilateral CNI, which is known to promote angiogenesis and improve erection in diabetic mice. We found a potent neurovascular regenerative effect of Hebp1 in CNI mice, demonstrating that exogenously delivered Hebp1 improved erectile function by promoting the survival of cavernous endothelial-mural cells and neurons. We further found that endogenous Hebp1 delivered by mouse cavernous pericyte (MCP)-derived extracellular vesicles promoted neurovascular regeneration in CNI mice. Moreover, Hebp1 achieved these effects by reducing vascular permeability through regulation of claudin family proteins. Our findings provide new insights into Hebp1 as a neurovascular regeneration factor and demonstrate its potential therapeutic application to various peripheral nerve injuries.
    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
    勃起功能障碍(ED),作为一种常见的男性疾病,会严重降低男性及其伴侣的生活质量。随着人类生活水平的提高,ED被认为是困扰男性的重要健康问题。然而,现有的治疗方法很难满足所有患者的需求,因此有必要开发新的治疗策略。外泌体,作为一类由具有双层膜结构的细胞分泌的囊泡,参与人体的各种生理和病理过程,被认为具有巨大的治疗潜力。本文综述了外泌体治疗ED动物模型的最新进展。并对未来的研究进行了展望,以期为临床试验和临床翻译提供依据。
    Erectile dysfunction (ED), as a common male disease, can seriously reduce the life quality of men and their partners. With the improvement of human living standards, ED is considered to be an important health issue that plagues men. However, it is difficult for existing therapeutic approaches to meet the needs of all patients, so it is necessary to develop novel treatment strategies. Exosomes, as a class of vesicles secreted by cells with bilayer membrane structure, are involved in various physiological and pathological processes in human body and considered to have great therapeutic potentials. This review summarizes the recent advances on exosome therapy with animal models of ED, and proposes the prospect of future research in order to provide a basis for clinical trials and clinical translation.
    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
    研究背景:海绵状神经损伤(CNI)是前列腺癌根治术和骨盆骨折后勃起功能障碍(ED)的主要原因。人脂肪干细胞(ASCs)的移植已被广泛用于恢复CNI-ED大鼠和患者的勃起功能。脐带血来源的MSCs(CBMSCs)与ASCs相比具有类似的低免疫原性,但是非常原始,并且由于脐带血库的广泛建立,在大规模商业应用中更有希望。然而,CBMSCs和ASCs是否对CNI-ED有不同的治疗效果,潜在的机制尚不清楚。材料与方法:采用双侧海绵体神经粉碎法建立双侧海绵体神经损伤(BCNI)大鼠模型。压碎后,立即海绵状注射ASC和CBMSC。勃起功能,Masson染色,在第4周和第12周评估阴茎组织的免疫荧光分析。经PKH-26标记的ASCs或CBMSCs经海绵体内注射,以在注射后3天确定ASCs或CBMSCs在阴茎中的存在和分化。体外实验包括细胞内ROS检测,线粒体膜电位测定,EdU细胞增殖染色,细胞凋亡试验,与ASC治疗相比,进行了蛋白芯片检测,以探讨CBMSC治疗的潜在机制。结果:CBMSC注射液能显著恢复勃起功能,挽救了海绵状体部平滑肌的损失,并增加平滑肌与胶原蛋白的比例。PKH-26标记的CBMSCs或ASCs不与海绵体中的内皮细胞或平滑肌细胞共定位。此外,条件培养基(CM)能显著抑制雪旺氏细胞的氧化应激,提高线粒体膜电位和增殖。在体内和体外,CBMSC组的治疗效果均优于ASC组。此外,CBMSC-CM中神经营养因子和基质金属蛋白酶的含量,尤其是NT4,VEGF,MMP1、MMP3显著高于ASC-CM。结论:CNI-ED大鼠海绵体内注射CBMSCs的勃起功能恢复优于ASCs,能促进神经再生,减少细胞外基质沉积。CBMSC移植将是未来CNI-ED再生的有希望的治疗策略。
    Background: Cavernous nerve injury (CNI) is the leading cause of erectile dysfunction (ED) after radical prostatectomy and pelvic fracture. Transplantation of human adipose-derived stem cells (ASCs) has been widely used to restore erectile function in CNI-ED rats and patients. Umbilical cord blood-derived MSCs (CBMSCs) are similarly low immunogenic but much primitive compared to ASCs and more promising in large-scale commercial applications due to the extensive establishment of cord blood banks. However, whether CBMSCs and ASCs have differential therapeutic efficacy on CNI-ED and the underlying mechanisms are still not clear. Materials and methods: A bilateral cavernous nerve injury (BCNI) rat model was established by crushing the bilateral cavernous nerves. After crushing, ASCs and CBMSCs were intracavernously injected immediately. Erectile function, Masson staining, and immunofluorescence analyses of penile tissues were assessed at 4 and 12 weeks. PKH-26-labeled ASCs or CBMSCs were intracavernously injected to determine the presence and differentiation of ASCs or CBMSCs in the penis 3 days after injection. In vitro experiments including intracellular ROS detection, mitochondrial membrane potential assay, EdU cell proliferation staining, cell apoptosis assay, and protein chip assay were conducted to explore the underlying mechanism of CBMSC treatment compared with ASC treatment. Results: CBMSC injection significantly restored erectile function, rescued the loss of cavernous corporal smooth muscles, and increased the ratio of smooth muscle to collagen. PKH-26-labeled CBMSCs or ASCs did not colocalize with endothelial cells or smooth muscle cells in the corpus cavernosum. Moreover, the conditioned medium (CM) of CBMSCs could significantly inhibit the oxidative stress and elevate the mitochondria membrane potential and proliferation of Schwann cells. Better therapeutic effects were observed in the CBMSC group than the ASC group both in vivo and in vitro. In addition, the content of neurotrophic factors and matrix metalloproteinases in CBMSC-CM, especially NT4, VEGF, MMP1, and MMP3 was significantly higher than that of ASC-CM. Conclusion: Intracavernous injection of CBMSCs exhibited a better erectile function restoration than that of ASCs in CNI-ED rats owing to richer secretory factors, which can promote nerve regeneration and reduce extracellular matrix deposition. CBMSC transplantation would be a promising therapeutic strategy for CNI-ED regeneration in the future.
    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
    简介:勃起功能障碍(ED)是前列腺癌根治术(RP)后的常见并发症,严重影响患者及其伴侣的生活质量。术后ED的主要触发因素是对海绵状神经的手术损伤,该海绵状神经控制阴茎勃起并沿前列腺前外侧延伸。尽管神经保护技术的引入和不断创新,相当数量的患者仍然患有中度海绵状神经损伤(CNI),这被认为是短暂的和可逆的。因此,术后早期阴茎康复治疗可通过促进海绵状神经再生和防止阴茎结构改变来挽救患者的勃起功能。目的:对CNI诱导的ED的分子发病机制进行全面概述,以及新的治疗策略及其潜在机制。方法:使用PubMed进行文献检索。搜索词包括勃起功能障碍,海绵状神经损伤,发病机制,通路,和治疗。结果:NOS/NO通路,氧化应激相关途径,RhoA/ROCK途径,转化生长因子-β(TGF-β),索尼克刺猬(Shh),硫化氢(H2S)参与CNI诱导ED的分子发病机制。多种神经营养因子,包括脑源性神经生长因子(BDNF),胶质细胞系源性神经营养因子(GDNF),和neurturin(NTN),被发现促进海绵状神经再生。新兴的治疗方法可以大致分为四类,即小分子和药物,基于干细胞的治疗(SCT),微能量疗法和富血小板血浆(PRP)疗法。结论:这些途径共同导致CNI后阴茎结构的不可逆损伤。促进上游神经再生和恢复下游阴茎异常分子信号的联合早期康复策略被认为可以挽救RP后患者的勃起功能。在未来的研究中,这些分子途径之间的串扰需要进一步澄清,以及神经支配损伤如何引起阴茎分子改变的问题也需要解决。
    Introduction: Erectile dysfunction (ED) is a common complication after radical prostatectomy (RP), and it seriously affects the quality of life in patients and their partners. The primary trigger of postoperative ED is surgical injury to the cavernous nerves that control penile erection and run along the anterolateral aspect of the prostate. Despite the introduction and ongoing innovation of nerve-sparing techniques, a significant number of patients still suffer from moderate cavernous nerve injury (CNI), which is thought to be transient and reversible. Therefore, early postoperative penile rehabilitation therapy may salvage patients\' erectile function by promoting cavernous nerve regeneration and preventing penile structural alterations. Aims: To present a comprehensive overview of the current molecular pathogenesis of CNI-induced ED, as well as novel therapeutic strategies and their potential mechanisms. Methods: A literature search was performed using PubMed. Search terms included erectile dysfunction, cavernous nerve injury, pathogenesis, pathway, and treatment. Results: The NOS/NO pathway, oxidative stress-related pathway, RhoA/ROCK pathway, transforming growth factor-β (TGF-β), sonic hedgehog (Shh), and hydrogen sulfide (H2S) are involved in the molecular pathogenesis of CNI-induced ED. Multiple neurotrophins, including brain-derived nerve growth factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and neurturin (NTN), were found to promote cavernous nerve regeneration. Emerging therapeutic approaches can be roughly summarized into four categories, namely small molecule and drug, stem cell-based therapy (SCT), micro-energy therapy and platelet-rich plasma (PRP) therapy. Conclusion: These pathways collectively lead to the irreversible damage to the penile structure after CNI. The combined early rehabilitation strategies of promoting upstream nerve regeneration and recovering abnormal molecular signals of downstream penis are presumed to save patients\' erectile function after RP. In future studies, the cross-talk between these molecular pathways needs to be further clarified, and the questions of how denervation injury induces the molecular alterations in the penis also need to be addressed.
    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
    神经源性勃起功能障碍(nED)是直肠癌和前列腺癌手术最常见和最棘手的术后并发症之一,有时会伴随患者终生。干细胞移植已被证明是一种有希望的治疗方法。然而,由于细胞的过度丢失和死亡以及损伤部位的不良积累以及传统的植入策略,治疗效果严重受损。在这里,设计了一种负载EPO的多功能水凝胶。通过添加儿茶酚-儿茶酚加合物增强了水凝胶的粘合性能和机械强度,因此,显着改善脂肪干细胞(ADSC)保留并挽救损伤部位的细胞损失。同时,EPO的持续释放有效改善了ADSC的活力和旁分泌活性,导致施万细胞的迁移增强和PC12细胞在体内的分化。双侧海绵体神经损伤大鼠模型,本干细胞-EPO-水凝胶植入策略可显著缓解勃起功能障碍.Tuj1的高表达和GFAP在主要骨盆神经节(MPG)中的低表达表明神经分化加速,而星形胶质细胞的发育受到抑制。此外,联合治疗恢复了eNOs的表达水平,nNO,阴茎组织中的α-SMA,建议阴茎康复。进一步分析Masson三色染色和海绵体细胞凋亡评价显示血管内皮含量的保存和去神经后阴茎纤维化的预防。总的来说,我们认为,这种联合策略不仅为恢复神经源性勃起功能,而且为干细胞治疗的临床转化提供了有希望的方法。
    Neurogenic erectile dysfunction (nED) is one of the most common and intractable postoperative complications of rectal and prostate cancer surgery and sometimes accompanies patients lifelong. The transplantation of stem cells has been proved a promising way for treatment. However, the therapeutic efficacy is severely impaired by excessive cell loss and death and poor accumulation in the injury site along with the traditional implantation strategy. Herein, an EPO-loaded multifunctional hydrogel was designed. The hydrogels\' adhesive property and mechanical strength were enhanced by adding catechol-catechol adducts, thus significantly improving adipose-derived stem cells (ADSC) retention and rescuing cell loss in the injury site. Meanwhile, the sustained release of EPO effectively ameliorated the viability and paracrine activity of ADSC, leading to enhanced migration of Schwann cells and differentiation of PC12 cells in vivo. On a bilateral cavernous nerve injury rat model, the present stem cell-EPO-hydrogel implanted strategy could significantly alleviate erectile dysfunction. The higher expression of Tuj1 and lower expression of GFAP in the major pelvic ganglia (MPG) indicated the acceleration of neural differentiation while the suppressing development of astrocytes. Also, the combined therapy restored the expression levels of eNOs, nNOs, and α-SMA in penile tissues, suggesting the rehabilitation of the penis. Further analysis of Masson trichrome staining and apoptosis evaluation of the corpus cavernosum showed the preservation of vascular endothelium content and the prevention of penile fibrosis after denervation. Overall, we believe that this combined strategy presents a promising way not only for restoring neurogenic erectile function but also for the clinical translation of stem cell therapy.
    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
    Pelvic surgery, even without direct cavernous nerve injury, carries a high risk of post-operative erectile dysfunction. The present studies were aimed at identifying molecular mechanisms by which pelvic surgery results in erectile dysfunction. As a model of pelvic surgery, male Sprague-Dawley rats underwent pelvic laparotomy, avoiding direct cavernous nerve injury. A second group of animals, serving as a model of direct cavernous nerve injury, underwent bilateral transection of the cavernous nerve. Cavernosometry demonstrated, that even in the absence of direct nerve injury, the pelvic surgery model exhibited significant erectile dysfunction 3 days post-operatively. Gene expression profiling also demonstrated that even in this animal model of nerve-sparing pelvic surgery, the profile of differentially expressed genes in cavernosal tissue was indicative of cavernous nerve injury. In addition, although 6 hr after surgery there were significant changes in circulating cytokine/chemokine levels, an inflammatory response in the major pelvic ganglion, cavernous nerve and cavernosal tissue was only observed 3 days post-surgery. Our results validate a rat model of pelvic surgery exhibiting erectile dysfunction and suggest systemic release of cytokines/chemokines following surgical trauma might mediate a pathological inflammatory response in tissues distal to the site of surgical trauma, indirectly resulting in cavernous nerve injury and erectile dysfunction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    勃起功能障碍(ED)是一种常见且使人衰弱的疾病,对生活质量有很大影响。ED的根本原因是阴茎平滑肌的凋亡,发生在海绵状神经损伤时,在前列腺切除术中,糖尿病和老年患者。我们正在开发肽两亲物(PA)纳米纤维水凝胶,作为Sonichedgehog蛋白向阴茎和海绵体神经的体内递送载体,以防止凋亡反应。我们研究了生物材料临床应用所需的两个重要方面:如果SHHPA抑制内在(caspase9)和外在(caspase8)凋亡机制,如果抑制一种凋亡机制迫使凋亡通过不同的机制发生。我们显示SHHPA抑制胱天蛋白酶9和8的凋亡机制,抑制caspase9并没有将信号转移到caspase8。SHHPA作为预防性ED治疗具有重要的临床潜力,通过管理内在和外在的凋亡机制。
    Erectile dysfunction (ED) is a common and debilitating condition with high impact on quality of life. An underlying cause of ED is apoptosis of penile smooth muscle, which occurs with cavernous nerve injury, in prostatectomy, diabetic and aging patients. We are developing peptide amphiphile (PA) nanofiber hydrogels as an in vivo delivery vehicle for Sonic hedgehog protein to the penis and cavernous nerve to prevent the apoptotic response. We examine two important aspects required for clinical application of the biomaterials: if SHH PA suppresses intrinsic (caspase 9) and extrinsic (caspase 8) apoptotic mechanisms, and if suppressing one apoptotic mechanism forces apoptosis to occur via a different mechanism. We show that SHH PA suppresses both caspase 9 and 8 apoptotic mechanisms, and suppressing caspase 9 did not shift signaling to caspase 8. SHH PA has significant clinical potential as a preventative ED therapy, by management of intrinsic and extrinsic apoptotic mechanisms.
    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