首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 828 毫秒
1.
Mutations in polycystin-1 (PC1) give rise to autosomal dominant polycystic kidney disease, an important and common cause of kidney failure. Despite its medical importance, the function of PC1 remains poorly understood. Here, we investigated the role of the intracellular polycystin-1, lipoxygenase, and α-toxin (PLAT) signature domain of PC1 using nuclear magnetic resonance, biochemical, cellular, and in vivo functional approaches. We found that the PLAT domain targets PC1 to the plasma membrane in polarized epithelial cells by a mechanism involving the selective binding of the PLAT domain to phosphatidylserine and l-α-phosphatidylinositol-4-phosphate (PI4P) enriched in the plasma membrane. This process is regulated by protein kinase A phosphorylation of the PLAT domain, which reduces PI4P binding and recruits β-arrestins and the clathrin adaptor AP2 to trigger PC1 internalization. Our results reveal a physiological role for the PC1-PLAT domain in renal epithelial cells and suggest that phosphorylation-dependent internalization of PC1 is closely linked to its function in renal development and homeostasis.  相似文献   

2.

Background

Laminins are central components of basement membranes and play important roles in cell adhesion, proliferation, and migration. However, the role of laminins in tumor progression has not been thoroughly investigated in meningiomas.

Objective

The aim of the present study is to evaluate the expression of laminin γ1 in various grades of meningiomas in Chinese patients.

Methods

In the current study, clinical and pathological data for 32 meningioma patients with various tumor grades were collected. The expression of laminin γ1 in each tumor was assessed by using quantitative real-time polymerase chain reaction (qPCR), Western blot and immunohistochemical analysis and was correlated with the meningioma grade, tumor recurrence and patient survival. Patient prognoses were attained and the progression-free survival was calculated based on the Kaplan-Meier method. A two-sided probability cutoff of 0.05 was chosen for statistical significance.

Results

A total of 32 meningioma patients with various pathological subtypes (WHO grade I: 13, grade II: 10 and grade III: 9) were enrolled in this study. The qPCR results showed that laminin γ1 mRNA expression was significantly higher in grade III meningiomas than in grade I meningiomas (p?<?0.05), although there was no significant difference in laminin γ1 expression between grade II and grade I meningiomas (p?>?0.05). Western blot and immunohistochemistry analysis confirmed that the expression of laminin γ1 protein was relatively higher in grade III meningiomas when compared with grade I meningiomas. Higher levels of laminin γ1 expression in meningiomas are associated with a significantly shorter tumor recurrence time (p?<?0.05) and a decreased patient survival time (p?<?0.05).

Conclusions

Our results suggest that laminin γ1 is associated with meningioma grades and could play a role in enhancing tumor invasion. Laminin γ1 could be used as a predictor for meningioma recurrence and patient survival. Furthermore, laminin γ1 may represent a druggable molecular target for future therapies for tumors that overexpress this marker.  相似文献   

3.
Tumor necrosis factor α (TNFα) is a multifunctional cytokine that regulates various cellular processes related to spermatogenesis. Two types of cell receptors, TNFR1 and TNFR2, mediate TNFα activity. In the present study, we sought to explore the association of TNFα -857C→T, TNFR1 36A→G, and TNFR2 676T→G polymorphisms with sperm concentration and motility. Two hundred ninety men were examined during infertility investigation; of those, 170 men were normozoospermic and 120 were oligospermic. Polymerase chain reaction analysis revealed significant differences in genotype distribution of the TNFR1 36A→G polymorphism between normozoospermic and oligospermic men. Men with oligozoospermia presented TNFR1 36A/A genotypes less frequently than normozoospermic men (P < .001). The presence of the TNFR1 36G allele was significantly increased in oligospermic men (P < .001). Furthermore, the presence of the TNFR1 36G allele was associated with lower sperm concentration in normozoospermic men (P < .03) and in the total study population (P < .001), and with lower sperm motility in normozoospermic men (P < .007) and in the total study population (P < .001). No significant associations were found between TNFα -857C→T and TNFR2 676T→G polymorphisms and semen quality. The TNFR1 36A allele is associated with increased sperm concentration and motility in our series, supporting the significance of TNFR1 gene in semen quality.  相似文献   

4.
Because of the increasing interest on stimulating fracture healing, knowledge about the role and chronology of growth factors during the healing process is important. The purpose of this study was to quantify the protein concentration of IGF-I and TGF-1 during rat tibial fracture healing 5, 10, and 15 days after fracture using ELISA methods and to analyze the distribution of the proteins and the related mRNA expression in the fracture callus by immunohistochemistry and in situ hybridization. The following three groups were analyzed: Fractured tibiae intramedullary stabilized with K-wires coated with IGF-I and TGF-1 compared with fractures stabilized with uncoated K-wires and unfractured tibiae. The weight of the callus increased during the healing process in both experimental groups. The protein concentration of IGF-I and TGF-1 in the fracture callus showed significant changes between the investigated time points and treatment groups compared with the unfractured tibia. IGF-I increased with healing time whereas TGF-1 revealed a constantly elevated level at the investigated time points. Mesenchymal cells, osteoblasts, osteocytes, proliferating and immature chondrocytes, and osteoclasts expressed both growth factors. No differences in the expression and localization pattern of the growth factors were detectable among the groups. Using the different methods for quantification and visualization of the growth factors, no differences (except the increased IGF-I concentration at day 15 in the growth factor group) were seen between the normal and the growth factor-stimulated fracture healing as an indication for physiological healing after exogenous growth factor treatment. Parts of this study have been presented at the annual meeting of the ORS 2002 (Orthopaedic Research Society, Dallas, Feb. 2002) (Trans Orthop Res Soc 48th, poster).  相似文献   

5.
腹腔镜膀胱根治性切除原位新膀胱技术现已成熟,并广泛开展,主要分为经腹膜外及腹膜内两种手术入径。经腹膜内途径为主流选择,但不能最大程度保留腹膜,而腹膜外途径,能够最大程度保留腹膜甚至能够完全保留腹膜,减少因腹膜缺失造成的术后并发症,同时可在围手术期严格按照加速康复外科(ERAS)理念管理。我科开展了ERAS理念下经腹膜外腹腔镜膀胱根治性切除术,将原位膀胱置于腹膜外,完全保留腹膜。本文就术中经腹膜外途径、盆底腹膜保留、重建的手术步骤及技术要点进行总结。  相似文献   

6.
7.
This study was designed to determine the expression and localization of the α-adrenoceptor (AR) subtypes in human ureteral tissue. The expression of the α-AR subtypes was examined by immunohistochemistry using subtype selective antibodies in proximal, mid and distal ureter. Ureter samples were obtained from pathological specimens of nephroureterectomy. A single pathologist scored the expression of receptor (grade 0, no staining; grade 1, 0–25% cells positive; grade 2, 26–50% cells positive; and grade 3, greater than 50%). We compared the mean grades of α-AR 1A, 1B and 1D and the percentage of receptor expression with grade 2 or more between receptor subtypes in each ureter level. The expression levels of all three subtypes are altered according to the level of ureter. In the proximal ureter, the mean grades of the α-1A, -1B and -1D receptors were 1.0 ± 0.5, 1.1 ± 0.6, and 2.1 ± 0.7. Corresponding grades in the mid and distal ureter were 1.1 ± 0.6, 0.8 ± 0.6 and 2.0 ± 0.8, and 2.0 ± 0.7, 1.8 ± 0.6 and 2.5 ± 0.5, respectively. When compared with percentage of high expression, in the proximal and mid ureter, α-1D high expression percentage was significantly higher than α-1A and -1B subtype (80, 10, 20, and 90, 20, 10%, respectively). In the distal ureter, α-1D expression was higher than α-1A and -1B subtype but there was no statistical significance (100, 80, 70%). The distal ureter had higher density of α-AR receptors than proximal and mid ureter. The expression of α-1A and α-1B AR in distal ureter was significantly higher than proximal and mid ureter. α-1D expression in distal ureter was also higher than proximal and mid ureter but was not statistically significant (p = 0.28, 0.19, respectively). Our results show that α-1A, -1B and -1D AR subtypes are localized in human ureter irrespective of location. The expression levels of subtypes are altered according to level of ureter and subtype. This study was supported by Seoul National University Hospital Clinical Research Institute (06-2005-164-0). An erratum to this article can be found at  相似文献   

8.
Cellular cholesterol homeostasis is important for normal β-cell function. Disruption of cholesterol transport by decreased function of the ATP-binding cassette (ABC) transporter ABCA1 results in impaired insulin secretion. Mice lacking β-cell ABCA1 have increased islet expression of ABCG1, another cholesterol transporter implicated in β-cell function. To determine whether ABCA1 and ABCG1 have complementary roles in β-cells, mice lacking ABCG1 and β-cell ABCA1 were generated and glucose tolerance, islet sterol levels, and β-cell function were assessed. Lack of both ABCG1 and β-cell ABCA1 resulted in increased fasting glucose levels and a greater impairment in glucose tolerance compared with either ABCG1 deletion or loss of ABCA1 in β-cells alone. In addition, glucose-stimulated insulin secretion was decreased and sterol accumulation increased in islets lacking both transporters compared with those isolated from knockout mice with each gene alone. Combined deficiency of ABCA1 and ABCG1 also resulted in significant islet inflammation as indicated by increased expression of interleukin-1β and macrophage infiltration. Thus, lack of both ABCA1 and ABCG1 induces greater defects in β-cell function than deficiency of either transporter individually. These data suggest that ABCA1 and ABCG1 each make complimentary and important contributions to β-cell function by maintaining islet cholesterol homeostasis in vivo.  相似文献   

9.
Diabetic nephropathy is a serious microvascular complication and one of the main causes of end-stage renal disease. L-Glutamine (LG) is naturally occurring amino acids with antidiabetic and antioxidant potential. The aim of present investigation was to evaluate the potential of LG against streptozotocin (STZ)-induced diabetic nephropathy (DN) in laboratory rats. DN was induced in male Wistar rats (200–220?g) by intraperitoneal administration of STZ (55?mg/kg). Animals were treated orally with either distilled water (10?mg/kg) or LG (250, 500, and 1000?mg/kg) or Sitagliptin (5?mg/kg). Various biochemical, molecular, and histological (hematoxylin–eosin and Masson’s trichrome stain) parameters were assessed. Administration of LG (500 and 1000?mg/kg) significantly inhibited (p?<?.05) STZ-induced alterations in serum and urine biochemistry (urine creatinine, uric acid, albumin, and BUN). It also significantly increased creatinine clearance rate. STZ induced increase in renal oxidonitrosative stress was significantly decreased (p?<?.05) by LG (500 and 1000?mg/kg) treatment. Upregulated renal KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expression after STZ administration was significantly inhibited (p?<?.05) by LG (500 and 1000?mg/kg) treatment. Correlation analysis also revealed that antidiabetic potential of LG attenuates STZ-induced elevated renal KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expression. Histopathological alteration induced by STZ in renal tissue was ameliorated by LG treatment. In conclusion, results of present investigation suggest that treatment with LG ameliorated STZ-induced DN via the inhibition of oxidonitrosative stress as well as downregulation of KIM-1, NGAL, TGF-β1, and collagen-1 mRNA expressions.  相似文献   

10.
Akirav E  Kushner JA  Herold KC 《Diabetes》2008,57(11):2883-2888
OBJECTIVE— β-Cell regeneration is a fundamental but elusive goal for type 1 diabetes research. Our objective is to review newer human and animal studies of β-cell destruction and regeneration and consider the implications for treatment of type 1 diabetes.RESEARCH DESIGN AND METHODS— Recent human and animal studies of β-cell destruction and regeneration in type 1 diabetes are reviewed.RESULTS— The loss of β-cells that characterizes type 1 diabetes reflects the net effects of destruction and regeneration. These processes have been examined in the nonobese diabetic (NOD) mouse; uncertainty remains about β-cell dynamics in humans. Islet inflammation stimulates β-cell replication that produces new insulin-positive cells. The regenerative process may tide the loss of overall β-cell function, but it also may enhance the autoimmune attack on β-cells by providing new epitopes. The highest rates of β-cell replication are at the time of diagnosis of diabetes in NOD mice, and if autoimmunity and islet inflammation are arrested, new β-cells are formed. However, the majority of β-cells after treatment with immune modulators such as anti-CD3 monoclonal antibody, and most likely during the “honeymoon” in human disease, are recovered β-cells that had been degranulated but present at the time of diagnosis of diabetes.CONCLUSIONS— Residual β-cells play a significant role for the design of therapeutic trials: they not only may respond to combination therapies that include stimulants of metabolic function but are also the potential source of new β-cells.Studies largely done in nonobese diabetic (NOD) mice have described a linear loss of β-cell mass from the time of initiation of insulitis through presentation with hyperglycemia and afterward until there is complete loss of β-cells (1,2). Results from the Diabetes Prevention Trial-1 (DPT-1) have described changes in insulin secretion from pre-diabetes to presentation with hyperglycemia, and other studies have depicted the progressive loss of insulin secretion after diagnosis (35). Whereas the data from animal studies and clinical investigations are largely consistent, other studies indicate that a simple linear loss of β-cell function may be an oversimplification of a process that involves an undulating downhill course. Variation in the rate of progression of β-cell loss may be due to waxing and waning of the inflammatory response because of exposure of new antigens; intercurrent insults to β-cells involving infectious agents, metabolic demands, or other factors; and a continuous attempt at β-cell regeneration that tides the progression. Inflammation itself appears to stimulate β-cell regeneration, but at the same time may expose new antigenic epitopes that become the drivers of a broadening autoimmune response (6). Ultimately, the failure to contain and counter this enlarging process results in presentation with clinical disease. Understanding the way in which the process develops and progresses has implications for treatment and reversal of the disease.

β-cell turnover is increased during progression of diabetes.

In NOD mice, β-cell proliferation increases with islet inflammation during progression of diabetes. By 4 weeks of age, the proportion of Ki67+insulin-positive cells in the islets of Langerhans in NOD mice is significantly increased compared with control NOD/scid mice that do not have insulitis (7). β-Cell proliferation continues to increase, with insulitis, during the development of disease; by 12 weeks of age, 3.03 ± 0.94% of insulin-positive cells are Ki67+. The highest levels of Ki67+insulin-positive cells are found at the time that the mice develop hyperglycemia. The increased rate of β-cell proliferation is found even before a significant decline in β-cell mass or an increase in glucose levels suggesting that metabolic demands per se are not responsible for the β-cell proliferation. Similar findings of heightened proliferation of β-cells were reported by Sreenan et al. (1) using bromodeoxyuridine (BrdU) labeling. These investigators also found an inverse change in β-cell mass and proliferation rates.The β-cell proliferation is linked to islet inflammation (7). It increases in the recipients of diabetogenic spleen cells that are transferred to NOD/scid recipients. By 4 weeks after transfer of cells, the rates of β-cell replication increases more than 10-fold compared with NOD/scid mice (from 0.22 ± 0.05% to 3.14 ± 1.07%, P = 0.007) (7). Measures that modulate islet inflammation decrease β-cell proliferation. After treatment with anti-CD3 monoclonal antibody (mAb), β-cell proliferation fell from 3.09 ± 0.8% (at diagnosis) to 1.57 ± 0.28% 3 weeks after treatment (P < 0.05). Moreover, when diabetogenic spleen cells were transferred into NOD/scid recipients together with CD4+CD25+ regulatory T-cells, the rate of diabetes decreased from 79 to 17% (P < 0.01), but β-cell proliferation also decreased from 2.95 ± 0.27 to 1.98 ± 0.26%.The increased rates of β-cell proliferation that are found in mice with insulitis result in an increase in the mass of “new” β-cells if autoimmunity is arrested. To demonstrate this directly, we treated hyperglycemic NOD mice with anti-CD3 mAb to reverse hyperglycemia, studied the changes in β-cell area after treatment, and enumerated the proportion of new β-cells after recovery of diabetes. We added BrdU to the drinking water of the mice after treatment with anti-CD3 mAb and enumerated the “new” β-cells in the islets after euglycemia was established. In the islets of these mice, 10.9 ± 1.38% of the β-cells were BrdU+ 3 weeks after mAb treatment compared with 4.22 ± 0.49% of the β-cells in untreated NOD/scid mice that were given BrdU over the same period of time (P < 0.001) (7).These findings are consistent with the recent report by Nir et al. (8) that there may be spontaneous recovery of β-cell mass and function in the absence of autoimmunity. In these studies, the investigators acutely eliminated 70–80% of β-cells by the transgenic expression of diphtheria toxin. When the transgene expression was quenched, hyperglycemia resolved over a 4-week period and β-cell mass increased. Dor, Melton, and colleagues (9) have previously observed that β-cell mass expansion primarily occurs via insulin-positive precursors. Indeed, diphtheria-mediated β-cell mass reduction primarily occurs by expansion of insulin-positive progenitors (8). New data indicate specialized progenitors do not contribute to β-cell mass growth or regeneration (10,11). Taken together, these studies suggest that β-cell mass in young rodents is highly dynamic and that non–autoimmune-stimulated β-cell regeneration primarily occurs via simple β-cell replication.Despite the consistent evidence for β-cell regeneration in rodents, it is not clear whether similar β-cell dynamics occur in humans. Butler et al. (12) did not find evidence for increased β-cell replication in patients with recent-onset diabetes who died of ketoacidosis compared with age-matched subjects. In’t Veld et al. (13) did identify Ki67+insulin-positive cells in islets with insulitis in anti-islet autoantibody–positive organ donors, but the percentage of islets in the pancreas with insulitis was small (3 and 9%). However, there are differences between the material sampled in these two reports and the pancreases of NOD mice that develop diabetes. The timing of the analysis is different in patients with diabetes compared with pre-diabetic NOD mice, and in the case of the organ donors, it is not clear whether the pathologic process that was studied would in fact lead to diabetes. Nonetheless, these observational studies identify a number of important differences between human disease and the NOD model. First, the proliferative responses of human islets may be different from those seen in rodents. Second, the inflammatory response in islets of patients with diabetes is clearly different from that seen in NOD mice in which inflammatory infiltrates that permeate and often obliterate islets are routinely seen (14). The insulitis that occurs in patients is reduced, involving 0–33% of islets and in only 52% of islets, and the modest cellular infiltrate is predominated by CD8+ T-cells (15,16).

The dynamic process of new β-cell formation and β-cell destruction creates an environment for expression and presentation of new antigenic targets.

The mechanisms involved in the destruction of β-cells have been the subject of intensive research and include direct lysis of β-cells by CD8+ T-cells or other lytic cells, the damaging effects of cytokines that are produced by effector CD4+ T-cells that recognize their antigenic targets on non–β-cells, or even by non–T-cells that release innate inflammatory mediators, such as interleukin (IL)-1β, which can damage β-cells and break tolerance through activation of adaptive responses (1725). The turnover of β-cells may fuel this process through the expression of new antigens or possibly by post-translational modification of proteins, which has been described in other disease settings (6,2628). Some experimental evidence suggests that β-cell death throughout the life of NOD mice during development primes autoreactive T-cells. The detection of dead β-cells in pancreatic lymph nodes by day 20 of age and the ability of isolated dendritic cells from these lymph nodes to stimulate the TCR transgenic diabetogenic T-cell, BDC2.5, provides an indication for the role of β-cell death and contraction during development in the priming of autoreactive T-cells (29). β-Cell injury and death may underlie the spreading of specificities of pathogenic autoimmune cells during progression of disease by exposing new epitopes (30). Epitope spreading allows for the activation of new T- and B-cell clones in response to novel antigens that are expressed in the target organ. Isolated T-cells from NOD mice showed a wider ability to respond to novel intramolecular determinants of GAD65, which increased with age (31). In fact, tolerized NOD mice showed reduced recognition of novel epitope by autoreactive T-cells and protection from disease. Not only intramolecular but also intermolecular epitope spreading is observed in the NOD mouse (32).At least one protein, reg, is involved in β-cell proliferation and can serve as an autoantigen (33,34). In the NOD mouse, array studies have shown reg gene expression during progression of disease as one of the products to which cellular immune responses ultimately develop (26). The recognition of new antigens may be the consequence or the cause of progression. Ott et al. (35) have examined the reactivity of T-cells from type 1 diabetic patients, people at high risk of developing diabetes, and healthy control subjects who respond to GAD65 and proinsulin in vitro. The authors demonstrated increased T-cell reactivity to GAD65 antigens and to proinsulin in both type 1 diabetic patients and high-risk individuals. T-cell reactivity to whole protein and the variable memory response in different type 1 diabetic patients led the authors to conclude that cryptic epitopes are recognized in type 1 diabetes. They found that epitope spreading occurs in these individuals, whereas these cryptic epitopes do not activate T-cells from individuals who do not progress to disease.The acquisition of antigenicity during progression of diabetes is more accurately described as instructive rather than stochastic. There is an orderly unfolding of antigenic responses over time: there is a hierarchy of antigens. Recent studies by Nakayama et al. and Krishnamurthy et al. support the notion that insulin is a “primary” antigen in NOD diabetes, whereas the islet-specific G6Pase catalytic subunit-related protein (IGRP), clearly an antigen recognized by diabetogenic T-cells later in the course of the disease, appears to be secondary. Insulin 1 and insulin 2 gene knockout mice with a mutated proinsulin transgene (in which residue 16 on the B-chain was changed to alanine) in NOD mice did not develop insulin autoantibodies, insulitis, or diabetes (36). NOD mice tolerized to proinsulin did not develop cytotoxic IGRP-reactive T-cells or diabetes, whereas mice tolerized to IGRP did not develop IGRP reactive T-cells but did develop diabetes (22). These results provide direct evidence that the response against IGRP is downstream of the response to proinsulin in this model. Human data are also consistent with this notion: autoantibody responses to insulin are highest in younger individuals and the risk of disease increases with the number of antigens that are recognized (3739). Most information on the specificities of antigenic targets is cross-sectional rather than prospective, but the available evidence supports a sequential rather than simultaneous appearance of autoantigen reactivity (38).

Presentation with hyperglycemia reflects a transient shift in the dynamic process that may be precipitated by acute islet inflammation, increased insulin resistance, or other factors.

As β-cell mass declines, a critical level is reached at which small disturbances in either insulin requirements or insulin production can have a large effect on the clinical picture. Acute viral infections are one example. Coxsackie B4 virus has been isolated from the blood of patients with new-onset diabetes, and certain of these isolates have been shown to cause diabetes in mice (40). Histopathologic studies have shown expression of IFN-α in the islets of individuals who have died at the time of presentation of type 1 diabetes (A. Poulous, personal communication). A recent study found evidence for infection with Coxsackie B4 enterovirus in the pancreata of three of six patients with type 1 diabetes whose pancreas was examined up to 9 months after diagnosis (a pancreas graft was studied from one patient) (41). Interestingly, in two of the patients, the number of β-cells was similar to that from three age-matched nondiabetic control subjects, whereas in three patients, the islets with enterovirus showed reduced insulin secretion in response to secretagogues and the virus from the islets could infect islets from nondiabetic donors and cause viral inclusions and pyknosis. There was NK cell (CD94+) and to a lesser extent T-cell infiltration, occasional B-cell and CD68+ cell infiltration, and IFN-α expression in three of the patients, suggesting ongoing or previous islet viral infection.In addition to reduced insulin production, insulin resistance has been suggested as an accelerator of diabetes at presentation. Insulin resistance has been thought to contribute to the frequent presentation of type 1 diabetes during adolescence when insulin sensitivity declines. Innate immune responses may account both for impaired insulin sensitivity as well as a decline in β-cell function (42). Koulmanda et al. (43) found that “triple therapy” with rapamycin, IL-2 Ig, and IL-15 Ig, which reversed hyperglycemia in NOD mice, dampened the expression of inflammatory genes including IL-1β, tumor necrosis factor-α, and granzyme in diabetic mice. Interestingly, these investigators found that the triple therapy restored insulin signaling in peripheral tissues suggesting that inflammatory mediators may affect responses as well as production of insulin. This is consistent with clinical findings of insulin resistance at the time of presentation of diabetes and the widely recognized role of inflammatory products such as tumor necrosis factor-α in mediating it (44,45). In addition, prospective human data from the Diabetes Prevention Trial have shown that factors other than a precipitous decline in insulin secretion account for clinical presentation. The comparison between the C-peptide responses to either a mixed meal or oral glucose before and after diagnosis showed only a modest difference, where the major effect is on the peak C-peptide response to the metabolic challenge. Basal insulin secretion is relatively well preserved at the time of diagnosis (3,5,46). In our studies of β-cell function before and after diagnosis, the C-peptide response to a mixed-meal test after diagnosis was 87 ± 7% (n = 31) of the prediagnosis response. It was on a third test, in a subset of the original subjects, that was done 6 months or more later, that a more dramatic decrease to 53 ± 13% (n = 10) of the response before diagnosis was seen. In an analysis of the C-peptide response to an oral glucose tolerance test, the total C-peptide area under the curve was reduced by 18% from 6 months before to after diagnosis. These studies suggest that in addition to the decline in C-peptide responses, additional factors are involved in the metabolic decompensation at presentation.

β-Cell area degranulation and regranulation?

In NOD mice, there is a precipitous drop in the β-cell area at presentation with hyperglycemia (2). The effects of the acute precipitants of disease may not be permanent and may be partially reversible. Indeed, the rapid recovery of β-cell function after the diagnosis during the “honeymoon” period suggests a reversible component to the metabolic dysfunction at diagnosis. Our studies of β-cell mass in NOD mice at diagnosis of type 1 diabetes are consistent with this notion. As noted above, there was a significant increase in β-cell area after treatment with anti-CD3 mAb, but only ∼11% of the β-cells were new (i.e., BrdU+ and insulin-positive). Therefore, ∼90% of the β-cells had been present in the islets at the time of presentation with hyperglycemia. The difference between the total area after correction of hyperglycemia (0.367 ± 0.065 mm2) and the β-cell area found at diagnosis (0.086 ± 0.04 mm2) plus the 11% of the β-cells that had been newly formed (0.037 mm2) does not account for the entire β-cell mass. This suggests that there had been β-cells in the islets at the time of diagnosis that could not be identified by conventional staining with insulin and accounted for over half of the β-cells that were found after correction of hyperglycemia. When we stained islets from mice with new-onset diabetes with antibodies to GLUT2, we identified GLUT2+ insulin cells in the islets at diagnosis, but all of the GLUT2+ cells in the islets after treatment with anti-CD3 mAb were insulin-positive. The GLUT2+ insulin cells that were found in the islets at diagnosis may represent a functionally “exhausted” or degranulated pool of cells that have not been destroyed and may recover. Thus, the precipitation of disease onset may involve a rapidly accelerating cycle in which an initial loss of β-cell function, due to inflammatory mediators, coupled with a reduced β-cell mass after autoimmune destruction, results in modest hyperglycemia, which is met by increased demands on the residual β-cells to produce insulin (Fig. 1). If the expansion of β-cell mass or improvement in function does not keep pace with the demands, additional β-cells may become functionally exhausted and degranulated leading to worsening degrees of hyperglycemia (47,48). The immediate recovery after the initial treatment with immune modulators and/or even insulin involves the net effect of β-cell replication during islet inflammation, but even more significantly, the recovery of β-cell function that was reduced at the time of diagnosis. It should be noted, however, that although insulin treatment and correction of the decompensated metabolic conditions results in temporary improvement in insulin responses in humans, a similar “honeymoon” does not occur in NOD mice treated with insulin. The differences between mice and humans could reflect differences in the relative pace of the autoimmune process or the significant impact of the precipitants of human disease. It may also reflect our relatively poor ability to control the glucose level in NOD mice with new-onset diabetes. This resulting condition, however, is precarious because modest increases in insulin requirements, inflammatory insults to the islets, or progression of the anti-islet adaptive response may result in recurrence of the cycle.Open in a separate windowFIG. 1.Changes in β-cells during progression of type 1 diabetes and the selection of interventions: the initial event that primes the adaptive immune response is not known but is postulated to involve β-cell death. Antigen-specific immune interventions (red arrow) have the greatest chance of efficacy in this early stage. With progression of the response, there are increased rates of β-cell proliferation but also a loss of β-cells and expansion of the immune response. A greater proportion of replicating β-cells is seen in the islet as insulitis progresses, and some β-cells are unable to keep up with the metabolic demands. Most likely, a broader approach to immune intervention is needed at this point, since the autoreactive repertoire is broader than at the earlier stages. Left untreated, there is continued loss of β-cells. However, interventions such as immune modulators can prevent the ongoing adaptive immune response against β-cells and, with metabolic treatment, recovery of degranulated β-cells can occur.

Implications for therapy of diabetes.

These new concepts regarding the dynamics of β-cell mass in type 1 diabetes have implications for the design of clinical interventions that are targeted to arrest the adaptive autoimmune response. First, the total amount of residual β-cell mass is an important variable in the outcome of immune intervention. Metabolic interventions will only be effective if there is sufficient β-cell mass with which they can work. Furthermore, some studies indicate that new β-cells are derived by replication of existing cells and therefore growth of the total β-cell mass will be more substantial with greater starting material (911). The clinical data have supported the importance of residual β-cell function in promoting responses to interventions. In the studies of anti-CD3 mAb by Keymeulen et al. (49), the greatest responses to the immune intervention were in individuals with the highest C-peptide levels at the time of diagnosis. To improve the recovery of diabetes in NOD mice after anti-CD3 mAb, we tested the addition of the GLP-1 receptor agonist, exendin-4, to treatment with anti-CD3 mAb (50). Previous studies had indicated that exendin-4 increased replication of β-cells in rodents and had glucose-lowering effects in mice and humans (5154). We found that exendin-4 enhanced the reversal of diabetes in mice in which the glucose level was <350 mg/dl at the time of diagnosis. These mice would be expected to have a residual β-cell mass that is greater than those with higher glucose levels at diagnosis in which the exendin-4 and anti-CD3 mAb were less effective. The total β-cell area and rate of β-cell proliferation was similar in islets from mice that did or did not receive exendin-4 with anti-CD3 mAb. The exendin-4 treatment did not have an effect on the immune response or the antigenicity of the islets. However, the glucose tolerance of the exendin-4–treated mice and the insulin content of their pancreases were improved compared with those treated with just the anti-CD3 mAb. Thus, it may be possible to enhance the efficacy of single immunologic agents by combining them with agents that have other mechanisms such as enhancing β-cell function. With this combination, the efficacy of the combination will depend on the residual β-cell mass that can be affected by the drug.Finally, in our studies, immune therapy diminished the replicative response of β-cells that are found during development of diabetes. More than 6 weeks after treatment of NOD mice with anti-CD3 mAb, β-cell replication fell to levels that were less than in younger mice but were still higher than in control (NOD/scid) mice (1.11 ± 0.15 vs. 0.37 ± 0.07%, P < 0.001) (7), possibly reflecting some degree of ongoing inflammation or residual β-cell deficit. The “boost” of β-cell replication that occurs during inflammation may therefore decline, but the recent studies by Nir et al. (8) imply that there may be spontaneous recovery of β-cell mass if inflammation and autoimmunity are prevented.In summary, clinical and preclinical studies suggest that there is a progressive loss of β-cell mass and function from the initiation of insulitis through presentation with hyperglycemia and afterward. However, the course is undulating and in mice, reflects the opposing factors of β-cell destruction by an expanding adaptive immune response, β-cell replication, and the effects of acute insults with infectious agents and other inflammatory mediators. The accelerated rates of β-cell turnover during progression may be an incendiary to the process by providing and presenting new antigenic targets. This model has largely been developed in mice; human studies have yet to confirm the β-cell replicative process or even the extent of insulitis that is found in the NOD mouse. At the time of diagnosis, a critical threshold of β-cell function has been crossed, but there is potentially recoverable β-cell mass that can restore β-cell function to a prediagnosis level. However, even with recovery, β-cell mass is significantly reduced. Therefore, increases in insulin demand or further progression of the immune process results in significant clinical consequences and further loss of β-cells. The time at which the progression of disease is identified and treated with immune modifiers is of practical importance, since the residual cells may represent the source of new β-cells and combination agents are more likely to be effective if there are β-cells that can respond.  相似文献   

11.
Recently, nephrin, podocin, -actinin, and WT1, which are located at the slit diaphragm and expressed by the podocyte, were found to be causative in congenital/familial nephrotic syndrome (NS), but their role in acquired NS remains unclear. We studied their expression in NS with the aim of disclosing their possible role in the development of proteinuria. Immunofluorescence, confocal microscopy, and image analysis were used to study the expression and the distribution in 19 children with primary NS, 9 with isolated hematuria, and 9 controls. All the children with NS presented with heavy proteinuria and foot process effacement was identified by electron microscopy. No proteinuria and foot process effacement was seen in the group with hematuria. A dramatic decrease of podocin expression was found in NS (86.66±22.74) compared with control groups (P=0.014). Furthermore, we also found the pattern of distribution of nephrin, podocin, and -actinin changed in children with NS. In conclusion, a dramatic decrease of podocin expression and abnormal distribution of nephrin, podocin, and -actinin were found in children with NS. No differences were found in children with isolated hematuria, suggesting involvement of these molecules in the development of proteinuria in primary NS.  相似文献   

12.
13.
We investigated the association between IL-1, IL-1ra, and TNF- gene polymorphisms and childhood nephrotic syndrome (NS). We analyzed the genetic polymorphism of IL-1, IL-1ra, and TNF- genes in 152 patients with childhood NS and 292 healthy adult controls. The C to T exchange at position –511 of IL-1 and the G to A at –308 of the TNF- gene were genotyped. Five alleles of the IL-1ra gene were identified and designated as IL1RN*1, IL1RN*2, IL1RN*3, IL1RN*4, and IL1RN*5, according to the variable number of tandem repeats in intron 2. The allele frequencies of IL-11 (-511C), IL-12 (-511T), TNF1 (-308G), and TNF2 (-308A) were 53.0, 47.0, 92.1, and 7.9%, respectively, in the childhood NS group. This was not significantly different from normal controls. In the childhood NS group, the allele frequencies of IL1RN*1, IL1RN*2, IL1RN*3, IL1RN*4, and IL1RN*5 were 90.8, 7.6, 1.6, 0, and 0% [IL1RN*1 odds ratio (OR)=0.296, P=0.0001, IL1RN*2 OR=3.902, P=0.0002]. A high allele frequency of IL1RN*2 and a lower allele frequency of IL1RN*1 were found in childhood NS, although there was no association with IL-1 and TNF-. A high allele frequency of the IL1RN*2 allele may affect disease susceptibility in childhood NS.  相似文献   

14.
15.
16.
17.

Introduction and hypothesis  

We investigated whether the expression of alpha-1 antitrypsin (ATT), neutrophil elastase (NE), and lysyl oxidase-like protein 1 (LOXL-1) vary within the vagina in subjects with pelvic organ prolapse (POP).  相似文献   

18.
19.
目的 探讨腹腔镜经肠系膜途径输尿管切开取石术治疗中上段输尿管结石临床应用价值。方法 回顾地分析2016年1月至2019年3月腹腔镜下经肠系膜途径治疗的68例输尿管中上段结石患者临床资料。记录患者的性别、年龄、手术时间、术中出血量、术后肠道恢复时间,术后症状改善和B超复查结果。结果 67例手术取石顺利,有1例结石上行到肾盂而行体外冲击波碎石治疗。手术时间(41.2±4.7)min,术中出血量(5.3±1.6)mL,术后肠道恢复时间(9.5±2.1)h。68例患者随访1~36个月,全部患者肾输尿管积水明显好转,无结石复发和输尿管切开缝合处狭窄。结论 对体形瘦、肠系膜脂肪少的中上段输尿管结石患者,腹腔镜经结肠系膜途径输尿管切开取石安全有效,可以减少创伤、缩短肠道恢复及手术时间。  相似文献   

20.

Summary

Parathyroid hormone (PTH) augments bone metabolism and bone mass when given intermittently. Enhanced blood flow is requisite to support high tissue metabolism. The bone arteries are responsive to all three PTH analogs, which may serve to augment skeletal blood flow during intermittent PTH administration.

Introduction

PTH augments bone metabolism. Yet, mechanisms by which PTH regulates bone blood vessels are unknown. We deciphered (1) endothelium-dependent and endothelium-independent vasodilation to PTH 1–84, PTH 1–34, and PTHrP 1–34, (2) the signaling pathways (i.e., endothelial nitric oxide synthase [eNOS], cyclooxygenase [COX], protein kinase C [PKC], and protein kinase A [PKA]), and (3) receptor activation.

Methods

Femoral principal nutrient arteries (PNAs) were given cumulative doses (10?13–10?8 M) of PTH 1–84, PTH 1–34, and PTHrP 1–34 with and without signaling pathway blockade. Vasodilation was also determined following endothelial cell removal (i.e., denudation), PTH 1 receptor (PTH1R) inhibition and to sodium nitroprusside (SNP; a nitric oxide [NO] donor).

Results

Vasodilation was lowest to PTH 1–34, and maximal dilation was highest to PTHrP 1–34. Inhibition of eNOS reduced vasodilation to PTH 1–84 (?80 %), PTH 1–34 (?66 %), and PTHrP 1–34 (?48 %), evidencing the contribution of NO. Vasodilation following denudation was eliminated (PTH 1–84 and PTHrP 1–34) and impaired (PTH 1–34, 17 % of maximum), highlighting the importance of endothelial cells for PTH signaling. Denuded and intact PNAs responded similarly to SNP. Both PKA and PKC inhibition diminished vasodilation in all three analogs to varying degrees. PTH1R blockade reduced vasodilation to 1, 12, and 12 % to PTH 1–84, PTH 1–34, and PTHrP 1–34, respectively.

Conclusions

Vasodilation of femoral PNAs to the PTH analogs occurred via activation of the endothelial cell PTH1R for NO-mediated events. PTH 1–84 and PTHrP 1–34 primarily stimulated PKA signaling, and PTH 1–34 equally stimulated PKA and PKC signaling.
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号