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 31 毫秒
1.
Obliterative bronchiolitis after lung transplantation is a chronic inflammatory and fibrotic condition of small airways. The fibrosis associated with obliterative bronchiolitis might be reversible. Matrix metalloproteinases (MMPs) participate in inflammation and tissue remodeling. MMP-2 localized to myofibroblasts in post-transplant human obliterative bronchiolitis lesions and to allograft fibrosis in a rat intrapulmonary tracheal transplant model. Small numbers of infiltrating T cells were also observed within the fibrosis. To modulate inflammation and tissue remodeling, the broad-spectrum MMP inhibitor SC080 was administered after the allograft was obliterated, starting at post-transplant day 21. The allograft lumen remained obliterated after treatment. Only low-dose (2.5 mg/kg per day) SC080 significantly reduced collagen deposition, reduced the number of myofibroblasts and the infiltration of T cells in association with increased collagenolytic activity, increased MMP-2 gene expression, and decreased MMP-8, MMP-9, and MMP-13 gene expression. In in vitro experiments using cultured myofibroblasts, a relatively low concentration of SC080 increased MMP-2 activity and degradation of type I collagen. Moreover, coculture with T cells facilitated persistence of myofibroblasts, suggesting a role for T-cell infiltration in myofibroblast persistence in fibrosis. By combining low-dose SC080 with cyclosporine in vivo at post-transplant day 28, partial reversal of obliterative fibrosis was observed at day 42. Thus, modulating MMP activity might reverse established allograft airway fibrosis by regulating inflammation and tissue remodeling.Chronic allograft dysfunction after lung transplantation is manifested by obliterative bronchiolitis (OB), a fibroproliferative obstructive lesion in small airways, and its clinical correlate, bronchiolitis obliterans syndrome (BOS).1,2 Once the fibrotic process of OB is initiated, conventional immunosuppression is usually ineffective.3 The traditional pathological perspective is that fibrosis is the end result of damage: scar tissue, with no possibility of return to the pre-existing structure.4 However, increasing evidence suggests that fibrosis still undergoes dynamic remodeling and is potentially a reversible process. For example, the resolution of liver fibrosis is well documented both clinically and experimentally. In animal experiments, up-regulation or overexpression of matrix metalloproteinases (MMPs) capable of degrading interstitial type I and type III collagen (including MMP-1,5 MMP-8,6 MMP-13,7and MMP-2 and MMP-148,9) is associated with the regression of liver fibrosis. Pulmonary fibrosis has also been shown to be conditionally reversible.10One possible mechanism rendering fibrosis unlikely to resolve is the aberrant persistence of myofibroblasts, an active form of fibroblasts positive for α-smooth muscle actin (α-SMA), which leads to production of extracellular matrix (ECM) in excess of MMP-dependent ECM degradation.11 Unresolved inflammation can be an important contributor to this mechanism.10 Accumulating evidence suggests that chronic fibrotic conditions are mediated by complex interactions between immune and nonimmune cells, in which the persistence of a relatively low grade of inflammation continuously stimulates resident stromal cells12,13 and provides survival signals to myofibroblasts.14 For instance, the resolution of liver fibrosis encountered in alcohol-induced and virus-related fibrosis occurs only after remedy of the underlying cause.15,16 Moreover, in experimental models of fibrosis, reversal of fibrosis has occurred in one-hit injury models such as bleomycin-induced pulmonary fibrosis,17 in which the initial tissue injury leads to fibrosis but the tissue injury or inflammation is not continuous.8,9Along those lines, OB after lung transplantation is a fibrotic and chronic inflammatory condition18 in which myofibroblasts persist.19 The intrapulmonary tracheal transplant model of OB is a unique animal model in which persistent alloantigen from the donor trachea within the pulmonary milieu causes continuous alloantigen-induced inflammation and results in robust fibrosis in the allograft lumen.20 We have previously demonstrated that myofibroblasts expressing high levels of collagen and MMP-2 and MMP-14 play a central role in the remodeling of established allograft airway fibrosis.20 Given that MMPs also play important but complex roles in the trafficking of immune responsive cells,20 MMPs involved in both tissue remodeling and inflammation may play key roles in the reversal of fibrosis.We therefore hypothesized that allograft airway fibrosis is a potentially reversible process involving MMPs. Here, we demonstrate expression patterns of MMPs in established human OB lesions and describe the roles of MMPs in the remodeling of collagen matrix, myofibroblasts, and immune responsive cells using in vivo and in vitro models with SC080, a general MMP inhibitor. Finally, we demonstrate for the first time reversibility of allograft airway fibrosis by combining immunosuppression with a low dose of SC080.  相似文献   

2.
The Commentary highlights the article by Humphreys et al, “Fate Tracing Reveals the Pericyte and Not Epithelial Origin of Myofibroblasts in Kidney Fibrosis” discussing hypothesis of renal progression.Regardless of the initial cause of renal injury, failing kidneys approaching end-stage disease typically show marked tubular atrophy and interstitial fibrosis. In addition, in both animal models and humans, once the number of functioning nephrons falls below a critical level, there is an inexorable progression of further nephron loss, tubular atrophy, and interstitial fibrosis. Therefore, it is clearly of importance to understand the pathophysiology of this process. Recent research on the progression of renal disease has centered on three related but independent hypotheses:
  • 1.The final common pathway of renal injury is interstitial fibrosis and therefore preventing interstitial fibrosis will slow the progressive decline in renal function.
  • 2.A major source of the fibroblasts that produce interstitial collagen in the injured kidney is tubular epithelial cells that have differentiated to fibroblasts and crossed the basement membrane. This process has been referred to as epithelial-mesenchymal transition (EMT) and elucidation of this process has been a major focus of research over the past decade. The belief is that understanding this process will allow interventions that will slow interstitial fibrosis.
  • 3.In glomerular disease, proteinuria is a major stimulus to the alteration of epithelial cell function and leads to EMT and interstitial fibrosis.
These three hypotheses have been widely accepted and, indeed, in most review articles the process of EMT is often stated as fact. However, as with all hypotheses, it is important to continue to test them with the best methods available. In this issue of The American Journal of Pathology, Humphreys et al1 have performed elegant studies that cast considerable doubt on the existence of EMT in the kidney.They have used genetic manipulation to develop mice that express protein labels exclusively in renal tubular epithelial cells. They then induced injury in the kidney and, by looking for labeled cells, were able to determine whether any tubular cells had moved into the interstitium or had acquired the ability to behave as fibroblasts.Using the cre/lox technique to label cells, which depends on crossing mice that express the enzyme cre-recombinase under the control of a cell-lineage specific promoter with mice expressing a reporter gene that is activated by cre-recombinase, Humphreys et al1 generated two types of mice: one in which all tubular epithelial cells except those in the collecting duct were labeled and one in which collecting duct cells were labeled. The authors then subjected these mice to two types of injury that led to interstitial fibrosis: unilateral ureteric obstruction (UUO) and ischemia-reperfusion injury. Histological examination of the kidneys postinjury revealed interstitial fibrosis with many cells in the interstitium expressing α-smooth muscle actin (α-SMA), a marker of myofibroblasts. However, the authors did not find any labeled tubular epithelial cells in the interstitium nor any tubular cells that expressed α-smooth muscle actin, implying that EMT did not occur.Humphreys et al1 performed a number of critical controls. One possible explanation for their data is that the marker protein is not expressed by fibroblast-like cells. They therefore used two different marker proteins and showed the same results with both. In addition, they examined mice that expressed the marker protein β-galactosidase in all cells and showed that it continued to be expressed by the interstitial cells in their model. They also studied whether the labeled epithelial cells could undergo changes in cell culture that other investigators have considered part of the process of EMT. They confirmed this by showing that when the cells were incubated with transforming growth factor-β, they expressed α-SMA and S100A4, markers that have generally been considered as evidence of EMT, while also continuing to express the marker protein.Thus, this study provides strong evidence against the process of EMT having a significant involvement in interstitial fibrosis in two models of renal fibrosis and complements other studies that have also cast doubt on its existence. In one such study, Faulkner et al2 studied an accelerated model of angiotensin II-induced renal fibrosis and found no evidence for an epithelial origin for interstitial fibroblasts.So where do the interstitial fibroblasts come from in these models? Another theory that has attracted support is that some interstitial fibroblasts are derived from circulating cells of bone marrow origin. Our group tested this hypothesis by using a mouse that expressed a reporter gene under the control of the promoter of the α-2 chain of type I collagen and found no evidence that cells from the bone marrow could synthesize collagen in the kidney after unilateral ureteric obstruction.3 Similarly, Lin et al4 failed to find a significant contribution of circulating cells to renal fibroblasts by using the promoter of the α-1 chain of type I collagen. In fact, it appears that the major source of interstitial collagen-producing cells is from fibroblasts that reside within the interstitium itself and particularly from cells in the adventitia of arterioles and arteries.2,5 In agreement with this, Humphreys et al1 also now show that cells of metanephric mesenchymal origin expressing platelet-derived growth factor receptor β (PDGFRβ), consistent with pericytes, are the major source of interstitial fibroblasts in unilateral ureteric obstruction.If it is true that EMT does not contribute significantly to interstitial fibrosis, and I think the evidence against EMT is now strong, one has to ask why this theory had achieved such widespread support and what is the evidence in its favor. In part it is because the expression epithelial mesenchymal transition has been applied to a range of phenomena, and there has been confusion as to how the term is used. At one end of the spectrum it has been used to describe changes that renal tubular epithelial cells can show in culture whereby they express makers such as α-SMA that are more typical of cells of mesenchymal origin. That this occurs is uncontroversial and indeed Humphreys et al1 show that their labeled epithelial cells express α-SMA after stimulation with transforming growth factor-β. It is also clear that when renal tubular cells are damaged in vivo they undergo morphological changes of dedifferentiation and may sometimes express markers such as α-SMA.6 However, the demonstration of these changes falls a long way short of what is usually meant by EMT, which is that these cells traverse the basement membrane and start to synthesize type I collagen. I think the evidence in favor of this is much more tenuous.A great deal of weight has been placed on the expression by tubular epithelial cells of a protein that has been referred to as fibroblast specific protein 1, which is a form of S100 protein, S100A4.7 Indeed, one of the major articles supporting a role for both tubular epithelial cells and circulating cells differentiating to interstitial fibroblasts relies mainly on the co-expression of S100A4.8 In that article, the authors used a lineage-specific marker for tubular epithelial cells and showed that in UUO there was cellular co-localization of the tubular marker and S100A4. However, the expression of this protein by epithelial cells inside the tubular basement membrane is no more evidence for their ability to become interstitial fibroblasts than is expression of another marker such as α-SMA. In addition, there is evidence that S100A4 can be expressed by leukocytes in injured kidneys,7 by podocytes in diabetic nephropathy,9 and by many other cell types including human monocytes and macrophages (reviewed in Mazzucchelli10), and so it is very doubtful whether it can be considered a fibroblast-specific protein.In summary, it is clear that when renal tubular epithelial cells are injured they undergo morphological and biochemical changes and may express proteins that can also be expressed by cells of mesenchymal origin. This is likely to be related to their ability to repair and regenerate after acute tubular damage. However, as the study by Humphreys et al1 demonstrates, there is little evidence that these altered cells can move into the interstitium and produce collagen, and the main collagen-producing cells are intrinsic renal cells, particularly perivascular cells. However, this does not exclude a possible role for communication between the damaged epithelial cells and interstitial cells that may be important in stimulating fibrosis.If EMT does not occur, what about the other two hypotheses mentioned earlier? I think these also need critical examination. The first states that the final common pathway of renal injury is interstitial fibrosis and therefore preventing interstitial fibrosis will slow the progressive decline in renal function. This is often stated as if it is self-evident. However, while it is obvious that almost all severely damaged kidneys show marked fibrosis, it is much less clear that preventing the fibrosis would have prevented loss of renal function. Morphologically, fibrosis is seen in association with atrophic tubules. It is likely that the fibrosis is a secondary response to the tubular damage and that preventing fibrosis would not necessarily alter the tubular degeneration. This is particularly relevant to glomerular diseases where glomerular scarring, if it involves the origin of the proximal tubule, leads to secondary tubular atrophy and fibrosis around the atrophic tubule. However, fibrosis in that case is a secondary response and preventing it would not alter the tubular damage.As has been pointed out by Kriz and LeHir,11 in a very thorough review, the hypothesis that fibrosis itself leads to progressive decline in renal function is dependent on fibrosis around a damaged nephron then causing injury to adjacent nephrons, but there is no experimental evidence for this. Indeed, careful morphological studies12 show that it is only in nephrons with irreversible glomerular injury that the tubules appear damaged with no evidence of spread to adjacent tubules. Thus, in glomerular disease at least, there is no evidence for progression of the disease at the level of the tubulointerstitium. Indeed, it may be that by replacing damaged nephrons with scar tissue the interstitial reaction confines the degenerative change to the damaged nephron. This does not exclude a role for interstitial fibrosis in progression of other processes such as transplant rejection but does emphasize the need for more careful evaluation of pathophysiology in different settings.What about the third hypothesis that in glomerular disease, proteinuria is a major stimulus to alteration of epithelial cell function and leads to EMT and interstitial fibrosis? Even if we accept that EMT does not occur, it is plausible that tubular cells damaged by protein, or other substances in the tubular lumen, might signal to cells in the interstitium and that this might stimulate interstitial fibroblasts to lay down collagen.13 This would also fit with the fact that the magnitude of proteinuria correlates well with the rate of progression of renal disease. However, much of the experimental evidence that has been put forward to support this is consistent with other mechanisms, and there are several authors who argue against its importance as discussed in detail in the review by Kriz and Le Hir.11 Careful analysis of experimental glomerular injury suggests that, even with severe glomerular injury, the proximal tubule remains healthy unless the glomerular damage encroaches on the glomerulotubular junction. In agreement with this, abrogation of protein uptake into proximal tubules in transgenic mice with a kidney-specific deficiency of megalin did not alter tubular degenerative changes in crescentic glomerulonephritis and, in fact, megalin-deficient tubular cells showed increased apoptosis.14In conclusion, I believe that all three of the hypotheses with which I began this commentary need careful re-evaluation. Their uncritical acceptance risks diverting research efforts from other areas that are potentially of more importance in the progression of renal disease including the control of scarring versus repair in glomeruli, and the role of the peritubular capillaries. The article by Humphreys et al1 is an important step in this re-evaluation.  相似文献   

3.
4.
5.
Matrix metalloproteinases (MMPs), a specialized group of enzymes capable of proteolytically degrading extracellular matrix proteins, have been postulated to play an important role in angiogenesis. It has been suggested that MMPs can regulate neovascularization using mechanisms other than simple remodeling of the capillary basement membrane. To determine the interplay between vascular endothelial growth factor (VEGF) and MMPs, we investigated the induction of angiogenesis by recombinant active MMPs and VEGF in vivo. Using a rat corneal micropocket in vivo angiogenesis assay, we observed that the active form of MMP-9 could induce neovascularization in vivo when compared with the pro- form of the enzyme as a control. This angiogenic response could be inhibited by neutralizing VEGF antibody, which suggests that MMPs acts upstream of VEGF. Additional in vitro studies using extracellular matrix loaded with radiolabeled VEGF determined that active MMPs can enzymatically release sequestered VEGF. Interestingly, in vivo angiogenesis induced by VEGF could be inhibited by MMP inhibitors, indicating that MMPs also act downstream of VEGF. In addition, inflammation plays an important role in the induction of angiogenesis mediated by both VEGF and MMPs. Our results suggest that MMPs act both upstream and downstream of VEGF and imply that potential combination therapies of VEGF and MMP inhibitors may be a useful therapeutic approach in diseases of pathological neovascularization.Angiogenesis, the sprouting of new capillaries from pre-existing blood vessels, is a multistep process requiring the degradation of the basement membrane, endothelial cell migration, endothelial cell proliferation, and capillary tube formation. Precise spatial and temporal regulation of extracellular proteolytic activity mediated by matrix-degrading enzymes appears to be important in the initial process of endothelial cell invasion into the extracellular matrix (ECM).1 Three families of enzymes, the matrix metalloproteinases (MMPs), a disintegrin and metalloprotease domain (ADAM) family, and a disintegrin-like and metalloprotease domain (reprolysin type) with thrombospondin type I repeats (ADAMTS) family2 mediate the proteolysis of ECM proteins.MMPs (eg, collagenases, gelatinases, and stromelysins) are a family of zinc binding, Ca2+-dependent neutral endopeptidases that can act together or in concert with other enzymes to degrade most components of the ECM.3,4 These enzymes have been implicated in invasive cell behavior and recent studies have indicated that MMPs play an important role in the regulation of angiogenesis.5–8 Mice deficient in MMP-2 (gelatinase A), MMP-9 (gelatinase B), or MMP-14 exhibit reduced angiogenesis in vivo,9–11 and members of the tissue inhibitor of metalloproteinase family are potent angiogenesis inhibitors.12,13A number of mechanisms by which remodeling of the ECM by MMPs and other proteases can regulate angiogenesis have been proposed.6,14,15 Since MMPs degrade proteins in the ECM, their primary function has been considered to be the breakdown of the capillary basement membrane to allow the migration of endothelial cells into the surrounding matrix. More recently, additional ectodomain shedding and release of matrix-bound angiogenic factors, cytokine receptors, and adhesion molecules, mediated by MMPs,5 have been suggested to contribute to this process. Tumorigenesis experiments have proposed that vascular endothelial growth factor (VEGF) may be released from the ECM by gelatinase B (MMP-9) and result in the angiogenic switch.5 Other in vitro studies have suggested that VEGF mediates its angiogenic effects by up-regulation of MMPs.16,17 In an effort to determine the interplay between VEGF and MMPs in the mediation of angiogenesis and to ascertain if MMPs act upstream or downstream of VEGF, we investigated the induction of angiogenesis in vivo by recombinant active MMPs and VEGF and the potential inhibitory activities of their respective inhibitors.  相似文献   

6.
7.
8.
Guillain-Barré syndrome (GBS) and chronic inflammatory demyelinating polyneuropathy (CIDP) are conditions that affect peripheral nerves. The mechanisms that underlie demyelination in these neuropathies are unknown. Recently, we demonstrated that the node of Ranvier is the primary site of the immune attack in patients with GBS and CIDP. In particular, GBS patients have antibodies against gliomedin and neurofascin, two adhesion molecules that play a crucial role in the formation of nodes of Ranvier. We demonstrate that immunity toward gliomedin, but not neurofascin, induced a progressive neuropathy in Lewis rats characterized by conduction defects and demyelination in spinal nerves. The clinical symptoms closely followed the titers of anti-gliomedin IgG and were associated with an important deposition of IgG at nodes. Furthermore, passive transfer of antigliomedin IgG induced a severe demyelinating condition and conduction loss. In both active and passive models, the immune attack at nodes occasioned the loss of the nodal clusters for gliomedin, neurofascin-186, and voltage-gated sodium channels. These results indicate that primary immune reaction against gliomedin, a peripheral nervous system adhesion molecule, can be responsible for the initiation or progression of the demyelinating form of GBS. Furthermore, these autoantibodies affect saltatory propagation by dismantling nodal organization and sodium channel clusters. Antibodies reactive against nodal adhesion molecules thus likely participate in the pathologic process of GBS and CIDP.Guillain-Barré syndrome (GBS) is a group of inflammatory neuropathies that affect peripheral nerves. In Europe, acute inflammatory demyelinating polyneuropathy (AIDP) is the most common form of GBS. Autopsy and biopsy studies indicated that both humoral and cellular immune reaction against Schwann cell or axonal antigens are implicated in GBS etiology.1 Early investigations have found that conduction defects closely correlate with myelin retraction and macrophage invasion in many patients.2, 3, 4, 5 Some GBS cases also involve acute demyelination without immune cell invasion and are primarily humorally mediated.6, 7 In particular, deposition of complement on the abaxonal surface of the Schwann cells has been shown during the early stage of GBS8, 9, 10 and in experimental allergic neuritis (EAN).11 In a recent study, we demonstrated that nodes of Ranvier and paranodes are the targets of the immune attack in GBS and in chronic inflammatory demyelinating polyneuropathy (CIDP).12 Notably, cell adhesion molecules (CAMs) at nodes or paranodes (gliomedin, neurofascin, and contactin) were recognized by IgG antibodies in patients with GBS or CIDP.12, 13 Autoantibodies against neurofascin and gliomedin were also detected in a rat model of AIDP and correlated with important conduction defects.14 This finding suggested that antibodies to nodal CAMs may participate to the pathogenesis of AIDP and CIDP. However, the exact mechanisms by which these humoral factors mediate demyelination and conduction defects are still elusive.Several CAMs are implicated in node formation and are responsible for the enrichment of voltage-gated sodium (Nav) channels at the nodes of Ranvier.15 At peripheral, nodes gliomedin and NrCAM are secreted into the nodal gap lumen and interact with neurofascin-186 (NF186) expressed at nodal axolemma.16, 17, 18, 19 This interaction is crucial for Nav channel aggregation at nodes.19, 20, 21 In addition, the paranodal axoglial junctions are made by the association of contactin and contactin-associated protein (Caspr) with neurofascin-155 (NF155), a variant expressed in glia.22 This adhesive junction forms a barrier to the lateral diffusion of nodal channels.19, 21, 23 In a rat model of AIDP, we found that the loss of NF186 and gliomedin at nodes preceded paranodal demyelination and the diffusion of Nav channels in demyelinated segments.14 This finding indicated that antibodies to nodal CAMs may participate to conduction defects by dismantling axoglial attachment at nodes and paranodes.We investigated whether immunity toward gliomedin and NF186 can trigger peripheral neuropathies and be responsible for demyelination in GBS patients. We found that immunization against gliomedin induced a biphasic condition associated with conduction loss and demyelination. Passive transfer of antibodies to gliomedin exacerbated the clinical signs of EAN and resulted in the disorganization of the nodes of Ranvier. Altogether, these results demonstrate that humoral immune response directed against nodal CAMs participates in conduction abnormalities in peripheral nerves and in the etiology of GBS and CIDP.  相似文献   

9.
Nonalcoholic fatty liver disease is an increasingly prevalent spectrum of conditions characterized by excess fat deposition within hepatocytes. Affected hepatocytes are known to be highly susceptible to ischemic insults, responding to injury with increased cell death, and commensurate liver dysfunction. Numerous clinical circumstances lead to hepatic ischemia. Mechanistically, specific means of reducing hepatic vulnerability to ischemia are of increasing clinical importance. In this study, we demonstrate that the glucagon-like peptide-1 receptor agonist Exendin 4 (Ex4) protects hepatocytes from ischemia reperfusion injury by mitigating necrosis and apoptosis. Importantly, this effect is more pronounced in steatotic livers, with significantly reducing cell death and facilitating the initiation of lipolysis. Ex4 treatment leads to increased lipid droplet fission, and phosphorylation of perilipin and hormone sensitive lipase – all hallmarks of lipolysis. Importantly, the protective effects of Ex4 are seen after a short course of perioperative treatment, potentially making this clinically relevant. Thus, we conclude that Ex4 has a role in protecting lean and fatty livers from ischemic injury. The rapidity of the effect and the clinical availability of Ex4 make this an attractive new therapeutic approach for treating fatty livers at the time of an ischemic insult.The incidence of obesity and fatty liver disease is increasing worldwide. Non alcoholic fatty liver disease (NAFLD) includes a spectrum of liver abnormalities ranging from simple steatosis with preserved synthetic function to end-stage liver disease requiring transplantation.1, 2 The cause of hepatic dysfunction related to steatosis remains incompletely defined.3 However, it is known that a steatotic liver has increased susceptibility to ischemic insults, such as those induced during liver resections and liver surgery,4, 5, 6 heart failure,7 and shock.8 In addition, steatotic livers are known to weather the ischemic insult of transplantation poorly,9 resulting in increased rates of primary nonfunction and initial graft dysfunction.10, 11 As such, fatty livers are routinely turned down for transplantation and this impacts transplant wait list morbidity and mortality.12 Thus, liver steatosis contributes to the public health burden and methods to mollify the adverse effects of liver steatosis are relevant across a large spectrum of hepatic diseases.The inability of a steatotic liver to withstand ischemic insult is directly related to increased post ischemic cell death, which can occur through necrosis and apoptosis. The fundamental connection between intracellular fat and poor hepatic cell survival13 is incompletely understood. However, it has been suggested that methods that decrease intracellular fat reverse this susceptibility and the use of glucagon-like peptide-1 (GLP-1) analogues is one such approach. GLP-1 is secreted from the L cells of the small intestine and its cognate receptor (GLP-1R) is present in several organs, such as the pancreas, brain, heart, kidney, and liver. Although it is well known for its incretin action,14 it also has pleotropic effects.15, 16, 17, 18, 19 In the liver we have shown that GLP-1 or its homologue Exendin 4 (Ex4) acts directly on steatotic hepatocytes to decrease their lipid content.20, 21 In addition, a cytoprotective action of Ex4 with improvement in cell survival has also been reported.22 Thus, we hypothesize that anti-steatotic effects of Ex4 in hepatocytes and cytoprotective effects in other organs make it a rational target for investigation in steatotic livers undergoing ischemia reperfusion injury (IRI), a common clinical scenario in people with NAFLD. In this study, we explore the role of Ex4 in protecting against necrosis and apoptosis, the two forms of cell death encountered in hepatic IRI, and we provide evidence to show that Ex4 stimulates lipolysis with a short course of treatment. To our knowledge, this is the first study showing a direct and rapid action of Ex4 in acutely reversing the vulnerability of a steatotic liver to ischemic insults, supporting the investigation of Ex4 as a potential therapeutic agent for treatment of people with NAFLD undergoing ischemic injury and at the time of procurement of a fatty liver for transplantation.  相似文献   

10.
Autophagy mediates bulk degradation and recycling of cytoplasmic constituents to maintain cellular homeostasis. In response to stress, autophagy is induced and may either contribute to cell death or serve as a cell survival mechanism. Very little is known about autophagy in renal pathophysiology. This study examined autophagy and its pathological role in renal cell injury using in vitro and in vivo models of ischemia−reperfusion. We found that hypoxia (1% O2) induced autophagy in cultured renal proximal tubular cells. Blockade of autophagy by 3-methyladenine or small-interfering RNA knockdown of Beclin-1 and ATG5 (two key autophagic genes) sensitized the tubular cells to hypoxia-induced apoptosis. In an in vitro model of ischemia−reperfusion, autophagy was not induced by anoxic (0% O2) incubation in glucose-free buffer, but was induced during subsequent recovery/reperfusion period. In this model, suppression of autophagy also enhanced apoptosis. In vivo, autophagy was induced in kidney tissues during renal ischemia−reperfusion in mice. Autophagy was not obvious during the ischemia period, but was significantly enhanced during reperfusion. Inhibition of autophagy by chloroquine and 3-methyladenine worsened renal ischemia/reperfusion injury, as indicated by renal function, histology, and tubular apoptosis. Together, the results demonstrated autophagy induction during hypoxic and ischemic renal injury. Under these pathological conditions, autophagy may provide a protective mechanism for cell survival.Autophagy is a cellular process of “self-eating” wherein various cytoplasmic constituents are broken down and recycled through the lysosomal degradation pathway.1 This process consists of several sequential steps, including sequestration of cytoplasmic portions by isolation membrane to form autophagosome, fusion of the autophagosome with lysosome to create an autolysosome, and degradation of the engulfed material to generate monomeric units such as amino acids.2 Identification of the autophagy-related genes (ATG) in yeast and their orthologs in other organisms including mammals demonstrates that autophagy is evolutionarily conserved in all eukaryotic cells. The ATG genes constitute the core molecular machinery of autophagy and function at the different levels to regulate autophagy induction, progression, and completion.1Autophagy occurs at basal level in most cells and contributes to the turnover of long-lived proteins and organelles to maintain intracellular homeostasis. In response to cellular stress, autophagy is up-regulated and can provide an adaptive strategy for cell survival, but may also directly or indirectly lead to cell demise.3–6 With the dual role in life and death, autophagy is involved in various physiological processes, and more importantly, linked to the pathogenesis of a wide array of diseases, such as neurodegeneration, cancer, heart disease, aging, and infections.1,2,6,7 However, it remains largely unknown how autophagy makes the life and death decisions of a stressed cell. Moreover, the conundrum is further complicated by the cross talk and coordinated regulation between autophagy and apoptosis.4,5,8Despite rapid progress of autophagy research in other organ systems, the role of autophagy in the pathogenesis of renal diseases was not recognized until very recently. In 2007, Chien et al9 suggested the first evidence of autophagy during renal ischemia−reperfusion in rats. Subsequent work by Suzuki et al10 further showed autophagy in ischemic mouse kidneys and notably, in transplanted human kidneys. In nephrotoxic models of acute kidney injury, we and others have demonstrated autophagy during cisplatin nephrotoxicity and have suggested a role for autophagy in renoprotection.11,12 A prosurvival role of autophagy was also shown in tubular cells during cyclosporine A nephrotoxicity.13 In contrast, Gozuacik et al14 suggested that autophagy may serve as a second cell killing mechanism that acts in concert with apoptosis to trigger kidney damage in tunicamycin-treated mice. A cell killing role for autophagy was also suggested by Suzuki et al10 during H2O2-induced renal tubular cell injury. As a result, whether autophagy is a mechanism of cell death or survival in renal pathology remains unclear.In this study, we have determined the role of autophagy in renal tubular cell injury using in vitro and in vivo models of renal ischemia−reperfusion. We show that autophagy is induced in these models. Importantly, blockade of autophagy sensitizes renal cells and tissues to injury by hypoxia and ischemia−reperfusion, suggesting a prosurvival role for autophagy.  相似文献   

11.
Adhesions between organs after abdominal surgery remain a significant unresolved clinical problem, causing considerable postoperative morbidity. Osteopontin (OPN) is a cytokine up-regulated after cell injury and tissue repair. Our previous studies have shown that blocking OPN expression at sites of cutaneous wounding resulted in reduced granulation tissue and scarring. We hypothesize that it may be possible to similarly reduce inflammation-associated fibrosis that causes small-bowel adhesions after abdominal surgery. By using a mouse model, we deliver OPN antisense oligodeoxynucleotides via Pluronic gel to the surface of injured, juxtaposed small bowel and show a significant reduction of inflammatory cell influx to the developing adhesion and a dramatic reduction in the resulting adhesion size. A significant reduction in α-smooth muscle actin expression and collagen deposition within the mature adhesion is also demonstrated. We see no impact on mortality, and the healing of serosal injury to intact bowel appeared normal given the reduced inflammatory response. Our studies suggest that dampening OPN levels might be a potentially important target for anti-adhesion therapeutics.The peritoneum is an extensive and complex organ consisting of a layer of mesothelial cells lining the peritoneal cavity and all organs within it.1 One of the main functions of the peritoneum is to allow friction-free movement between abdominal viscera and the peritoneal wall.2 Any surgery that breaches the peritoneal lining causes injury to the peritoneum, which responds by raising inflammatory signals that attract innate immune cells in parallel with a wound repair response and subsequent fibrosis.3–5 This almost invariably results in permanent peritoneal adhesion formation.6 The result can be tethering of adjacent small-bowel loops that may lead to abdominal pain7 and/or bowel obstruction,8 which is a significant cause of postoperative morbidity in clinical practice. Readmission rates secondary to adhesional complications are as high as 5% to 10% after abdominal surgery.9,10 Adhesion prevention options in clinical practice are limited to either barrier methods11 or flotation fluids,12 which use the concept of keeping damaged peritoneal surfaces separated during their healing process; however, these options are of limited effectiveness.13,14 Pathophysiological manipulation of the cascade events leading to fibrosis has been investigated,15–18 but none has led to a clinically usable product. Herein, we investigate whether therapeutic strategies used to block scar formation after skin healing might also be effective during peritoneal repair. Microarray studies of wound tissues from wild-type mice versus PU.1 mice (lacking neutrophils, macrophages, and mast cells) reveal an inflammation-dependent gene, osteopontin (OPN), that is expressed by wound granulation tissue fibroblasts, coincident with a skin wound inflammatory response.19,20 PU.1 mice heal skin wounds without the standard inflammatory cascade, which results in less fibrosis and scarring at the healed wound site.19 OPN acts both as a secreted chemokine-like protein and as part of an intracellular signaling complex.21 It plays key roles in several processes associated with tissue repair, including cell adhesion, migration, and survival.21,22 Short-term local knockdown of OPN in cutaneous wounds leads to decreased granulation tissue and reduced scar formation.23 In this study, we investigate whether these effects are transferable to peritoneal repair and also might block i.p. fibrosis.  相似文献   

12.
13.
Molecular chaperones of the heat shock protein-90 (Hsp90) family promote cell survival, but the molecular requirements of this pathway in tumor progression are not understood. Here, we show that a mitochondria-localized Hsp90 chaperone, tumor necrosis factor receptor-associated protein-1 (TRAP-1), is abundantly and ubiquitously expressed in human high-grade prostatic intraepithelial neoplasia, Gleason grades 3 through 5 prostatic adenocarcinomas, and metastatic prostate cancer, but largely undetectable in normal prostate or benign prostatic hyperplasia in vivo. Prostate lesions formed in genetic models of the disease, including the transgenic adenocarcinoma of the mouse prostate and mice carrying prostate-specific deletion of the phosphatase tensin homolog tumor suppressor (Ptenpc−/−), also exhibit high levels of TRAP-1. Expression of TRAP-1 in nontransformed prostatic epithelial BPH-1 cells inhibited cell death, whereas silencing of TRAP-1 in androgen-independent PC3 or DU145 prostate cancer cells by small interfering RNA enhanced apoptosis. Targeting TRAP-1 with a novel class of mitochondria-directed Hsp90 inhibitors, ie, Gamitrinibs, caused rapid and complete killing of androgen-dependent or -independent prostate cancer, but not BPH-1 cells, whereas reintroduction of TRAP-1 in BPH-1 cells conferred sensitivity to Gamitrinib-induced cell death. These data identify TRAP-1 as a novel mitochondrial survival factor differentially expressed in localized and metastatic prostate cancer compared with normal prostate. Targeting this pathway with Gamitrinibs could be explored as novel molecular therapy in patients with advanced prostate cancer.Apart from skin tumors, prostate cancer is the most commonly diagnosed malignancy in men in the United States.1 Despite progress in early diagnosis,2 and prolongation of patient survival,3 the disease still carries significant morbidity and mortality, with its advanced and metastatic phase claiming over 30,000 deaths per year in the United States alone. Similar to the genetic heterogeneity of most epithelial malignancies, prostate cancer progresses through a stepwise acquisition of multiple molecular changes,4 of which insensitivity to androgen deprivation,5 emergence of an ‘osteomimetic’ phenotype responsible for metastatic tropism to the bone,6 and deregulated cell proliferation and cell survival,7 are pivotal traits.In this context, advanced prostate cancer is almost invariably associated with a heightened anti-apoptotic threshold,4 which may contribute to disease progression and resistance to therapy. This process often involves aberrant resistance to mitochondrial cell death,8 with reduced organelle permeability to solutes, and attenuated release of mitochondrial apoptogenic proteins in the cytosol.9 The regulators of such ‘mitochondrial permeability transition’ normally triggered by cell death stimuli are still largely elusive, but knockout data in mice have identified pro-apoptotic Bcl-2 family proteins and the mitochondrial matrix immunophilin, cyclophilin D, as pivotal effectors of this process, controlling the integrity of the mitochondrial outer membrane,8 and the opening a permeability transition pore,10,11 respectively.Recent data have shown that molecular chaperones of the heat shock protein-90 (Hsp90) family,12 may function as novel regulators of mitochondrial permeability transition,13 especially in tumor cells.14 Accordingly, Hsp90, and its ortholog, tumor necrosis factor receptor-associated protein-1 (TRAP-1) are abundantly localized to mitochondria of tumor, but not most normal cells, and antagonize cyclophilin D-dependent pore-forming function, potentially via a protein (re)folding mechanism.14 Consistent with a general role of Hsp90 as a drug target in prostate cancer,15 this mitochondria-compartmentalized cytoprotective pathway could provide a novel therapeutic target to enhance tumor cell apoptosis.14In the current study, we demonstrate that TRAP-1 is dramatically expressed in all lesions that comprise the entire natural history of human prostate cancer, as well as genetic disease models in rodents, but undetectable in the normal prostate. Importantly, we show that Gamitrinibs, a novel class of small molecule Hsp90 antagonists selectively engineered to target the pool of these chaperones in mitochondria,16 cause sudden prostate cancer cell death without affecting nontransformed prostatic epithelium.  相似文献   

14.
MCM7 is one of the pivotal DNA replication licensing factors in controlling DNA synthesis and cell entry into S phase. Its expression and DNA copy number are some of the most predictive factors for the growth and behavior of human malignancies. In this study, we identified that MCM7 interacts with the receptor for activated protein kinase C 1 (RACK1), a protein kinase C (PKC) adaptor, in vivo and in vitro. The RACK1 binding motif in MCM7 is located at the amino acid 221-248. Knocking down RACK1 significantly reduced MCM7 chromatin association, DNA synthesis, and cell cycle entry into S phase. Activation of PKC by 12-O-tetradecanoylphorbol-13-acetate dramatically decreased MCM7 DNA replication licensing and induced cell growth arrest. Activation of PKC induced redistribution of RACK1 from nucleus to cytoplasm and decreased RACK1-chromatin association. The MCM7 mutant that does not bind RACK1 has no DNA replication licensing or oncogenic transformation activity. As a result, this study demonstrates a novel signaling mechanism that critically controls DNA synthesis and cell cycle progression.Miniature chromosome maintenance (MCM) proteins were initially identified from autonomously replicating sequence in Saccharomyces cerevisiae. Mutations of some of these proteins, such as MCM7 or MCM3 result in loss of the large chunk of yeast chromosomes in yeast. MCM7 cDNA encodes a 543-amino acid protein and is ubiquitously expressed in all tissues. A large body of studies indicate that MCM7 is a critical component of DNA replication licensing complex in the yeast and xenopus.1–4 Some studies suggest that MCM4, MCM6, and MCM7 complex contains DNA helicase activity.5,6 DNA replication licensing complex is multimeric and phase specific. In yeast, DNA replication licensing proteins, such as MCM2-7 and several replication origin binding proteins, such as Cdc6, germinin, and Cdt1, form DNA replication licensing complex in G1 phase to enable DNA replication and to promote cell cycle entry into S phase. Initial implication of MCM7 involvement in human malignancies came from positive immunostaining of MCM7 in several human malignancies, including endometrial carcinoma,7 melanoma,8 esophageal adenocarcinoma,9 colorectal adenocarcinoma,10 oral squamous cell carcinoma,11 glioblastoma,12 and thyroid cancer.13 The first study addressing the oncogenic role of MCM7 in prostate cancer came from genome analysis of prostate cancer by performing a genome wide copy number analysis using biotin-labeled genome DNA on Affymetrix U95av2 chip.14 The DNA copy number of MCM7 was found to increase severalfold accompanied with a concomitant increase of MCM7 mRNA level. Subsequent validation analyses suggest that either copy number and/or protein level increase of MCM7 are associated with prostate cancer relapse and metastasis. Amplification of MCM7 was also found in esophageal carcinoma.9 The magnitude of MCM7 amplification correlates with the expression of MCM7, tumor grades, and the aggressiveness of esophageal cancer.9 It is presumed that amplification of MCM7 is the driving force of MCM7 overexpression in primary human malignancies. MCM7 is probably the primary target of Rb, the tumor suppressor that controls cell entry into S phase.15 There is growing evidence that other signaling pathways also regulate MCM7 activity.Receptor for activated protein kinase C 1 (RACK1), was initially identified as an adaptor of several protein kinase C (PKC) isoforms.16 The binding of RACK1 and PKC anchor PKC to its substrate to initiate second messenger signaling. It is suggested, according to recent studies that RACK1 interacts with a variety of other signaling molecules, including ras-GTPase activating protein,17 dynamin-1,18 src,19 integrins,20 PTPμ,21 phosphodiesterase,22 hypoxia induced factor-1,23 and so forth, that play an important role in several physiological processes, including, growth, hypoxia response, migration, adhesion, and cell differentiation. RACK1 only binds PKC activated by diacylglycerol or phorbol ester, but not quiescent PKC. In this study, we showed that RACK1 binds with MCM7 N-terminus. The MCM7/RACK1 interaction appears essential for DNA replication activity of MCM7.  相似文献   

15.
16.
Systemic sclerosis results in tissue fibrosis due to the activation of fibroblasts and the ensuing overproduction of the extracellular matrix. We previously reported that the absence of α2-antiplasmin (α2AP) attenuated the process of dermal fibrosis; however, the detailed mechanism of how α2AP affects the progression of fibrosis remained unclear. The goal of the present study was to examine the role of α2AP in fibrotic change. We observed significantly higher levels of α2AP expression in the skin of bleomycin-injected systemic sclerosis model mice in comparison with the levels seen in control mice. We also demonstrated that α2AP induced myofibroblast differentiation, and the absence of α2AP attenuated the induction of myofibroblast differentiation. Moreover, we found that connective tissue growth factor induced the expression of α2AP through both the extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways in fibroblasts. Interestingly, α2AP also induced transforming growth factor-β expression through the same pathways, and the inhibition of ERK1/2 and JNK slowed the progression of bleomycin-induced fibrosis. Our findings suggest that α2AP is associated with the progression of fibrosis, and regulation of α2AP expression by the ERK1/2 and JNK pathways may be an effective antifibrotic therapy for the treatment of systemic sclerosis.Systemic sclerosis (SSc) affects the skin and the internal organs, resulting in tissue fibrosis. Although the disease process involves immunological mechanisms, vascular damage, and activation of fibroblasts, the pathogenesis of SSc remains to be further elucidated. Fibrotic diseases are characterized by excessive scarring due to excessive production, deposition, and contraction of the extracellular matrix (ECM). This process usually occurs over many months and years, and can lead to organ dysfunction or death. Connective tissue growth factor (CTGF) is constitutively overexpressed in fibrotic lesions such as in scleroderma,1 liver,2 renal,3,4 lung,5 and pancreatic fibrosis.5 CTGF acts as a downstream effecter of at least some of the profibrotic effects of transforming growth factor-β (TGF-ß),6 and promotes fibroblast proliferation, myofibroblasts differentiation, matrix production, and granulation tissue formation.7,8Human and murine α2-antiplasmin (α2AP) are serpins (serine protease inhibitors) with a molecular weight of 65 to 70 kd,9 which rapidly inactivate plasmin, resulting in the formation of a stable inactive complex, plasmin-α2AP.10 Tissue fibrosis is generally considered to arise due to a failure of the normal wound healing response to terminate.11 Previous our studies show that α2AP is associated with the wound healing and the fibrosis.12,13 In addition, it has been reported that the level of plasmin-α2AP complex in plasma is elevated in SSc patients.14 These findings suggest that α2AP may be associated with the progression of fibrotic disease, but the physiological roles of α2AP are not precisely understood. We herein report that α2AP plays an important role in the progression of fibrosis.  相似文献   

17.
Cancer stem-like cell subpopulations, referred to as “side-population” (SP) cells, have been identified in several tumors based on their ability to efflux the fluorescent dye Hoechst 33342. Although SP cells have been identified in the normal human endometrium and endometrial cancer, little is known about their characteristics. In this study, we isolated and characterized the SP cells in human endometrial cancer cells and in rat endometrial cells expressing oncogenic human K-Ras protein. These SP cells showed i) reduction in the expression levels of differentiation markers; ii) long-term proliferative capacity of the cell cultures; iii) self-renewal capacity in vitro; iv) enhancement of migration, lamellipodia, and, uropodia formation; and v) enhanced tumorigenicity. In nude mice, SP cells formed large, invasive tumors, which were composed of both tumor cells and stromal-like cells with enriched extracellular matrix. The expression levels of vimentin, α-smooth muscle actin, and collagen III were enhanced in SP tumors compared with the levels in non-SP tumors. In addition, analysis of microdissected samples and fluorescence in situ hybridization of Hec1-SP-tumors showed that the stromal-like cells with enriched extracellular matrix contained human DNA, confirming that the stromal-like cells were derived from the inoculated cells. Moreober, in a Matrigel assay, SP cells differentiated into α-smooth muscle actin-expressing cells. These findings demonstrate that SP cells have cancer stem-like cell features, including the potential to differentiate into the mesenchymal cell lineage.Recently, adult stem cells have been identified in several mature tissues, such as the adult intestine,1 skin,2 muscle,3 blood,4 and the nervous system5–7 A stem cell is an undifferentiated cell that is defined by its ability to both self-renew and to produce mature progeny cells.8 Stem cells are classified based on their developmental potential as totipotent, pluripotent, oligopotent, and unipotent. Adult somatic stem cells were originally thought to be tissue specific and only able to give rise to progeny cells corresponding to their tissue of origin. Recent studies, however, have shown that adult mammalian stem cells are able to differentiate across tissue lineage boundaries,9,10 although this “plasticity” of adult somatic stem cells remains controversial.Stem cell subpopulations (“side-population” (SP) cells) have been identified in many mammals, including humans, based on the ability of these cells to efflux the fluorescent dye Hoechst 33342.11 Recent evidence suggests that the SP phenotype is associated with a high expression level of the ATP-binding cassette transporter protein ABCG2/Bcrp1.12 Most recently, established malignant cell lines, which have been maintained for many years in culture, have also been shown to contain SP cells as a minor subpopulation.13The human endometrium is a highly dynamic tissue undergoing cycles of growth, differentiation, shedding, and regeneration throughout the reproductive life of women. Endometrial adult stem/progenitor cells are likely responsible for endometrial regeneration.14 Rare populations of human endometrial epithelial and stromal colony-forming cells15 and SP cells16,17 have been identified. Although coexpression of CD146 and PDGFRβ isolates a population of mesenchymal stem like cells from human endometrium,18 specific stem cell markers of endometrium remain unclear. Recently, Gotte et al19 demonstrated that the adult stem cell marker Musashi-1 was coexpressed with Notch-1 in a subpopulation of endometrial cells. Furthermore, they showed that telomerase and Musashi-1-expressing cells were significantly increased in proliferative endometrium, endometriosis, and endometrial carcinoma tissue, compared with secretary endometrium, suggesting the concept of a stem cell origin of endometriosis and endometrial carcinoma.Recent evidence suggests that cancer stem-like cells exist in several malignant tumors, such as leukemia20,21 breast cancer,22 and brain tumors,23 and that these stem cells express surface markers similar to those expressed by normal stem cells in each tissue.20,24Development of endometrial carcinoma is associated with a variety of genetic alterations. For example, increased expression and activity of telomerase25,26 and frequent dysregulation of signaling pathways have been observed in endometrial carcinoma. Some of these pathways are important determinants of stem cell activity (Wnt-β-catenin and PTEN).27–29 These suggest a stem cell contribution to endometrial carcinoma development.Recently, we isolated SP cells from the human endometrium. These SP cells showed long-term proliferating capacity in cultures and produced both gland and stromal-like cells. Additionally, they were able to function as progenitor cells.16 In this study, we isolated and characterized SP cells from human endometrial cancer cells and from rat endometrial cells expressing oncogenic [12Val] human K-Ras protein and demonstrated their cancer stem-like cell phenotypes.  相似文献   

18.
Colony-stimulating factor (CSF)-1 controls the survival, proliferation, and differentiation of macrophages, which are recognized as scavengers and agents of the innate and the acquired immune systems. Because of their plasticity, macrophages are endowed with many other essential roles during development and tissue homeostasis. We present evidence that CSF-1 plays an important trophic role in postnatal organ growth and kidney repair. Notably, the injection of CSF-1 postnatally enhanced kidney weight and volume and was associated with increased numbers of tissue macrophages. Moreover, CSF-1 promotes postnatal renal repair in mice after ischemia-reperfusion injury by recruiting and influencing macrophages toward a reparative state. CSF-1 treatment rapidly accelerated renal repair with tubular epithelial cell replacement, attenuation of interstitial fibrosis, and functional recovery. Analysis of macrophages from CSF-1-treated kidneys showed increased expression of insulin-like growth factor-1 and anti-inflammatory genes that are known CSF-1 targets. Taken together, these data suggest that CSF-1 is important in kidney growth and the promotion of endogenous repair and resolution of inflammatory injury.Macrophages are versatile cells that have been long recognized as immune effectors where their recruitment to sites of injury is a fundamental feature of inflammation. Although their role in host defense has been well documented, macrophages and their precursors are also important during embryogenesis, normal tissue maintenance, and postnatal organ repair.1,2 Almost all developing organs contain a population of resident monocytes that infiltrate very early during organogenesis and persist throughout adult life.3–6 In addition to their phagocytic capabilities during tissue remodeling-associated apoptosis,5,7 fetal macrophages have many trophic effects that promote tissue and organ growth.6,8,9Colony-stimulating factor (CSF)-1 controls the differentiation, proliferation, and survival of macrophages by binding to a high-affinity cell-surface tyrosine kinase receptor (CSF-1R), encoded by the c-fms proto-oncogene that is expressed on macrophages and their progenitors.6 CSF-1 is critical for both adult and embryonic macrophage development. This is manifested by multiple organ growth deficiencies observed in osteopetrotic (Csf1op/Csf1op) mice that have a spontaneous mutation in the csf-1 gene. These mice show growth restriction and developmental abnormalities of the bones, brain, and reproductive and endocrine organs,10–13 a phenotype that can be rescued by injection of exogenous CSF-1 or insertion of a csf-1 transgene.14–16In adult organs, there is considerable heterogeneity of monocytes and macrophages with distinct subsets defined by phenotype, function, and the differential expression of cell surface markers.17–19 Subpopulations of macrophages directly contribute to wound healing and tissue repair, supporting the concept that some macrophage phenotypes can promote organ regeneration after a pro-inflammatory state of injury.20 The concept of macrophage polarization states has emerged; the M1 “classically activated” pro-inflammatory cell type apparently opposed by an M2 “alternatively activated” immune regulatory macrophage.18 In general, these two states are thought to be analogous to the opposing T helper 1 and T helper 2 immune responses, although in both cases this model is probably too simplistic. Functionally, it is more likely that distinct subpopulations of macrophages may exist in the same tissue and play critical roles in both the injury and recovery phases of inflammatory scarring.20Our previous study provided evidence that the addition of CSF-1 to a developing murine kidney promotes a growth and differentiation response that is accompanied by increased numbers of macrophages.3 Furthermore, with the use of expression profiling we demonstrated that fetal kidney, lung, and brain macrophages share a characteristic gene expression profile that includes the production of factors important in the suppression of inflammation and the promotion of proliferation.3 Embryonic macrophages appear to play a positive trophic role that may have parallel reparative functions in many adult tissues undergoing repair and cellular replacement.1,20 A number of studies have suggested that infiltrating macrophages along with the trophic factors they release participate in tissue repair of the kidney,20–22 brain,23 skin,24,25 lung,26 liver,27 heart,28 gastrointestinal tract,29,30 and skeletal muscle.31,32 Indeed, the pleiotrophic roles for CSF-1 in reproduction, development of multiple organ systems, and maternal-fetal interactions during pregnancy by macrophage-mediated processes have also been well defined.2,33,34To determine the physiological relevance of CSF-1 as a component of the mammalian growth regulatory axis, CSF-1 was administered to neonatal mice. We report that CSF-1 administration to newborn mice increased body weight and kidney weight and volume and was associated with increased numbers of macrophages. Our results also establish that CSF-1 injection into mice after ischemia-reperfusion (IR) injury promoted endogenous repair with characteristic rapid re-epithelialization of the damaged tubular epithelium, leading to functional recovery. Flow cytometric and gene expression analyses were used to delineate the macrophage profile present in the kidneys during the early and resolution phase of IR injury with and without CSF-1 therapy. We thus provide evidence that CSF-1 recruits macrophages to the reparative site and influences their phenotype, partly through an insulin-like growth factor (IGF)-1 signaling response. Therefore, macrophages under the stimulus of CSF-1 in an acute setting of renal disease markedly accelerate renal cell replacement and tissue remodeling while attenuating downstream interstitial extracellular matrix accumulation.  相似文献   

19.
20.
Although various guidelines and position papers have discussed, in the past, the ethical aspects of genetic testing in asymptomatic minors, the European Society of Human Genetics had not earlier endorsed any set of guidelines exclusively focused on this issue. This paper has served as a background document in preparation of the development of the policy recommendations of the Public and Professional Committee of the European Society of Human Genetics. This background paper first discusses some general considerations with regard to the provision of genetic tests to minors. It discusses the concept of best interests, participation of minors in health-care decisions, parents'' responsibilities to share genetic information, the role of clinical genetics and the health-care system in communication within the family. Second, it discusses, respectively, the presymptomatic and predictive genetic testing for adult-onset disorders, childhood-onset disorders and carrier testing.Although various guidelines and position papers have discussed, in the past, the ethical aspects of genetic testing in asymptomatic minors,1, 2 the European Society of Human Genetics had not earlier endorsed any set of guidelines exclusively focused on this issue. This background paper was preceded by an in-depth research on the topic by Eurogentest.3 Eurogentest (http://www.eurogentest.org aims to develop the necessary infrastructure, tools, resources, guidelines and procedures that will structure, harmonize and improve the overall quality of all the EU genetic services at the molecular, cytogenetic, biochemical and clinical level.4 Attention has also been paid to the provision of appropriate counselling related to genetic testing, the education of patients and professionals, as well as to the ethical, legal and social issues surrounding testing. The focus of the ethics unit of Eurogentest was oriented towards the study of the ethical issues related to genetic testing in minors. This work was the starting point for this background paper, which has been prepared and supported by different types of evidence. First, research has been performed on the existing recommendations regarding predictive genetic testing in minors1 and carrier testing,2 with the intention of identifying areas of agreement and disagreement. Second, the literature on medico–ethical and medico–legal aspects of predictive genetic testing in minors,5 carrier testing,6, 7 the position of minors8 and patient rights9 was studied. Third, a systematic literature review was performed to gather information regarding the attitudes of the different stakeholders (minors, health-care professionals, parents and relatives of the affected individuals) towards genetic testing in asymptomatic minors.10, 11 Fourth, the attitudes of European clinical geneticists regarding genetic testing in asymptomatic minors were gathered.12, 13, 14In 2007, contacts were made with the Public and Professional Policy Committee of the European Society of Human Genetics with the aim of developing policy recommendations on the issue. On the basis of a decision of the PPPC meeting during the ESHG conference in Nice (June 2007), an ad hoc committee, consisting of Pascal Borry (Eurogentest), Kris Dierickx (Eurogentest), Angus Clarke, Gerry Evers-Kiebooms (PPPC) and Martina Cornel (PPPC), was created. This ad hoc committee met on 15 November 2007 to discuss a first draft of a background paper and recommendations that were prepared by Pascal Borry under the supervision of Kris Dierickx. A revised version was discussed during a PPPC meeting in Amsterdam (April 2008) and Barcelona (June 2008). In order not to repeat issues that have been discussed elsewhere, reference will often be made to the above-referenced publications.  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