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.
Rosacea is a chronic inflammatory condition of facial skin estimated to affect more than 16 million Americans. Although the pathogenesis of rosacea is not fully understood, recent evidence in vitro as well as in vivo has supported the role of increased levels of the trypsin-like serine protease, kallikrein 5, in initiating an augmented inflammatory response in rosacea. The increase in the quantity and magnitude of biological activity of kallikrein 5 leads to production of greater quantities of cathelicidin (LL-37), an antimicrobial peptide associated with increases in innate cutaneous inflammation, vasodilation, and vascular proliferation, all of which are characteristic features of rosacea. In this article, the authors review the literature supporting the role of kallikrein 5 in the pathophysiology of rosacea, including how therapeutic interventions modulate the effects of kallikrein 5, thus providing further support for this pathophysiological model that at least partially explains many of the clinical features of cutaneous rosacea.Cutaneous rosacea (rosacea) is a chronic inflammatory facial skin disorder noted most commonly in individuals of northern European descent, although people of any ethnicity or skin color may be affected.1-4 The visible manifestations with central facial predominance are characteristic of rosacea, including erythema, papules, pustules, telangiectasias, and phymatous changes.1-4 However, persistent (nontransient) erythema involving the central face that intensifies during flares and the presence of telangiectasias, which are also accentuated mostly on the central face, are the core clinical features that support a diagnosis of rosacea.1-9 Papules and pustules are not consistently present in rosacea, characterizing only those individuals with rosacea who exhibit the papulopustular subtype of the disease.3-6 In fact, papulopustular lesions never emerge in many individuals affected by rosacesa, and phymatous changes affect only a relatively small number of the rosacea-affected population; however, central facial erythema is present to some extent in essentially all people with rosacea.1-8Why do some people get rosacea and others do not? Although the entire explanation that would fully answer this question remains elusive, current evidence suggests that individuals affected by rosacea exhibit rosacea-prone skin, which inherently displays dysregulation of two main systems present within skin—the neurovascular/neuroimmune system and the immune detection/response system (innate immunity).3,5-8,19 Both of these systems normally serve physiological functions related to how skin responds to exogenous changes or insults (i.e., changes in temperature, exposure to microbial pathogens). However, in rosacea, both the cutaneous neurovascular/neuroimmune system and the immune detection/response system are dysregulated, with both demonstrating augmented responses that correlate with clinical manifestations commonly seen in patients with cutaneous rosacea.Neurovascular/neuroimmune dysregulation, which includes both anatomic and physiochemical differences present in rosacea-prone skin as compared to healthy facial skin, appears to be a major contributor that exacerbates the vasodilation of facial skin vasculature with increased facial blood flow that occurs during a rosacea flare.3,5-7,17,18,20 This increased vasodilation in rosacea-affected skin, which can be acute or subacute in onset, is commonly referred to as flushing.1,3,4,6,7,17,20 Neurosensory symptoms (i.e., stinging, burning) are often associated with or exacerbated during a rosacea flare.1,3-8 Exogenous factors that are commonly recognized by patients as triggers, which seem to induce a flare, include increased ambient heat/warmth and certain spices (i.e., capsaicin), all of which can induce signaling of neurogenic inflammation via specific receptor channels (transient receptor potential vanilloid [TRPV] subfamily) shown to be increased in rosacea-prone skin.3,6,17,18 The immune detection/response dysregulation of rosacea is evidenced by the upregulation of the pattern recognition receptor, toll-like receptor 2 (TLR2) and the cathelicidin innate immunity pathway.3,5-17,19,21,22 Ultraviolet light (UV) exposure, another recognized trigger factor associated with flares of rosacea, produces changes that induce ligand-binding of TLR2, which signals innate inflammation.3,5-16,19,21 Lastly, upregulated production of several matrix metalloproteases (MMPs) has been demonstrated in rosacea, further contributing to cascades of inflammation and degradation of the dermal matrix.1,3,5-7,19 Accentuated immune detection/response as a major component of the pathophysiology of rosacea has been discussed extensively in the literature and is addressed in more detail as a major subject of this article.5-7,10-16,19,21,22Although the pathophysiology of rosacea is not completely understood, dysregulation of the innate immune detection/response system plays a significant role in the inflammatory and vascular responses seen in this condition.5-7,10-16,19,21,22 As a known inducer of innate and cellular inflammation, increased vascularity, and angiogenesis, cathelicidin (LL-37), an antimicrobial peptide that physiologically provides near-immediate innate defense against several microbial organisms, has been investigated to determine its potential role in the pathophysiology of rosacea.10,11,23,24 Results have shown that patients with rosacea express elevated levels of LL-37 in facial skin, with this increased expression attributed to abnormally high levels of the trypsin-like serine protease enzyme, kallikrein 5 (KLK5), which selectively cleaves an inactive precursor protein (hCAP18) to form the biologically active antimicrobial peptide (LL-37).10,22 Investigations of the mechanism of action of two agents proven to be effective in reducing papulopustular lesions and perilesional erythema in rosacea, topical azelaic acid (AzA) and oral doxycycline, demonstrated direct and indirect inhibition of KLK5, respectively.25-29 In one study with AzA 15% gel, the reduction in KLK5 activity correlated with clinical improvement of rosacea.29 In this review, the authors further describe the role of KLK5 in the pathophysiology of rosacea, including the inflammatory cascades that result from increased KLK5 expression, as well as a more detailed discussion of different therapies shown to inhibit the progression of this cascade.  相似文献   

2.
Cutaneous collagenous vasculopathy is a rare microangiopathy of dermal blood vessels. Clinically indistinguishable from generalized essential telangiectasia, this condition is diagnosed by its unique histological appearance. In contrast to other primary telangiectatic processes, cutaneous collagenous vasculopathy has dilated vascular structures that contain deposits of eosinophilic hyaline material within the vessel walls. To date, cutaneous collagenous vasculopathy has been described in a total of 19 cases in the medical literature. The first several cases were described exclusively in middle-aged to elderly men. Though it has now been described in both men and women, cutaneous collagenous vasculopathy is still most often described in middle-aged to older adults. No particular disease or medication has been linked to the development of cutaneous collagenous vasculopathy, and the etiology remains unknown. In this case series, the authors present three additional patients diagnosed with cutaneous collagenous vasculopathy and discuss their clinical and histopathologic features.Cutaneous collagenous vasculopathy (CCV) is a rare, idiopathic microangiopathy first reported in 2000 by Salama and Rosenthal.1 CCV has characteristic microscopic findings, including dilated capillaries and post-capillary venules with marked collagen deposition,2 which are features essential to diagnosis. Clinically, CCV presents as blanchable, non-urticating macules that typically begin on the lower extremities and then spread to the trunk and upper extremities. 1,3-6 Due to its clinical similarity to generalized essential telangiectasia (GET), dermatologists may not biopsy these patients, potentially causing CCV to be underdiagnosed and under-reported. 4,5,7,8 To date, CCV has been described in approximately equal numbers in men and women of Caucasian race with patients ranging from 16 to 83 years of age.4,9 The majority of cases have been diagnosed in patients with other concomitant diseases, most commonly hypertension and cardiovascular disease,3,5,6,9,10 but also in patients with autoimmune conditions2,5,8,9 and diabetes mellitus.5,10 Additionally, in most described cases, patients were taking at least one medication on an intermittent or ongoing basis. 1,2,4-7,9,1° Despite this observation, specific medical conditions or medications are yet to be linked to the development of CCV. In this article, the authors present three female patients who have been diagnosed with CCV along with their clinical and histopathological features.  相似文献   

3.
Tear trough deformities are a sign of facial aging. The anatomical base is well understood. In many patients, minimal invasive surgical procedures are useful to improve appearance. Here, the authors describe the use of monophasic hyaluronic acid dermal filler and calcium hydroxylapatite injection for correction. Forty female patients with a mean age of 50 years have been treated. On average, an improvement of one class of Hidman’s severity score could be achieved by single treatment. Mean duration of the effect was 10.1 months for hyaluronic acid and 12.8 months for calcium hydroxylapatite. Adverse effects were mild and temporary. Patients satisfaction was high (95%).Considering facial aesthetics, the periorbital region is one of the most important regions. Facial aging and loss of volume can be recognized easily due to the delicate structure of periorbital skin and subcutaneous tissue. Tear trough or nasojugal groove describes the concavity at the border of the lower eyelid and medial cheek. This hollow area elicits a tired or wearied appearance. The shadow created by this groove is commonly perceived as dark circles.1The tear trough represents also a border between loose tissue of the lower lid and more compact structures of the cheek. This line or border is defined by an anchoring fascia (septum orbitale) to the periosteum of the inferomedial arcus marginalis. This leads to a separation of adjacent fat compartments of the face—inferior orbital from medial cheek and nasolabial fat pads. In contrast to other prominent facial folds, there is an additional structure separating superficial and deep fat pads—the orbicularis retaining ligament.1 This ligament is stronger in the medial portion, but weaker in the central part. With age, a further weakening can lead to herniation of orbital fat.Recently, Wong et al2 succeeded in isolation and characterization of the tear trough ligament. This is an osteocutaneous ligament found on the maxilla between the palpebral and the orbital parts of the musculus orbicularis. It commences medially just inferior to the anterior lacrimal crest and continues laterally as orbicularis retaining ligament.2In the infraorbital region, the superficial musculo-aponeurotic system (SMAS) consists of a network of small fibrous septae, which traverse perpendicularly between fat lobules to the dermis and deeply into facial muscles or periosteum. Loss of maxillary bone projection negatively affects appearance with increasing age. Severity of tear trough deformity correlates with the anterior angle of the anterior face of maxillary sinus.3,4 Other factors contributing to an aged appearance are loss of skin elasticity, festoons, and musculus orbicularis prominence. The loss of volume indirectly contributes to the tear trough deformity,5,6 which can be associated with lower lid eye bags. The major factors contributing to tear trough deformity can be described by the trinity of relaxation, atrophy, and ptosis. This makes the correction of tear trough deformities one of the most challenging topics for minimal invasive facial aesthetic procedures.7,8Although various surgical techniques have been developed to correct tear trough deformity, non-surgical techniques have gained a wider acceptance by both patients and medical doctors.9,10  相似文献   

4.

OBJECTIVE

We investigated the effects of 18 confirmed type 2 diabetes risk single nucleotide polymorphisms (SNPs) on insulin sensitivity, insulin secretion, and conversion of proinsulin to insulin.

RESEARCH DESIGN AND METHODS

A total of 5,327 nondiabetic men (age 58 ± 7 years, BMI 27.0 ± 3.8 kg/m2) from a large population-based cohort were included. Oral glucose tolerance tests and genotyping of SNPs in or near PPARG, KCNJ11, TCF7L2, SLC30A8, HHEX, LOC387761, CDKN2B, IGF2BP2, CDKAL1, HNF1B, WFS1, JAZF1, CDC123, TSPAN8, THADA, ADAMTS9, NOTCH2, KCNQ1, and MTNR1B were performed. HNF1B rs757210 was excluded because of failure to achieve Hardy-Weinberg equilibrium.

RESULTS

Six SNPs (TCF7L2, SLC30A8, HHEX, CDKN2B, CDKAL1, and MTNR1B) were significantly (P < 6.9 × 10−4) and two SNPs (KCNJ11 and IGF2BP2) were nominally (P < 0.05) associated with early-phase insulin release (InsAUC0–30/GluAUC0–30), adjusted for age, BMI, and insulin sensitivity (Matsuda ISI). Combined effects of these eight SNPs reached −32% reduction in InsAUC0–30/GluAUC0–30 in carriers of ≥11 vs. ≤3 weighted risk alleles. Four SNPs (SLC30A8, HHEX, CDKAL1, and TCF7L2) were significantly or nominally associated with indexes of proinsulin conversion. Three SNPs (KCNJ11, HHEX, and TSPAN8) were nominally associated with Matsuda ISI (adjusted for age and BMI). The effect of HHEX on Matsuda ISI became significant after additional adjustment for InsAUC0–30/GluAUC0–30. Nine SNPs did not show any associations with examined traits.

CONCLUSIONS

Eight type 2 diabetes–related loci were significantly or nominally associated with impaired early-phase insulin release. Effects of SLC30A8, HHEX, CDKAL1, and TCF7L2 on insulin release could be partially explained by impaired proinsulin conversion. HHEX might influence both insulin release and insulin sensitivity.Impaired insulin secretion and insulin resistance, two main pathophysiological mechanisms leading to type 2 diabetes, have a significant genetic component (1). Recent studies have confirmed 20 genetic loci reproducibly associated with type 2 diabetes (213). Three were previously known (PPARG, KCNJ11, and TCF7L2), whereas 17 loci were recently discovered either by genome-wide association studies (SLC30A8, HHEX-IDE, LOC387761, CDKN2A/2B, IGF2BP2, CDKAL1, FTO, JAZF1, CDC123/CAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, NOTCH2, KCNQ1, and MTNR1B), or candidate gene approach (WFS1 and HNF1B). The mechanisms by which these genes contribute to the development of type 2 diabetes are not fully understood.PPARG is the only gene from the 20 confirmed loci previously associated with insulin sensitivity (14,15). Association with impaired β-cell function has been reported for 14 loci (KCNJ11, SLC30A8, HHEX-IDE, CDKN2A/2B, IGF2BP2, CDKAL1, TCF7L2, WFS1, HNF1B, JAZF1, CDC123/CAMK1D, TSPAN8/LGR5, KCNQ1, and MTNR1B) (6,12,13,1638). Although associations of variants in HHEX (1622), CDKAL1 (6,2126), TCF7L2 (22,2730), and MTNR1B (13,31,32) with impaired insulin secretion seem to be consistent across different studies, information concerning other genes is limited (12,1825,27,3338). The mechanisms by which variants in these genes affect insulin secretion are unknown. However, a few recent studies suggested that variants in TCF7L2 (22,3942), SLC30A8 (22), CDKAL1 (22), and MTNR1B (31) might influence insulin secretion by affecting the conversion of proinsulin to insulin. Variants of FTO have been shown to confer risk for type 2 diabetes through their association with obesity (7,16) and therefore were not included in this study.Large population-based studies can help to elucidate the underlying mechanisms by which single nucleotide polymorphisms (SNPs) of different risk genes predispose to type 2 diabetes. Therefore, we investigated confirmed type 2 diabetes–related loci for their associations with insulin sensitivity, insulin secretion, and conversion of proinsulin to insulin in a population-based sample of 5,327 nondiabetic Finnish men.  相似文献   

5.

OBJECTIVE

Oxyntomodulin (OXM) is a glucagon-like peptide 1 (GLP-1) receptor (GLP1R)/glucagon receptor (GCGR) dual agonist peptide that reduces body weight in obese subjects through increased energy expenditure and decreased energy intake. The metabolic effects of OXM have been attributed primarily to GLP1R agonism. We examined whether a long acting GLP1R/GCGR dual agonist peptide exerts metabolic effects in diet-induced obese mice that are distinct from those obtained with a GLP1R-selective agonist.

RESEARCH DESIGN AND METHODS

We developed a protease-resistant dual GLP1R/GCGR agonist, DualAG, and a corresponding GLP1R-selective agonist, GLPAG, matched for GLP1R agonist potency and pharmacokinetics. The metabolic effects of these two peptides with respect to weight loss, caloric reduction, glucose control, and lipid lowering, were compared upon chronic dosing in diet-induced obese (DIO) mice. Acute studies in DIO mice revealed metabolic pathways that were modulated independent of weight loss. Studies in Glp1r−/− and Gcgr−/− mice enabled delineation of the contribution of GLP1R versus GCGR activation to the pharmacology of DualAG.

RESULTS

Peptide DualAG exhibits superior weight loss, lipid-lowering activity, and antihyperglycemic efficacy comparable to GLPAG. Improvements in plasma metabolic parameters including insulin, leptin, and adiponectin were more pronounced upon chronic treatment with DualAG than with GLPAG. Dual receptor agonism also increased fatty acid oxidation and reduced hepatic steatosis in DIO mice. The antiobesity effects of DualAG require activation of both GLP1R and GCGR.

CONCLUSIONS

Sustained GLP1R/GCGR dual agonism reverses obesity in DIO mice and is a novel therapeutic approach to the treatment of obesity.Obesity is an important risk factor for type 2 diabetes, and ∼90% of patients with type 2 diabetes are overweight or obese (1). Among new therapies for type 2 diabetes, peptidyl mimetics of the gut-derived incretin hormone glucagon-like peptide 1 (GLP-1) stimulate insulin biosynthesis and secretion in a glucose-dependent manner (2,3) and cause modest weight loss in type 2 diabetic patients. The glucose-lowering and antiobesity effects of incretin-based therapies for type 2 diabetes have prompted evaluation of the therapeutic potential of other glucagon-family peptides, in particular oxyntomodulin (OXM). The OXM peptide is generated by post-translational processing of preproglucagon in the gut and is secreted postprandially from l-cells of the jejuno-ileum together with other preproglucagon-derived peptides including GLP-1 (4,5). In rodents, OXM reduces food intake and body weight, increases energy expenditure, and improves glucose metabolism (68). A 4-week clinical study in obese subjects demonstrated that repeated subcutaneous administration of OXM was well tolerated and caused significant weight loss with a concomitant reduction in food intake (9). An increase in activity-related energy expenditure was also noted in a separate study involving short-term treatment with the peptide (10).OXM activates both, the GLP-1 receptor (GLP1R) and glucagon receptor (GCGR) in vitro, albeit with 10- to 100-fold reduced potency compared with the cognate ligands GLP-1 and glucagon, respectively (1113). It has been proposed that OXM modulates glucose and energy homeostasis solely by GLP1R agonism, because its acute metabolic effects in rodents are abolished by coadministration of the GLP1R antagonist exendin(939) and are not observed in Glp1r−/− mice (7,8,14,15). Other aspects of OXM pharmacology, however, such as protective effects on murine islets and inhibition of gastric acid secretion appear to be independent of GLP1R signaling (14). In addition, pharmacological activation of GCGR by glucagon, a master regulator of fasting metabolism (16), decreases food intake in rodents and humans (1719), suggesting a potential role for GCGR signaling in the pharmacology of OXM. Because both OXM and GLP-1 are labile in vivo (T1/2 ∼12 min and 2–3 min, respectively) (20,21) and are substrates for the cell surface protease dipeptidyl peptidase 4 (DPP-4) (22), we developed two long-acting DPP-4–resistant OXM analogs as pharmacological agents to better investigate the differential pharmacology and therapeutic potential of dual GLP1R/GCGR agonism versus GLP1R-selective agonism. Peptide DualAG exhibits in vitro GLP1R and GCGR agonist potency comparable to that of native OXM and is conjugated to cholesterol via a Cys sidechain at the C-terminus for improved pharmacokinetics. Peptide GLPAG differs from DualAG by only one residue (Gln3→Glu) and is an equipotent GLP1R agonist, but has no significant GCGR agonist or antagonist activity in vitro. The objective of this study was to leverage the matched GLP1R agonist potencies and pharmacokinetics of peptides DualAG and GLPAG in comparing the metabolic effects and therapeutic potential of a dual GLP1R/GCGR agonist with a GLP1R-selective agonist in a mouse model of obesity.  相似文献   

6.

OBJECTIVE

Significant new data suggest that metabolic disorders such as diabetes, obesity, and atherosclerosis all posses an important inflammatory component. Infiltrating macrophages contribute to both tissue-specific and systemic inflammation, which promotes insulin resistance. The complement cascade is involved in the inflammatory cascade initiated by the innate and adaptive immune response. A mouse genomic F2 cross biology was performed and identified several causal genes linked to type 2 diabetes, including the complement pathway.

RESEARCH DESIGN AND METHODS

We therefore sought to investigate the effect of a C3a receptor (C3aR) deletion on insulin resistance, obesity, and macrophage function utilizing both the normal-diet (ND) and a diet-induced obesity mouse model.

RESULTS

We demonstrate that high C3aR expression is found in white adipose tissue and increases upon high-fat diet (HFD) feeding. Both adipocytes and macrophages within the white adipose tissue express significant amounts of C3aR. C3aR−/− mice on HFD are transiently resistant to diet-induced obesity during an 8-week period. Metabolic profiling suggests that they are also protected from HFD-induced insulin resistance and liver steatosis. C3aR−/− mice had improved insulin sensitivity on both ND and HFD as seen by an insulin tolerance test and an oral glucose tolerance test. Adipose tissue analysis revealed a striking decrease in macrophage infiltration with a concomitant reduction in both tissue and plasma proinflammatory cytokine production. Furthermore, C3aR−/− macrophages polarized to the M1 phenotype showed a considerable decrease in proinflammatory mediators.

CONCLUSIONS

Overall, our results suggest that the C3aR in macrophages, and potentially adipocytes, plays an important role in adipose tissue homeostasis and insulin resistance.The complement system is an integral part of both the innate and adaptive immune response involved in the defense against invading pathogens (1). Complement activation culminates in a massive amplification of the immune response leading to increased cell lysis, phagocytosis, and inflammation (1). C3 is the most abundant component of the complement cascade and the convergent point of all three major complement activation pathways. C3 is cleaved into C3a and C3b by the classical and lectin pathways, and iC3b is generated by the alternative pathway (2,3). C3a has potent anaphylatoxin activity, directly triggering degranulation of mast cells, inflammation, chemotaxis, activation of leukocytes, as well as increasing vascular permeability and smooth muscle contraction (3). C3a mediates its downstream signaling effects by binding to the C3a receptor (C3aR), a Gi-coupled G protein–coupled receptor. Several studies have demonstrated a role for C3a and C3aR in asthma, sepsis, liver regeneration, and autoimmune encephalomyelitis (1,3). Therefore, targeting C3aR may be an attractive therapeutic option for the treatment of several inflammatory diseases.Increasing literature suggests that metabolic disorders such as diabetes, obesity, and atherosclerosis also possess an important inflammatory component (47). Several seminal reports have demonstrated that resident macrophages can constitute as much as 40% of the cell population of adipose tissue (79) and can significantly affect insulin resistance (1018). Several proinflammatory cytokines, growth factors, acute-phase proteins, and hormones are produced by the adipose tissue and implicated in insulin resistance and vascular homeostasis (47,19). An integrated genomics approach was performed with several mouse strains to infer causal relationships between gene expression and complex genetic diseases such as obesity/diabetes. This approach identified the C3aR gene as being causal for omental fat pad mass (20). The C3aR−/− mice were shown to have decreased adiposity as compared with wild-type mice on a regular diet (20). Monocytes and macrophages express the C3aR (2128). Increased C3a levels also correlate with obesity, diabetes, cholesterol, and lipid levels (2934). We therefore sought to investigate the specific role of the C3aR in insulin resistance, obesity, and macrophage function utilizing both normal diet and the diet-induced obesity model.  相似文献   

7.

OBJECTIVE

SH2B1 is a SH2 domain-containing adaptor protein expressed in both the central nervous system and peripheral tissues. Neuronal SH2B1 controls body weight; however, the functions of peripheral SH2B1 remain unknown. Here, we studied peripheral SH2B1 regulation of insulin sensitivity and glucose metabolism.

RESEARCH DESIGN AND METHODS

We generated TgKO mice expressing SH2B1 in the brain but not peripheral tissues. Various metabolic parameters and insulin signaling were examined in TgKO mice fed a high-fat diet (HFD). The effect of SH2B1 on the insulin receptor catalytic activity and insulin receptor substrate (IRS)-1/IRS-2 dephosphorylation was examined using in vitro kinase assays and in vitro dephosphorylation assays, respectively. SH2B1 was coexpressed with PTP1B, and insulin receptor–mediated phosphorylation of IRS-1 was examined.

RESULTS

Deletion of peripheral SH2B1 markedly exacerbated HFD-induced hyperglycemia, hyperinsulinemia, and glucose intolerance in TgKO mice. Insulin signaling was dramatically impaired in muscle, liver, and adipose tissue in TgKO mice. Deletion of SH2B1 impaired insulin signaling in primary hepatocytes, whereas SH2B1 overexpression stimulated insulin receptor autophosphorylation and tyrosine phosphorylation of IRSs. Purified SH2B1 stimulated insulin receptor catalytic activity in vitro. The SH2 domain of SH2B1 was both required and sufficient to promote insulin receptor activation. Insulin stimulated the binding of SH2B1 to IRS-1 or IRS-2. This physical interaction inhibited tyrosine dephosphorylation of IRS-1 or IRS-2 and increased the ability of IRS proteins to activate the phosphatidylinositol 3-kinase pathway.

CONCLUSIONS

SH2B1 is an endogenous insulin sensitizer. It directly binds to insulin receptors, IRS-1 and IRS-2, and enhances insulin sensitivity by promoting insulin receptor catalytic activity and by inhibiting tyrosine dephosphorylation of IRS proteins.Insulin decreases blood glucose both by promoting glucose uptake into skeletal muscle and adipose tissue and by suppressing hepatic glucose production. In type 2 diabetes, the ability of insulin to reduce blood glucose is impaired (insulin resistance) because of a combination of genetic and environmental factors, resulting in hyperglycemia. Insulin resistance is not only the hallmark but also a determinant of type 2 diabetes.Insulin binds to and activates the insulin receptor. Insulin receptor tyrosyl phosphorylates insulin receptor substrates (IRS-1, -2, -3, and -4). IRS proteins, particularly IRS-1 and IRS-2, initiate and coordinate multiple downstream pathways, including the phosphatidylinositol 3-kinase/Akt pathway (1). Genetic deletion of IRS-1, IRS-2, or Akt2 causes insulin resistance in mice, indicating that the IRS protein/phosphatidylinositol 3-kinase/Akt2 pathway is required for regulation of glucose homeostasis by insulin (25). Insulin receptor and IRS proteins are negatively regulated by various intracellular molecules, including PTP1B, Grb10, Grb14, SOCS1, SOCS3, JNK, PKCθ, S6K, and IKKβ (623). The relative contribution of these negative regulators to the progression of insulin resistance has been extensively studied (624). However, insulin signaling is likely to also be modulated by positive regulators. In this study, we demonstrate that SH2B1 is a novel endogenous insulin sensitizer.SH2B1 is a member of the SH2B family of adapter proteins that also includes SH2B2 (APS) and SH2B3 (Lnk). SH2B1 and SH2B2 are expressed in multiple tissues, including insulin target tissues (e.g., skeletal muscle, adipose tissue, liver, and the brain); by contrast, SH2B3 expression is restricted to hematopoietic tissue (25,26). Structurally, SH2B family members have an NH2-terminal dimerization domain, a central pleckstrin homology domain, and a COOH-terminal Src homology 2 (SH2) domain. The dimerization domain mediates homodimerization or heterodimerization between different SH2B proteins (27). SH2B1 and SH2B2 bind via their SH2 domains to a variety of tyrosine phosphorylated proteins, including JAK2 and insulin receptor, in cultured cells (28). Genetic deletion of SH2B1 results in marked leptin resistance, obesity, insulin resistance, and type 2 diabetes in mice, demonstrating that SH2B1 is required for the maintenance of normal body weight, insulin sensitivity, and glucose metabolism (2932). Surprisingly, SH2B2-null mice have normal body weight and slightly improved insulin sensitivity (32,33), suggesting that SH2B1 and SH2B2 have distinct functions in vivo. However, it remains unclear whether SH2B1 cell autonomously regulates insulin sensitivity in peripheral insulin target tissues because systemic deletion of SH2B1 causes obesity, which may cause insulin resistance in SH2B1-null mice.We generated a mouse model in which recombinant SH2B1 is specifically expressed in the brain of SH2B1-null mice (TgKO) using transgenic approaches (31). Neuron-specific restoration of SH2B1 corrects both leptin resistance and obesity, suggesting that neuronal SH2B1 regulates energy balance and body weight by enhancing leptin sensitivity (31). Consistent with these conclusions, polymorphisms in the SH2B1 loci are linked to leptin resistance and obesity in humans (3436). In this work, we demonstrate that deletion of SH2B1 in peripheral tissues impairs insulin sensitivity independent of obesity in TgKO mice. Moreover, we demonstrate that SH2B1 directly promotes insulin responses by stimulating insulin receptor catalytic activity and by protecting IRS proteins from tyrosine dephosphorylation.  相似文献   

8.
Vasculopathy is a major complication of diabetes; however, molecular mechanisms mediating the development of vasculopathy and potential strategies for prevention have not been identified. We have previously reported that C-peptide prevents diabetic vasculopathy by inhibiting reactive oxygen species (ROS)-mediated endothelial apoptosis. To gain further insight into ROS-dependent mechanism of diabetic vasculopathy and its prevention, we studied high glucose–induced cytosolic and mitochondrial ROS production and its effect on altered mitochondrial dynamics and apoptosis. For the therapeutic strategy, we investigated the vasoprotective mechanism of C-peptide against hyperglycemia-induced endothelial damage through the AMP-activated protein kinase α (AMPKα) pathway using human umbilical vein endothelial cells and aorta of diabetic mice. High glucose (33 mmol/L) increased intracellular ROS through a mechanism involving interregulation between cytosolic and mitochondrial ROS generation. C-peptide (1 nmol/L) activation of AMPKα inhibited high glucose–induced ROS generation, mitochondrial fission, mitochondrial membrane potential collapse, and endothelial cell apoptosis. Additionally, the AMPK activator 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside and the antihyperglycemic drug metformin mimicked protective effects of C-peptide. C-peptide replacement therapy normalized hyperglycemia-induced AMPKα dephosphorylation, ROS generation, and mitochondrial disorganization in aorta of diabetic mice. These findings highlight a novel mechanism by which C-peptide activates AMPKα and protects against hyperglycemia-induced vasculopathy.C-peptide and insulin are cosecreted in equimolar amounts into the circulation from the pancreatic β-cells of Langerhans (1). C-peptide deficiency is a prominent attribute of type 1 diabetes (1). Deficiencies of C-peptide and insulin may also occur in the late stages of type 2 diabetes as a result of progressive loss of β-cells (24). Recent evidence demonstrates a beneficial role for C-peptide in diabetic neuropathy (1,5,6), nephropathy (1,6,7), and vascular dysfunction (1,5) and inflammation (1). C-peptide protects against diabetic vascular damage by promoting nitric oxide (NO) release (8) and suppressing nuclear factor-κB (9), which suppresses leukocyte-endothelium interactions (8,9). C-peptide may prevent atherosclerosis by inhibiting vascular smooth muscle proliferation and migration (10) and reducing venous neointima formation (11). However, the molecular mechanism by which C-peptide prevents diabetes complications is not understood well enough to permit its clinical implementation.Generation of reactive oxygen species (ROS) in response to high glucose is the leading cause of endothelial damage and diabetic vasculopathy (12). Protein kinase C (PKC)-dependent NADPH oxidase is considered a major cytosolic mediator of ROS generation in endothelial cells (13,14) that play a central role in hyperglycemia-induced endothelial cell apoptosis and vascular complications (1517). Overproduction of intracellular ROS by mitochondria also occurs during the development of hyperglycemia-induced vascular complications (12,18,19). Altered mitochondrial dynamics due to mitochondrial fission were recently linked with endothelial dysfunction in diabetes (20,21). However, the mechanisms regulating production of cytosolic and mitochondrial ROS and their individual functions in regulating mitochondrial dynamics and apoptosis remain to be elucidated.AMP-activated protein kinase (AMPK) is an intracellular energy and stress sensor (22) and is an emerging target for preventing diabetes complications (23), as exhibited by the most common antihyperglycemic drugs, rosiglitazone (24) and metformin (25). AMPK prevents apoptosis of endothelial cells (2628) by inhibiting ROS generation by NADPH oxidase (24,29) and mitochondria (30). Additionally, AMPK dephosphorylation is associated with diabetes (22,31,32). It has been reported that C-peptide inhibits high glucose–induced mitochondrial superoxide generation in renal microvascular endothelial cells (7). We recently demonstrated a key role for C-peptide in preventing high glucose–induced ROS generation and apoptosis of endothelial cells through inhibition of transglutaminase (17). However, the mechanism underlying C-peptide–mediated inhibition of intracellular ROS production and subsequent apoptosis remains unclear. Thus, we hypothesized that the potential protective role of C-peptide could be attributed to activation of AMPK, which results in reduced hyperglycemia-induced production of intracellular ROS and altered mitochondrial dynamics that suppress apoptosis of endothelial cells.In this study, we sought to elucidate the mechanism by which C-peptide protects against hyperglycemia-induced ROS production and subsequent endothelial cell damage. We examined the beneficial effect of C-peptide through AMPKα activation and subsequent protection against hyperglycemia-induced production of intracellular ROS, dysregulation of mitochondrial dynamics, mitochondrial membrane potential (∆Ψm) collapse, and apoptosis of endothelial cells. These studies were confirmed in vivo in mice with streptozotocin-induced diabetes using C-peptide supplement therapy delivered through osmotic pumps. Thus, our study implicates C-peptide replacement therapy as a potentially significant approach for preventing diabetes complications.  相似文献   

9.
Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy is a rare autoimmune disorder. The clinical spectrum of symptoms is diverse; the diagnosis relying on the presence of at least two out of the three main conditions defining the syndrome: chronic mucocutaneous candidiasis, hypoparathyroidism, and Addison''s disease.Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), also called autoimmune polyendocrine syndrome type I (APS-1), is a rare organ-specific autosomal recessive disease (OMIM 240300).13 Although APECED may occur worldwide, it remains most common in Iranian Jews (1:9,000),4 Sardinians (1:14,500),512 Finns (1:25,000),13 Slovenians (1:43,000),14 Norwegians (1:80,000),15 Polish (1:1,29,000),16 and Japanese (1:10,000,000).17 Multiple organ failures in APECED are caused by a progressive loss of tolerance against self-antigens resulting in an immunological attack and secondary destruction of the adrenals, parathyroid glands, β cells of islet of Langerhans,18 stomach, and small intestine.13 Many of the auto-antigens have been identified and characterized.9,12,19  相似文献   

10.

OBJECTIVE

Analysis of energy expenditure (EE) in mice is essential to obesity research. Since EE varies with body mass, comparisons between lean and obese mice are confounded unless EE is normalized to account for body mass differences. We 1) assessed the validity of ratio-based EE normalization involving division of EE by either total body mass (TBM) or lean body mass (LBM), 2) compared the independent contributions of LBM and fat mass (FM) to EE, and 3) investigated whether leptin contributes to the link between FM and EE.

RESEARCH DESIGN AND METHODS

We used regression modeling of calorimetry and body composition data in 137 mice to estimate the independent contributions of LBM and FM to EE. Subcutaneous administration of leptin or vehicle to 28 obese ob/ob mice and 32 fasting wild-type mice was used to determine if FM affects EE via a leptin-dependent mechanism.

RESULTS

Division of EE by either TBM or LBM is confounded by body mass variation. The contribution of FM to EE is comparable to that of LBM in normal mice (expressed per gram of tissue) but is absent in leptin-deficient ob/ob mice. When leptin is administered at physiological doses, the plasma leptin concentration supplants FM as an independent determinant of EE in both ob/ob mice and normal mice rendered leptin-deficient by fasting.

CONCLUSIONS

The contribution of FM to EE is substantially greater than predicted from the metabolic cost of adipose tissue per se, and the mechanism underlying this effect is leptin dependent. Regression-based approaches that account for variation in both FM and LBM are recommended for normalization of EE in mice.The maintenance of stable body weight is achieved through a process termed “energy homeostasis” that matches energy intake to energy expenditure (EE) over long time intervals (1). Accordingly, when animals experience a sustained increase of energy intake (e.g., during consumption of an energy-rich highly palatable diet), an adaptive increase of metabolic rate can help to limit the associated weight gain (2). However, the ability to quantify adaptive changes of EE is confounded in that larger animals tend to have a higher metabolic rate than smaller ones. Therefore, to reliably detect changes in EE that are not due simply to differences in body size per se, EE must be normalized to body mass using a method that eliminates this confounding effect. To date, most rodent studies of obesity use ratio-based normalization methods whereby EE is divided by either total body mass (TBM) or lean body mass (LBM) (36). However, these two methods can give widely divergent results when applied to the same data (3,4,6,7).A recent Diabetes Perspectives article (7) cogently reviewed the confounding effect of normalizing EE via division by TBM in mice, particularly when groups being compared differ in fat mass (FM). Accompanying this caution was the recommendation that EE be normalized via division by LBM instead (7) on grounds that FM consumes much less energy than LBM. Despite its intuitive appeal, dividing EE by LBM is theoretically problematic as a means to remove the influence of body size variation from group comparisons. The linear relationship between EE and either TBM or LBM is typically characterized by a positive y (EE) intercept term (813) due to heterogeneity inherent in the EE of various tissues comprising LBM (14). Consequently, dividing resting or average EE by either TBM or LBM mathematically forces heavier individuals to have a lower normalized EE than smaller ones (813), a concept first articulated >60 years ago (8). One approach that has been forwarded to obviate this mathematical bias is to use allometric scaling (15) wherein a TBM scaling exponent b and scaling coefficient a must be identified based on the data (15,16) such that EE divided by TBMb assumes the constant expected value a. This approach, however, is limited by interpretational and other difficulties (17), and the notion that a predetermined fixed TBM scaling exponent can be applied universally has been challenged (16,18).Normalizing EE in human studies is now accomplished using multiple regression methods that adjust group comparisons of EE for differences in body mass so as to eliminate the influence of body size variation per se from evaluations of key independent variables such as ethnicity, sex, genotype, or nutritional status (9,1928). Although multiple regression has been used in animal studies (11,2935), the relative importance of FM and LBM as determinants of metabolic rate in mice remains an open question. Indeed, both human and animal investigations suggest that the energy cost of FM in vivo is greater than expected on the basis of its intrinsic metabolic rate (20,28,31,36). This possibility is consistent with evidence that changes in FM can influence metabolic rate at least in part through homeostatic adjustments of EE that promote body weight stability (2,3742). Testing this hypothesis, however, requires the application of valid strategies for normalizing EE to body size.In the current work, we demonstrate in a large sample of mice that ratio-based normalization of EE is problematic, even when LBM is used in lieu of TBM. Moreover, multiple regression analysis indicates that variation in FM makes a surprisingly large contribution to EE. These findings lead us to support recommendations for the broad use of regression-based approaches to normalizing EE in mice that take both FM and LBM into account (912,19,43,44). Based on the hypothesis that the effect of FM on EE reflects adaptive responses involving the adipocyte hormone leptin, we asked 1) whether the effect of FM on EE is absent in ob/ob mice that lack a leptin signal, 2) if the plasma leptin level supplants FM as an independent determinant of EE when leptin is administered to ob/ob mice at physiological doses, and 3) whether in wild-type (WT) mice rendered leptin-deficient by fasting, the plasma leptin level emerges as a determinant of EE when physiological replacement is achieved by exogenous leptin administration. Our results confirm each of these predictions and therefore implicate circulating leptin in the mechanism whereby FM variation affects EE.  相似文献   

11.
Objective: To evaluate the efficacy, safety, and pain of in-office “painless” aminolevulinic acid photodynamic therapy aimed at decreasing treatment-associated pain in patients undergoing removal of actinic keratoses. Design: Prospective split-face study comparing short aminolevulinic acid incubation times of 15 minutes followed by extended exposure (60 minutes) of continuous blue light versus conventional aminolevulinic acid photodynamic therapy. Prospective assessment of pain in patients undergoing in-office “painless” aminolevulinic acid photodynamic therapy. Setting: Clinical practice office. Participants: Three patients with actinic keratoses participated in the split-face study and 101 in the pain assessment study. Measurements: Evaluations in the split-face study included removal of actinic keratoses, skin temperature, and pain measured on a 10-point visual analog scale. Pain was assessed using the visual analog scale in the pain assessment study. Results: In the split-face study, in-office “painless” aminolevulinic acid photodynamic therapy resulted in a 52-percent reduction in lesions versus 44 percent for conventional aminolevulinic acid photodynamic therapy. Maximum pain scores of in-office “painless” aminolevulinic acid photodynamic therapy were all 0 at each time point, and the average score for conventional aminolevulinic acid photodynamic therapy was 7. Baseline skin temperatures increased from a baseline of 29 to 32°C to 34 to 35°C by minute 10 of blue light activation on both sides of the face. Results from the pain assessment study indicated no or minimal (scores 0-2) pain in nearly all patients who received in-office “painless” aminolevulinic acid photodynamic therapy as monotherapy or in combination with 5-fluoruacil or imiquimod used as pretreatments. Conclusions: In-office “painless” aminolevulinic acid photodynamic therapy appears to be effective for removing actinic keratoses and is associated with little or no pain in nearly all patients. This procedure should be evaluated in large-scale controlled trials.Actinic keratoses (AKs) are part of the spectrum between photodamaged skin and invasive squamous cell carcinoma (SCC).1-5 They are a major health care concern because of their increasing prevalence worldwide,6-10 economic impact,9-11 and decreased quality of life of affected individuals.10,12 Results from observational studies have indicated that AKs evolve into primary invasive SCC or in situ SCC at a rate ranging between 1/1,000 lesions per year13 to 0.60 percent at one year and 2.57 percent at four years.14 It is recommended that all AKs be treated because it is not currently possible to predict which will evolve into invasive SCC.15-17A variety of therapeutic modalities are used to treat AKs.1,18-20 Focally destructive therapies, such as cryotherapy,21 electrodessication and curettage,22 and shave excision23 are most often used to treat individual AKs. Large areas of actinically damaged skin require “field therapies” such as 5-fluoruacil (5-FU),24-26 imiquimod,26-30 diclofenac gel,31-33 ingenol mebutate,34,36 aminolevulinic acid photodynamic therapy (ALA PDT)35,37 and methyl-aminolevulinic acid PDT (MAL PDT),38,39 chemical peels,40 dermabrasion,41-43 and laser resurfacing.44,45PDT produces reactive oxygen species that result in tissue destruction46 and it destroys AKs because of the preferential accumulation of the photosensitizing molecule, protoporphyrin IX (PpIX) within AKs following topical application of pro-drugs ALA47 and MAL.48 PDT is safe and effective for treatment of large surface skin areas, provides good adherence because it is performed under supervision in a clinic setting, has minimal post-treatment downtime versus other AK field therapies, and produces good-to-excellent cosmetic outcomes with minimal potential for scarring.49,50 PDT also has several drawbacks, most notably pain during the first few minutes of light activation phase.51-53 Nearly two-thirds of patients undergoing ALA PDT report this pain as “moderate-severe” following 1, 2, or 3-hour ALA incubations.54 Pain with PDT has been related to cellular destruction and inflammation and possibly a direct effect of PDT on nerve fibers55-57; it has now become clear that PDT-related pain is associated with PpIX tissue accumulation based on fluorescence and the fluence rate of the activating light source.58 Topical anesthetics,55 cooling devices,59-61 nerve blockade,61,62 and treatment interruption63 have limited efficacy in managing PDT-related pain, which can lead to reluctance of patients to undergo future PDT treatments.A novel approach to minimizing discomfort during PDT, daylight-mediated PDT, uses a brief (30-minute) incubation period followed by 1.5 to 2.5 hours of daylight exposure.64-66 The shortened incubation period is designed to minimize PpIX build-up in the targeted tissue prior to daylight PpIX activation, and photobleaching prevents further buildup of PpIX and minimizes patient discomfort.67,68 Limitations to daylight-mediated PDT include dependence on favorable weather conditions and patient adherence to the treatment protocol outside the clinic.65Based on the efficacy and improved tolerability of daylight-mediated PDT, an in-office painless (IOP) ALA PDT protocol was developed. It involves applying ALA topically to actinically damaged skin, incubating without occlusion for 15 minutes, and then 60 minutes of continuous blue light activation. This report summarizes results from a split-face comparison of IOP ALA PDT and a standard short (75 minute) ALA incubation protocol followed by the standard 1,000 seconds of blue light activation carried out in three patients, and assessment of pain associated with this treatment in 101 patients undergoing 121 treatments over a two-year time period. In the latter study, IOP ALA PDT was employed as either a full-face monotherapy or in combination with one week of prior treatment with 5% and 0.5% 5-FU and 3.75% and 5% imiquimod.  相似文献   

12.

OBJECTIVE

The medial hypothalamus mediates leptin-induced glucose uptake in peripheral tissues, and brain melanocortin receptors (MCRs) mediate certain central effects of leptin. However, the contributions of the leptin receptor and MCRs in individual medial hypothalamic nuclei to regulation of peripheral glucose uptake have remained unclear. We examined the effects of an injection of leptin and the MCR agonist MT-II into medial hypothalamic nuclei on glucose uptake in peripheral tissues.

RESEARCH DESIGN AND METHODS

Leptin or MT-II was injected into the ventromedial (VMH), dorsomedial (DMH), arcuate nucleus (ARC), or paraventricular (PVH) hypothalamus or the lateral ventricle (intracerebroventricularly) in freely moving mice. The MCR antagonist SHU9119 was injected intracerebroventricularly. Glucose uptake was measured by the 2-[3H]deoxy-d-glucose method.

RESULTS

Leptin injection into the VMH increased glucose uptake in skeletal muscle, brown adipose tissue (BAT), and heart, whereas that into the ARC increased glucose uptake in BAT, and that into the DMH or PVH had no effect. SHU9119 abolished these effects of leptin injected into the VMH. Injection of MT-II either into the VMH or intracerebroventricularly increased glucose uptake in skeletal muscle, BAT, and heart, whereas that into the PVH increased glucose uptake in BAT, and that into the DMH or ARC had no effect.

CONCLUSIONS

The VMH mediates leptin- and MT-II–induced glucose uptake in skeletal muscle, BAT, and heart. These effects of leptin are dependent on MCR activation. The leptin receptor in the ARC and MCR in the PVH regulate glucose uptake in BAT. Medial hypothalamic nuclei thus play distinct roles in leptin- and MT-II–induced glucose uptake in peripheral tissues.Leptin is an adipocyte hormone that inhibits food intake and increases energy expenditure (1). The hypothalamus is a principal target of leptin in its regulation of energy metabolism (25). The arcuate nucleus (ARC) is the most well characterized of hypothalamic nuclei in terms of its role in the central effects of leptin (25). The ARC contains two populations of leptin-responsive neurons: pro-opiomelanocortin (POMC)-expressing neurons, which release the potent anorexic peptide α-melanocyte–stimulating hormone, and neurons that release two potent orexigenic peptides, agouti-related peptide (AgRP) and neuropeptide Y (NPY) (25). α-Melanocyte–stimulating hormone activates the melanocortin receptor (MCR), whereas AgRP competitively inhibits this receptor and NPY functionally antagonizes MCR signaling (6). Both sets of neurons project to second-order MCR-expressing neurons within the hypothalamus, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and lateral hypothalamus, as well as to other brain regions such as the brain stem (2,4,7,8). Leptin inhibits food intake through reciprocal regulation of POMC and AgRP/NPY neurons in the ARC and consequent activation of MCR in hypothalamic nuclei, including the PVH (5,6,7,9). Mice lacking the melanocortin 3 (MC3R) or 4 (MC4R) receptor show increased adiposity and feeding efficiency (4). Restoration of MC4R expression in certain sets of PVH neurons prevented hyperphagia and reduced body weight in MC4R-null mice (9). In addition to that in the ARC, the leptin receptor Ob-Rb in other hypothalamic nuclei has also been shown to regulate energy intake and adiposity. Neurons positive for steroidogenic factor 1 (SF1; also known as Ad4BP) (10,11) are largely restricted to the VMH in the adult brain. Leptin depolarizes these neurons, and specific ablation of the leptin receptor in SF1-positive cells induced obesity and increased susceptibility to a high-fat diet in mice (12).The leptin receptor in the brain also regulates glucose metabolism in certain peripheral tissues (1317). Treatment with leptin ameliorates diabetes in lipodystrophic mice and humans (18,19). Intravenous or intracerebroventricular administration of leptin markedly increased whole-body glucose turnover and glucose uptake by certain tissues in mice without any substantial change in plasma insulin or glucose levels (13). We have also previously shown that microinjection of leptin into the medial hypothalamus, such as into the VMH, but not into the lateral hypothalamus, preferentially increased glucose uptake in skeletal muscle, heart, and brown adipose tissue (BAT) (1416). Restoration of Ob-Rb expression in the ARC and the VMH of the Ob-Rb–mutated Koletsky rat by adenovirus- or adeno-associated virus–mediated gene transfer improved peripheral insulin sensitivity and reduced plasma glucose concentration (17,20). Ablation of suppressor of cytokine signaling 3 (SOCS3) in SF1-positive cells (10,11) improved glucose homeostasis in mice fed a high-fat diet (21). Furthermore, intracerebroventricular injection of the MCR agonist (MT-II) increased whole-body glucose turnover and expression of GLUT4 in skeletal muscle (22). Ob-Rb in the ARC and the VMH as well as the brain melanocortin pathway are thus implicated in the regulation of glucose uptake in peripheral tissues as well as in energy metabolism. However, little is known about the contributions of the leptin receptor and MCR in individual medial hypothalamic nuclei to regulation of glucose uptake in peripheral tissues, as opposed to their roles in the regulation of food intake and leanness.We have now examined the acute effects of microinjection of leptin and MT-II into the VMH, ARC, DMH, and PVH, all of which express Ob-Rb, MC3R, and MC4R at a high level (37,2325), on glucose uptake in peripheral tissues of mice in vivo. Our results suggest that the VMH mediates stimulatory actions of leptin and MT-II on glucose uptake in skeletal muscle, heart, and BAT, whereas the leptin receptor in the ARC as well as MCRs in PVH regulate glucose uptake in BAT. The medial hypothalamic nuclei thus appear to play distinct roles in the regulation of glucose uptake in peripheral tissues by leptin and MT-II.  相似文献   

13.
14.
15.
Vitiligo is one of the most common dermatological disorders, appearing as one or more white macules or patches and affecting up to two percent of the population worldwide. The undesirable aesthetic properties of vitiligo, especially facial, may result in significant negative psychosocial effects, particularly a rate of depression twice that of the general population. While there is no cure, there are several treatment options, notably depigmentation in severe cases. Monobenzone is the most potent depigmenting agent. However, its use is limited due to the permanent and potent nature of the drug. This case presents an example of when timely and aggressive treatment with monobenzone is warranted, demonstrating excellent clinical response, which resulted in a significant increase in the quality of life in a patient with severe vitiligo.Vitiligo is among one of the most common dermatological disorders, affecting up to two percent of the population worldwide.1,2 A chronic and usually progressive disorder, vitiligo presents discretely before 20 years of age, although first presentation in later life may occur as well.2 Clinically, it appears as one or more well-circumscribed, hypopigmented, white macules or patches. This is due to the acquired autoimmune destruction of melanocytes, most often in areas of greater pigmentation, such as the face and dorsum of the hands where they are most exposed to UV radiation.2 Apart from the cosmetic appearance, it is usually asymptomatic, although there is a greater tendency for sunburns and pruritis.2The undesirable aesthetic properties of vitiligo, especially facial, may result in significant negative psychosocial effects, notably a rate of depression twice that of the general population.3 In some cultures, vitiligo is not well understood. The depigmentation of vitiligo is thought to result from sexually transmitted infections, or of leprosy, and can have a damaging effect on educational, social, and employment opportunities.1,2,4 Patients may feel embarrassed or ashamed of such a visible disorder. Studies have shown that vitiligo is associated with a greater burden of disease to patients, especially those in populations with dark skin.1,2,4 Therefore, treatment, although not medically necessary, provides large psychosocial gains for the patient, increasing their quality of life.There is no cure for vitiligo. Current treatment for vitiligo attempts to either increase or decrease pigmentation in order to achieve cosmetically pleasing results and increase the patient''s self-esteem.5 Repigmentation tends to require a prolonged treatment course and yield minimal positive results.6 Strong topical steroids are generally the first line of treatment, with only a 50- to 75-percent repigmentation rate.5,6 Tactrolimus, an immunosuppressive, and calcipotriene, a vitamin D analogue, are alternative topical repigmenting agents, with a similar efficacy as the topical steroids.6 When such treatment fails, psoralen plus ultraviolet A radiation (PUVA) and narrow-band ultraviolet B radiation (NB-UVB) are effective alternatives.5,6 However, PUVA can be carcinogenic and NB-UVB has low efficacy; both require prolonged treatments.6,7 Alternative treatments exist, such as melanocyte transplantations. However, despite the type of treatment, repigmentation still remains difficult and time consuming, especially with advanced vitiligo.Due to the difficulties with repigmentation, it is often easier to achieve depigmentation, especially when vitiligo affects more than 50 percent of the body.57 It is, however, a more aggressive approach and its use is considered on an individual basis because of the irreversible changes and increased sensitivity to sunburn of the treated areas. Several treatment modalities exist. Phenols, lasers, cryotherapy, and depigmenting systemic agents, such as imatinib, imiquimod, and diphencyprone, are often considered.7 Monobenzone (monobenzyl ether of hydroquinone, MBEH) is usually the treatment of choice of depigmentation therapy for severe cases of vitiligo, and MBEH is usually used in concentrations of 20 to 40 percent to achieve the desired permanent depigmentation.5,7 It achieves its effects by inducing the necrotic death of melanocytes.7 Topical all-trans-retinoic acid (RA), a vitamin A derivative, causes mild depigmentation and when used in combination with MBEH, has synergistic effects, yielding depigmentation in a short amount of time.7 Nair et al8 have proposed that the RA enhances the absorption of monobenzone by melanocytes through the inactivation of their glutathione-dependent defense mechanisms.7,8 Side effects of MBEH include skin irritation, contact dermatitis, ocular side effects, exogenous ochronosis, and difficulties in predicting response.5,911 There can be repigmentation because of sun exposure or rarely as a reaction to the drug.5,7,10,11 Due to these side effects, MBEH treatment can be somewhat controversial, and its use has been limited in some countries, such as the Netherlands, which has restricted it since 1990.7 MBEH has been approved by the United States Food and Drug Administration since 1952 for permanent depigmentation of extensive vitiligo.  相似文献   

16.

OBJECTIVE

To test the graft-promoting effects of mesenchymal stem cells (MSCs) in a cynomolgus monkey model of islet/bone marrow transplantation.

RESEARCH DESIGN AND METHODS

Cynomolgus MSCs were obtained from iliac crest aspirate and characterized through passage 11 for phenotype, gene expression, differentiation potential, and karyotype. Allogeneic donor MSCs were cotransplanted intraportally with islets on postoperative day (POD) 0 and intravenously with donor marrow on PODs 5 and 11. Recipients were followed for stabilization of blood glucose levels, reduction of exogenous insulin requirement (EIR), C-peptide levels, changes in peripheral blood T regulatory cells, and chimerism. Destabilization of glycemia and increases in EIR were used as signs of rejection; additional intravenous MSCs were administered to test the effect on reversal of rejection.

RESULTS

MSC phenotype and a normal karyotype were observed through passage 11. IL-6, IL-10, vascular endothelial growth factor, TGF-β, hepatocyte growth factor, and galectin-1 gene expression levels varied among donors. MSC treatment significantly enhanced islet engraftment and function at 1 month posttransplant (n = 8), as compared with animals that received islets without MSCs (n = 3). Additional infusions of donor or third-party MSCs resulted in reversal of rejection episodes and prolongation of islet function in two animals. Stable islet allograft function was associated with increased numbers of regulatory T-cells in peripheral blood.

CONCLUSIONS

MSCs may provide an important approach for enhancement of islet engraftment, thereby decreasing the numbers of islets needed to achieve insulin independence. Furthermore, MSCs may serve as a new, safe, and effective antirejection therapy.Multipotent mesenchymal stem cells (MSCs) (1,2) can deliver immunomodulatory signals (37) that inhibit allogeneic T-cell responses through downregulation of the proinflammatory cytokines TNF-α and IFN-γ and production of the regulatory cytokines/molecules IL-10, hepatocyte growth factor (HGF), TGF-β, vascular endothelial growth factor (VEGF), indoleamine 2,3-dioxygenase, galectin-1, prostaglandin E2, nitric oxide, and matrix metalloproteinase-2 and -9 (3,812). Inflammatory signals, such as IFN-γ, can activate and upregulate MSC suppressive activities (9,13). These cells are able to migrate to sites of injury after intravenous injection (14,15). Their use in clinical trials and experimental models is based on their immunomodulatory and regenerative properties (1,7,16). Clinically, MSCs have been observed to enhance donor bone marrow cell (DBMC) engraftment and chimerism (17,18). Therefore, cotransplantation of MSCs that secrete immunomodulatory cytokines and growth factors might enhance islet survival and function. In experimental mouse models, intravenously infused MSCs are capable of migrating to pancreatic islets (19,20). Systemic infusion of MSCs in murine models of diabetes was accompanied by delayed onset of diabetes, improved glycemic levels, reduced pancreatic insulitis, and pancreatic tissue regeneration (19,2125), as well as prevention of autoimmune destruction of β-cells via induction of regulatory T-cells (Tregs) (26). Cotransplantation of syngeneic MSCs with a marginal mass of allogeneic islets under the kidney capsule of streptozotocin (STZ)-induced diabetic mice resulted in prolonged normoglycemia (11). Cotransplantation of syngeneic MSC with a marginal mass of allogeneic islets has been performed in the omentum (27) and kidney capsule (28) of STZ-induced diabetic rats, with enhanced islet graft survival as compared with animals receiving islets alone. In this study, cynomolgus monkey MSCs were characterized and donor MSCs were examined for the ability to promote intraportal islet engraftment as well as chimerism in recipients of islet/DBMC transplants. In addition, we tested the use of donor or third-party MSCs to reverse episodes of islet allograft rejection.  相似文献   

17.
Inflammation contributes to the pathogenesis of acute kidney injury. Dendritic cells (DCs) are immune sentinels with the ability to induce immunity or tolerance, but whether they mediate acute kidney injury is unknown. Here, we studied the distribution of DCs within the kidney and the role of DCs in cisplatin-induced acute kidney injury using a mouse model in which DCs express both green fluorescence protein and the diphtheria toxin receptor. DCs were present throughout the tubulointerstitium but not in glomeruli. We used diphtheria toxin to deplete DCs to study their functional significance in cisplatin nephrotoxicity. Mice depleted of DCs before or coincident with cisplatin treatment but not at later stages experienced more severe renal dysfunction, tubular injury, neutrophil infiltration and greater mortality than nondepleted mice. We used bone marrow chimeric mice to confirm that the depletion of CD11c-expressing hematopoietic cells was responsible for the enhanced renal injury. Finally, mixed bone marrow chimeras demonstrated that the worsening of cisplatin nephrotoxicity in DC-depleted mice was not a result of the dying or dead DCs themselves. After cisplatin treatment, expression of MHC class II decreased and expression of inducible co-stimulator ligand increased on renal DCs. These data demonstrate that resident DCs reduce cisplatin nephrotoxicity and its associated inflammation.Innate immune responses are pathogenic in both ischemic and toxic acute renal failure. In response to renal injury, inflammatory chemokines and cytokines are produced both by renal parenchymal cells, such as proximal tubule epithelial cells, and resident or infiltrating leukocytes.14 The elaborated chemokines and cytokines, including TNF-α, IL-18, keratinocyte-derived chemokine, and monocyte chemoattractant protein 1, subsequently recruit additional immune cells to the kidney, such as neutrophils, T cells, monocytes, and inflammatory dendritic cells (DCs), which may cause further injury through pathways that are not fully defined.2,512 DCs are sentinels of the immune system and under steady-state conditions induce tolerance by various mechanisms, including production of TGF-β, IL-10, or indoleamine 2,3-dioxygenase1316; expression of PDL-1, PDL-2, or FcγR2B17,18; clonal deletion of autoreactive T cells19; and induction of T regulatory cells via the inducible co-stimulator (ICOS) pathway.2023 In response to pathogens or products of tissue injury, DCs mature and initiate immunity or inflammatory diseases.24,25 Monocytes recruited to inflamed tissue can also differentiate into inflammatory DCs and mediate defense against pathogens or contribute to inflammatory tissue responses.12,2628Although DCs represent a major population of immune cells in the kidney,29 their role in renal disease is poorly defined. Liposomal clodronate has been used to study the pathophysiologic role of phagocytic cells, which include DCs and macrophages.3,3032 An alternative DC-specific approach uses expression of the simian diphtheria toxin receptor (DTR) driven by the CD11c promoter to target DCs for DT-mediated cell death.24 This model has been used extensively to study the role of DCs in various physiologic and pathophysiologic contexts32,33; however, its application in kidney disease has been limited to recent studies of immune complex–mediated glomerulonephritis.12,23We have reported that inflammation plays an important role in the pathogenesis of cisplatin-induced acute kidney injury (AKI).1,4,5,34 Given the dearth of information regarding the role of renal DCs in AKI, this study examined the renal DC population and the impact of its depletion on cisplatin nephrotoxicity. We show that DCs are the most abundant population of renal resident leukocytes and form a dense network throughout the tubulointerstitium. Renal DCs displayed surface markers that distinguished them from splenic DCs. Using a conditional DC depletion model, we determined that DC ablation markedly exacerbates cisplatin-induced renal dysfunction, structural injury, and infiltration of neutrophils.  相似文献   

18.

OBJECTIVE

Glucokinase (GCK) and glucose-6-phosphatase catalytic subunit 2 (G6PC2) regulate the glucose-cycling step in pancreatic β-cells and may regulate insulin secretion. GCK rs1799884 and G6PC2 rs560887 have been independently associated with fasting glucose, but their interaction on glucose-insulin relationships is not well characterized.

RESEARCH DESIGN AND METHODS

We tested whether these variants are associated with diabetes-related quantitative traits in Mexican Americans from the BetaGene Study and attempted to replicate our findings in Finnish men from the METabolic Syndrome in Men (METSIM) Study.

RESULTS

rs1799884 was not associated with any quantitative trait (corrected P > 0.1), whereas rs560887 was significantly associated with the oral glucose tolerance test 30-min incremental insulin response (30′ Δinsulin, corrected P = 0.021). We found no association between quantitative traits and the multiplicative interaction between rs1799884 and rs560887 (P > 0.26). However, the additive effect of these single nucleotide polymorphisms was associated with fasting glucose (corrected P = 0.03) and 30′ Δinsulin (corrected P = 0.027). This additive association was replicated in METSIM (fasting glucose, P = 3.5 × 10−10 30′ Δinsulin, P = 0.028). When we examined the relationship between fasting glucose and 30′ Δinsulin stratified by GCK and G6PC2, we noted divergent changes in these quantitative traits for GCK but parallel changes for G6PC2. We observed a similar pattern in METSIM.

CONCLUSIONS

Our data suggest that variation in GCK and G6PC2 have additive effects on both fasting glucose and insulin secretion.Genome-wide association (GWA) studies have identified several loci for type 2 diabetes (15) and type 2 diabetes–related quantitative traits (620). Two of these loci, glucokinase (GCK) (2123) and glucose-6-phosphatase catalytic subunit 2 (G6PC2) (9,10), regulate the critical glucose-sensing mechanism within pancreatic β-cells. Mutations in GCK confer susceptibility to maturity-onset diabetes of the young (MODY)-2 (2426), and a −30 GCK promoter variant (rs1799884) has been shown to be associated with β-cell function (21), fasting glucose, and birth weight (23). Chen et al. (9) demonstrated an association between the G6PC2 region and fasting glucose, an observation replicated by Bouatia-Naji et al. (10). Although fasting glucose levels are associated with both GCK and G6PC2, there has been no evidence that genetic variation at these two loci contribute a risk for type 2 diabetes, suggesting contribution to mild elevations in glycemia.GCK phosphorylates glucose to glucose-6-phosphate, whereas G6PC2 dephosphorylates glucose-6-phosphate back to glucose, forming a glucose cycle previously demonstrated to exist within pancreatic β-cells (27,28). The important role of GCK in glucose sensing by pancreatic islets has been demonstrated by numerous studies, and other studies suggest a role for glucose cycling in insulin secretion and diabetes (2830), implying that the balance between GCK and G6PC2 activity is important for determining glycolytic flux, ATP production, and subsequent insulin secretion. This was validated by a demonstration that direct manipulation of glucose cycling alters insulin secretion (31,32).BetaGene is a study in which we are performing detailed phenotyping of Mexican American probands with recent gestational diabetes mellitus (GDM) and their family members to obtain quantitative estimates of body composition, insulin sensitivity (SI), acute insulin response (AIR), and β-cell compensation (disposition index) with the goal of identifying genes influencing variations in type 2 diabetes–related quantitative traits (3335). Based on the evidence that variation in GCK (rs1799884) and G6PC2 (rs560887) are independently associated with fasting glucose concentrations and both are crucial to glucose cycling in β-cells, we hypothesized that interaction between these loci may be associated not just with fasting glucose but also with measures of insulin secretion or β-cell function. We tested this hypothesis in the BetaGene Study and, for replication, in a separate sample of Finnish men participating in the METabolic Syndrome in Men (METSIM) Study (36).  相似文献   

19.

OBJECTIVE

To assess insulin action on peripheral glucose utilization and nonesterified fatty acid (NEFA) suppression as a predictor of coronary artery calcification (CAC) in patients with type 1 diabetes and nondiabetic controls.

RESEARCH DESIGN AND METHODS

Insulin action was measured by a three-stage hyperinsulinemic-euglycemic clamp (4, 8, and 40 mU/m2/min) in 87 subjects from the Coronary Artery Calcification in Type 1 Diabetes cohort (40 diabetic, 47 nondiabetic; mean age 45 ± 8 years; 55% female).

RESULTS

Peripheral glucose utilization was lower in subjects with type 1 diabetes compared with nondiabetic controls: glucose infusion rate (mg/kg FFM/min) = 6.19 ± 0.72 vs. 12.71 ± 0.66, mean ± SE, P < 0.0001, after adjustment for age, sex, BMI, fasting glucose, and final clamp glucose and insulin. Insulin-induced NEFA suppression was also lower in type 1 diabetic compared with nondiabetic subjects: NEFA levels (μM) during 8 mU/m2/min insulin infusion = 370 ± 27 vs. 185 ± 25, P < 0.0001, after adjustment for age, sex, BMI, fasting glucose, and time point insulin. Lower glucose utilization and higher NEFA levels, correlated with CAC volume (r = −0.42, P < 0.0001 and r = 0.41, P < 0.0001, respectively) and predicted the presence of CAC (odds ratio [OR] = 0.45, 95% CI = 0.22–0.93, P = 0.03; OR = 2.4, 95% CI = 1.08–5.32, P = 0.032, respectively). Insulin resistance did not correlate with GHb or continuous glucose monitoring parameters.

CONCLUSIONS

Type 1 diabetic patients are insulin resistant compared with nondiabetic subjects, and the degree of resistance is not related to current glycemic control. Insulin resistance predicts the extent of coronary artery calcification and may contribute to the increased risk of cardiovascular disease in patients with type 1 diabetes as well as subjects without diabetes.Cardiovascular disease (CVD) remains the leading cause of death in individuals with type 1 diabetes (14). Although hyperglycemia appears to be the primary mediator of microvascular disease (5,6), its role in macrovascular disease is less clear (4). Tight glycemic control improves, but does not normalize CVD risk, and correlation of GHb to CVD risk remains controversial (715). In addition, standard prediction rules for CVD risk do not accurately predict CVD in type 1 diabetic populations (16). Thus, the mechanism of accelerated atherosclerosis in type 1 diabetes is unclear and identification of those patients at highest risk and most in need of aggressive risk factor modification is inaccurate.In the general population, insulin resistance has been implicated as an important contributor to accelerated atherosclerosis (1725). Although type 1 diabetes is primarily a disease of insulin deficiency, previous studies have demonstrated insulin resistance and suggested that CVD may also be linked to insulin resistance in type 1 diabetes (10,2632). As early as 1968, Martin et al. (30) demonstrated an “impaired glucose assimilation index” and an inverse association between this index and prevalent macrovascular disease in type 1 diabetic subjects. More recently, the Pittsburgh Epidemiology of Diabetes Complications Study (10) found no correlation between GHb and coronary artery disease outcomes. However, in addition to other known CVD risk factors, estimated glucose disposal rate correlated inversely with these outcomes. Similar correlations of estimated insulin resistance or a surrogate of insulin resistance (waist-to-hip ratio) to coronary artery disease were also found in the Diabetes Control and Complications Trial (DCCT) and the EURODIAB study (33). These data suggest that an estimate of insulin resistance may add to CVD risk prediction in type 1 diabetes. In addition, elevated nonesterified fatty acid (NEFA) levels have been proposed to mediate the increased atherosclerotic risk associated with insulin resistance in the general population (18,3437). It is not known whether the defects in insulin action in type 1 diabetes extend beyond glucose utilization to NEFA suppression.The Coronary Artery Calcification in Type 1 Diabetes (CACTI) study has followed a cohort of type 1 diabetic subjects and similar nondiabetic controls with electron beam computed tomography for measurement of coronary artery calcification (CAC) and CVD outcomes for 6 years (15,38). We hypothesized that type 1 diabetic subjects would be more insulin resistant than nondiabetic controls in terms of both glucose utilization and NEFA suppression, and that both measures of insulin resistance would correlate with CAC, a marker of the extent of coronary atherosclerosis.  相似文献   

20.
Referral to a nephrologist before initiation of chronic dialysis occurs less frequently for blacks than whites, but the reasons for this disparity are incompletely understood. Here, we examined the contribution of racial composition by zip code on access and quality of nephrology care before initiation of renal replacement therapy (RRT). We retrospectively studied a cohort study of 92,000 white and black adults who initiated RRT in the United States between June 1, 2005, and October 5, 2006. The percentage of patients without pre-ESRD nephrology care ranged from 30% among those who lived in zip codes with <5% black residents to 41% among those who lived in areas with >50% black residents. In adjusted analyses, as the percentage of blacks in residential areas increased, the likelihood of not receiving pre-ESRD nephrology care increased. Among patients who received nephrology care, the quality of care (timing of care and proportion of patients who received a pre-emptive renal transplant, who initiated therapy with peritoneal dialysis, or who had a permanent hemodialysis access) did not differ by the racial composition of their residential area. In conclusion, racial composition of residential areas associates with access to nephrology care but not with quality of the nephrology care received.Clinical practice guidelines for chronic kidney disease emphasize the importance of timely referral to a nephrologist for patients expected to require renal replacement therapy.13 Nevertheless, approximately 33% of end-stage renal disease (ESRD) patients in the United States do not see a nephrologist before initiation of chronic dialysis.4,5 Lack of timely access to nephrology care is associated with several adverse outcomes after initiation of dialysis including higher mortality rates,69 higher rates of hospitalization,10 lower rates of renal transplantation,11,12 delayed creation of arteriovenous fistulae,13 lower rates of achievement of dialysis treatment targets,14,15 and a lower likelihood of receiving home-based dialysis therapies such as peritoneal dialysis and home hemodialysis.3,1618In the United States, black dialysis patients are less likely than white patients to have received nephrology care before onset of ESRD.9,1921 They are also less likely to receive a pre-emptive kidney transplant,22,23 select peritoneal dialysis over hemodialysis,24 and have a vascular access in place at onset of hemodialysis.25,26 Several factors may contribute to these disparities including differences in insurance status, level of education, physician knowledge or biases, and patient preferences.9,2729 In addition, geographic factors such as proximity to dialysis facilities and degree of urbanization also affect access to nephrology care.3033A substantial proportion of black patients are also more likely to live in areas where most other residents are black. A recent study demonstrated that both black and white dialysis patients living in predominantly black zip codes were less likely to receive a kidney transplant than patients living in other areas.34 Although levels of income, wealth and education tended to be lower among residents of predominantly black zip codes, lower transplant rates among dialysis patients living in these areas were not completely explained by these measures. Patients who live in predominantly black zip codes may face unique barriers to care that are not explained by measured socioeconomic characteristics of those zip codes. We therefore hypothesized that dialysis patients living in zip codes with a greater proportion of black residents would be less likely to have received pre-ESRD care and less likely to have received high-quality nephrology care than patients living in other zip codes. We also hypothesized that these relationships would not be completely explained by differences in zip code socioeconomic status or patient race.  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