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 531 毫秒
1.

OBJECTIVE

Type 1 diabetes is an autoimmune disease characterized by the destruction of insulin-producing β-cells. NOD mice provide a useful tool for understanding disease pathogenesis and progression. Although much has been learned from studies with NOD mice, increased understanding of human type 1 diabetes can be gained by evaluating the pathogenic potential of human diabetogenic effector cells in vivo. Therefore, our objective in this study was to develop a small-animal model using human effector cells to study type 1 diabetes.

RESEARCH DESIGN AND METHODS

We adoptively transferred HLA-A2–matched peripheral blood mononuclear cells (PBMCs) from type 1 diabetic patients and nondiabetic control subjects into transgenic NOD-scid/γcnull/HLA-A*0201 (NOD-scid/γcnull/A2) mice. At various times after adoptive transfer, we determined the ability of these mice to support the survival and proliferation of the human lymphoid cells. Human lymphocytes were isolated and assessed from the blood, spleen, pancreatic lymph node and islets of NOD-scid/γcnull/A2 mice after transfer.

RESULTS

Human T and B cells proliferate and survive for at least 6 weeks and were recovered from the blood, spleen, draining pancreatic lymph node, and most importantly, islets of NOD-scid/γcnull/A2 mice. Lymphocytes from type 1 diabetic patients preferentially infiltrate the islets of NOD-scid/γcnull/A2 mice. In contrast, PBMCs from nondiabetic HLA-A2–matched donors showed significantly less islet infiltration. Moreover, in mice that received PBMCs from type 1 diabetic patients, we identified epitope-specific CD8+ T cells among the islet infiltrates.

CONCLUSIONS

We show that insulitis is transferred to NOD-scid/γcnull/A2 mice that received HLA-A2–matched PBMCs from type 1 diabetic patients. In addition, many of the infiltrating CD8+ T cells are epitope-specific and produce interferon-γ after in vitro peptide stimulation. This indicates that NOD-scid/γcnull/A2 mice transferred with HLA-A2–matched PBMCs from type 1 diabetic patients may serve as a useful tool for studying epitope-specific T-cell–mediated responses in patients with type 1 diabetes.NOD mice develop spontaneous diabetes due to autoimmune destruction of pancreatic β-cells. These mice have served as a useful tool for understanding many aspects of type 1 diabetes (1,2). For example, the identification of certain pathogenic epitopes were originally found in NOD mice and subsequently observed in the blood of type 1 diabetic patients (3). The major shortcoming of these studies is their inability to evaluate the human autoreactive effector cells directly. Researchers have identified a number of pathogenic autoreactive epitopes of CD4+ and CD8+ T cells that recognize and result in β-cell killing. Epitopes for islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), insulin, and preproinsulin have been identified in the islets of NOD mice (4). T cells specific for epitopes from these proteins and other islet proteins, including islet amyloid polypeptide (IAPP), have been found in the blood of type 1 diabetic patients (5). At the same time, however, many of the findings in NOD mice have not directly translated to human type 1 diabetes. Importantly, a large number of therapies appear to “cure” diabetes in NOD mice, but these therapies have not readily translated to humans. Anti-CD3 antibody therapy, which is extremely effective in NOD mice (6,7), is less effective in patients (8,9). These immunomodulatory therapies still leave concerns about their effects, as discussed by Santamaria (10). Other immune therapies aimed at targeting B cells, such as anti-CD20 monoclonal antibody treatment, also offer short-term CD19+ B-cell depletion and partially preserve β-cell function (11).Development of humanized mice in which HLA-matched peripheral blood mononuclear cells (PBMCs) from type 1 diabetic patients are adoptively transferred into immune-deficient mice would provide a means of studying human immune cells without the restrictions inherent to human studies (12). Specifically, it would be possible to identify human autoreactive epitope-specific T cells that infiltrate the islets directly ex vivo. A small-animal model that recapitulates the clinical manifestations of type 1 diabetes would also assist in identifying novel therapeutic targets and in developing and testing novel immunotherapeutic agents. Furthermore, we would be able to investigate the mechanisms involved in disease pathogenesis of many other autoimmune diseases, especially those with disease-associated epitopes, shared between humans and mice (13,14).During the past several years, many investigators have used human hematopoietic stem cells (HSCs) for engraftment into immunodeficient mice (1517). These studies have attempted to develop a complete human immune system in a murine host. In many cases, successful engraftment and cell differentiation was observed. Most recently, investigators showed that functional human CD4+ and CD8+ T cells developed after being transferred into immune-deficient HLA transgenic mice and that these T cells demonstrated HLA-restricted responses (18). In contrast, our goal was not to recapitulate the entire autoimmune process; rather, we sought to develop a humanized mouse model that would be useful for identifying pre-existing autoreactive diabetogenic circulating T cells from type 1 diabetic patients that are important in the direct pathogenesis of type 1 diabetes.In NOD mice, β-cell–specific CD8+ T-cell clones are found in the peripheral blood and pancreatic islets (19). In patients with type 1 diabetes, epitope-specific T cells display T-lymphocyte cytotoxic activity toward human β-cells (20). Humans and mice also share many of the protein sequences of identified epitopes (21). Therefore, because it is likely that lymphocytes that have been exposed to islet antigens circulate within the blood of patients with type 1 diabetes, we adoptively transferred peripheral blood mononuclear cells (PBMCs) from HLA-A2–matched type 1 diabetic patients into transgenic NOD-scid/γcnull/HLA-A*0201 (NOD-scid/γcnull/A2) mice (22).

TABLE 1

Type 1 diabetic and nondiabetic haplotype-matched PBMC donors used in transfer experiments*
Patient IDAgeSexDiabetes duration (years)
Type 1 diabetes (n = 10)
 A1AdultF48
 A19AdultF41
 A21AdultM22
 A25AdultM12
 A27AdultM12
 A31AdultM25
 A33AdultM12
 A37AdultM22
 H82ChildF5
 H96ChildF13
NDD (n = 9)
 NDD1AdultMNA
 NDD2AdultMNA
 NDD3AdultFNA
 NDD4AdultFNA
 NDD5AdultMNA
 NDD6AdultFNA
 NDD7AdultMNA
 NDD8AdultFNA
 NDD9AdultMNA
Open in a separate windowID, identification; NA, not applicable.*All PBMC donors were HLA-A2 haplotype.After transfer of human PBMCs, we were able to show engraftment and islet cell infiltration of the PBMCs from the type 1 diabetic donors. Further, the islet-infiltrating cells were enriched in T cells that recognize diabetogenic epitopes and underwent additional expansion after transfer to a secondary recipient.  相似文献   

2.
3.
Adenopathy and extensive skin patch overlying a plasmacytoma is a very rare syndrome featuring a red-to-brown, violaceous skin patch along with a plasmacytoma. Only 11 case reports exist in the literature. Skin biopsies from the cutaneous patch overlying the plasmacytoma exhibit a dermal vascular hyperplasia with increased surrounding dermal mucin. Radiation therapy is used to treat and cure the plasmacytoma.Adenopathy and extensive skin patch overlying a plasmacytoma (AESOP) syndrome is a very rare constellation of findings seen in patients with a yet-to-be diagnosed solitary plasmacytoma.1,2 There are only 11 cases reported in the literature; the first report dates back to Sheinker in 1938 (3

TABLE 1

Summary of patients with AESOP syndrome ranked by ascending age
PATIENT/REFERENCEAGE/SEXLYMPH-ADENOPATHYNEUROPATHYPLASMA-CYTOMA SITEOTHER DISEASESMONOCLONAL IMMUNO-GLOBULIN (IG)TREATMENTOUTCOME AFTER TREATMENT
11,218/Male+-1st, 2nd, 3rd ribsNoneIgGRadiationCured
2134/Male++5th ribNoneNoneRadiationFavorable
31,239/Male++SternumNoneUnknownNoneDied 15 months later
41,242/Male++SternumPOEMS, Castleman’s diseaseIgA λChemoUnkown
51,243/Male++SkullCastleman’s diseaseIgG λSurgery and radiationCured
61,2,454/Male++ScapulaOsteolysisNoneRadiationNo follow up
71,2,558/Male++ClavicleNoneNoneRadiationFavorable
8264/Female--6th ribNoneIgG λNot knownNo follow up
9166/Male++6th ribPOEMSIgG λSurgeryDied 4.5 years later
10168/Female++SternumPOEMSIgG λSurgery and radiationFavorable
11173/Male+-SternumNoneNoneRadiationFavorable
Open in a separate window  相似文献   

4.
The Aliskiren in the Evaluation of Proteinuria in Diabetes (AVOID) trial demonstrated that adding aliskiren, an oral direct renin inhibitor, at a dosage of 300 mg/d to the highest approved dosage of losartan and optimal antihypertensive therapy reduces albuminuria over 6 mo among patients with type 2 diabetes, hypertension, and albuminuria. The cost-effectiveness of this therapy, however, is unknown. Here, we used a Markov model to project progression to ESRD, life years, quality-adjusted life years, and lifetime costs for aliskiren plus losartan versus losartan. We used data from the AVOID study and the Irbesartan in Diabetic Nephropathy Trial (IDNT) to estimate probabilities of progression of renal disease. We estimated probabilities of mortality for ESRD and other comorbidities using data from the US Renal Data System, US Vital Statistics, and published studies. We based pharmacy costs on wholesale acquisition costs and based costs of ESRD and transplantation on data from the US Renal Data System. We found that adding aliskiren to losartan increased time free of ESRD, life expectancy, and quality-adjusted life expectancy by 0.1772, 0.1021, and 0.0967 yr, respectively. Total expected lifetime health care costs increased by $2952, reflecting the higher pharmacy costs of aliskiren and losartan ($7769), which were partially offset by savings in costs of ESRD ($4860). We estimated the cost-effectiveness of aliskiren to be $30,500 per quality-adjusted life year gained. In conclusion, adding aliskiren to losartan and optimal therapy in patients with type 2 diabetes, hypertension, and albuminuria may be cost-effective from a US health care system perspective.Recent clinical trials have shown that reductions in albuminuria with agents that inhibit the renin-angiotensin-aldosterone system (RAAS; e.g., angiotensin-converting enzyme inhibitors or angiotensin receptor blockers), as defined by a urinary albumin-creatinine ratio (UACR) or urinary albumin excretion rate (UAER), can delay progression of renal disease in patients with hypertension and type 2 diabetes.15 Albuminuria is associated with fatal and nonfatal cardiovascular events and progression toward ESRD,1,2,6 and reductions in albuminuria have been widely used as surrogate markers of renoprotection.7,8The Aliskiren in the Evaluation of Proteinuria in Diabetes (AVOID) trial was a multicenter, randomized, double-blind study to assess the efficacy and safety of adding aliskiren, an oral direct renin inhibitor, at a dosage of 300 mg/d to the highest approved dosage of losartan (100 mg/d) and optimal antihypertensive therapy in patients with type 2 diabetes, hypertension, and albuminuria.9 At the end of 6 mo of follow-up, aliskiren 300 mg/d significantly reduced mean UACR by 20% (P = 0.0009) and overnight UAER by 18% versus placebo (P = 0.009). Although the AVOID trial demonstrated that adjunctive treatment with aliskiren 300 mg/d reduces albuminuria during 6 mo in these patients, it did not examine the potential long-term clinical and economic consequences of such treatment.The objective of this study was to evaluate, from the US health care system perspective, the potential cost-effectiveness of lifetime treatment with aliskiren 300 mg/d plus losartan 100 mg/d and optimal antihypertensive therapy (aliskiren plus losartan) versus lifetime treatment with losartan 100 mg/d and optimal antihypertensive therapy alone (losartan only) in patients with type 2 diabetes, hypertension, and albuminuria. We evaluated cost-effectiveness using a Markov model (Figure 1).10 The initial distribution of the population across disease states and probabilities of progression from microalbuminuria (MA) and early overt nephropathy (EON) to advanced overt nephropathy (AON) were from the AVOID trial (11Open in a separate windowFigure 1.Markov state transition model. Ovals represent health states. Patients are assumed to transition among states every 6 mo on the basis of transition probabilities in
Health StateCycles/AgesAliskiren + LosartanLosartan OnlySource or Reference
Beginning of CycleEnd of Cycle
MAEONFirst 6-mo cycle0.27160.3291AVOID
Subsequent cycles0.27740.3510AVOID
AONFirst 6-mo cycle0.01080.0266AVOID
Subsequent cycles0.00000.0000AVOID
DeathUS age-/gender-specific rates × 2.016,30
EONMAFirst 6-mo cycle0.25030.1446AVOID
Subsequent cycles0.00000.0000Assumption
AONFirst 6-mo cycle0.07940.0987AVOID
Subsequent cycles0.08040.0995AVOID
DeathUS age-/gender-specific rates × 4.416,30
AONMAFirst 6-mo cycle0.09720.0624AVOID
Subsequent cycles0.00000.0000Assumption
EONFirst 6-mo cycle0.35470.3754AVOID
Subsequent cycles0.00000.0000Assumption
DSCYear 1a0.03510.035116
Year 2a0.02300.023016
Year 3a0.02140.021416
Year 4+a0.01590.015916
ESRD-dialysisYear 1a0.01570.015716
Year 2a0.01040.010416
Year 3a0.01250.012516
Year 4+a0.01290.012916
DeathUS age-/gender-specific rates × 4.416,30
DSCESRD-dialysis0.32000.320016
DeathUS age-/gender-specific rates × 4.416,30
ESRD-dialysisESRD-transplant0.02530.025316
DeathAged 50 to 59 yr0.08570.085729
Aged 60 to 64 yr0.10390.103929
Aged 65 to 69 yr0.12060.120629
Aged 70 to 79 yr0.15640.156429
Aged ≥80 yr0.21220.212229
ESRD-transplantESRD-dialysis0.06090.060916
DeathAged 50 to 59 yr0.02700.027029
Aged 60 to 64 yr0.03760.037629
Aged 65 to 69 yr0.04990.049929
Aged 70 to 79 yr0.06150.061529
Aged ≥80 yr0.10810.108129
Open in a separate windowData are probability of transition from health state at beginning of cycle to health state at end of cycle. States are mutually exclusive (e.g., patients with AON and DSC transition to DSC state).aYears since entering state; excludes first 6-mo cycle.  相似文献   

5.
Primary Cutaneous Blastomycosis as a Cause of Acute Respiratory Distress Syndrom: Case Report and Literature Review     
Jason J. Emer  Joel B. Spear 《The Journal of clinical and aesthetic dermatology》2009,2(3):22-30
Blastomycosis is a fungal infection caused by Blastomyces dermatitidis. Exposure in endemic regions frequently occurs when spores in soil are disturbed and subsequently inhaled. Less commonly, primary cutaneous blastomycosis may follow after traumatic inoculation of the fungus into the skin. Most patients infected with blastomycosis are asymptomatic, but an unfortunate small number present with fulminant disease. Rarely, the infection can affect organs, such as the skin, bone, or genitourinary system. In a small percentage of cases, blastomycosis may cause acute respiratory distress syndrome, which is associated with a very high mortality rate. Increased survival rates have been shown when the clinician has a high index of suspicion and facilitates rapid evaluation and initiation of the appropriate therapy. We present a rare case of a patient presenting with primary cutaneous blastomycosis that progressed to disseminated disease causing acute respiratory distress syndrome. High clinical suspicion, prompt diagnostic testing, and therapy with amphotericin B, confirmed the diagnosis and resulted in a swift recovery.Blastomycosis is an infection caused by the fungus Blastomyces dermatitidis, which is endemic to areas of the United States and Canada including the Great Lakes Region and the Mississippi and Ohio River Valleys.1,2 Exposure occurs in moist wooded areas during outdoor activities when fungal microhabitats existing in soil are disturbed. Inhaled conidia can result in a primary lung infection, which may then become disseminated.3,4 Pulmonary disease is the most common manifestation of blastomycosis with isolated lung disease occurring in 60 to 75 percent of infected people.5 In the remaining group, dissemination to skin, bone, genitourinary, and other organ systems can be seen.4 In fewer than 10 percent of cases, blastomycosis can progress to acute respiratory distress syndrome (ARDS) with dyspnea, tachypnea, and hypoxemia as systemic manifestations.612Palmer and McFadden reported the first case of disseminated blastomycosis causing ARDS in 1968.13 Since then, few studies have identified disseminated blastomycosis as a cause of ARDS (14 Thus, early suspicion of disseminated disease is especially helpful in endemic areas since rapid institution of therapy can improve morbidity and mortality.

Table 1

Case reports of blastomycosis causing acute respiratory distress syndrome in the literature
AuthorYearNo. of PatientsSymptomsVentilationInitial TreatmentUltimate TreatmentSurvival
Palmer et al1319681fatigue, general malaise, cough with sputum, shortness of breath, fever0 of 1penicillinamphotericin B0/1 (0%)
Griffith et al1119791fever, malaise, dyspnea, cough1 of 1penicillin + gentamicin + methylprednisolone + digoxinamphotericin B + hydroxystilbamidine1/1 (100%)
Lockridge et al819791pleuritic chest pain, fever, shortness of breath, cough with sputum production1 of 1aspirinmultiple antibiotics + amphotericin B + high-dose steroids0/2 (0%)
Evans et al719822fever, cough, right upper lobe pneumonia, dyspnea1 of 21) isoniazid + ethambutol 2) cephalothin + erythromycin1) amphotericin B + hydroxystilbamidine + highdose methylprednisolone 2) cefazolin + tobramycin + erythromycin + isoniazid + sulfamethoxazole + methylprednisolone0/2 (0%)
Atkinson et al9019831fever, cough, dyspnea1 of 1cephalothinmultiple antibitoics + isoniazide + highdose methylprednisolone0/1 (0%)
Thiele et al9119841left ear pain and discharge1 of 1penicillin + chloramphenicol + tobramycin + nafcillinisoniazid + rifampin + ethambutol + ketoconazole + amphotericin B0/1 (0%)
Skillrud et al6019851dyspnea on exertion, fatigue, shaking chills, nearsyncopal episode1 of 1cefazolin + cefoxitin + erythromycin + tobramycin + clindamycinamphotericin B1/1 (100%)
Unger9219861cough, fever, bloody sputum1 of 1multiple antibioticsamphotericin B1/1 (100%)
MacDonald9319901cough, dyspnea1 of 1amphotericin Bamphotericin B1/1 (100%)
Renston et al9419921fever, chills, sweats, cough, pain and swelling of right knee1 of 1multiple antibiotics + antituberculosis drugsamphotericin B1/1 (100%)
Meyer et al6199310cough, chills, fever, dyspnea9 of 10multiple antibioticsamphotericin B + ketoconazole + rifampin5/10 (50%)
Craft5919951fever, cough, chills, hemoptysis, rightsided pleuritic chest pain1 of 1multiple antibioticsfluconazole + rifampin + pyrazinamide + amikacin + ethambutol + vancomycin + isoniazid + amphotericin B + itraconazole1/1 (100%)
Mukkamala et al9519972cough with sputum production, night sweats, weight loss, headache, backache, shortness of breath2 of 2amphotericin Bamphotericin B0/2 (0%)
Mundey et al1020011fever, chills, cough without sputum, weakness, diarrhea1 of 1ampicillin/sulbactam + ceftazidime + gentamicinazithromycin + amphotericin B0/1 (0%)
Amini9620071fever, cough0 of 1piperacillin/tazobactum + levofloxacinamphotericin B0/1 (0%)
Gauthier et al4820076fever, cough, skin lesions0 of 6immunosuppressive agents + antibiotics + antiviral agentsamphotericin B, itraconazole, voriconazole2/6 (33%)
Watts et al9720071suprapubic pain with inability to urinate, fever, pruritic lesions of arms, face, and trunk, right ankle pain1 of 1gatifloxacinvancomycin + piperacillin/tazobactum +ciprofloxacin + amphotericin B0/1 (0%)
Open in a separate windowBlastomycosis may seldom present as a primary cutaneous lesion after traumatic inoculation from laboratory or autopsy exposure, animal bites or scratches, and outdoor trauma.15,16 A total of 22 cases of cutaneous inoculation blastomycosis were compiled for a major article, reviewing literature from 1903 to 2002.17 The clinical presentation of infected patients is variable and symptoms of infection may be absent, chronic, acute, or even fulminant. Primary cutaneous blastomycosis is often mistaken for other cutaneous entities such as keratoacanthoma, squamous cell carcinoma, tuberculosis, tertiary syphilis, leprosy, or bacterial pyoderma.18We describe a rare case of a patient with an unhealed cutaneous lesion who underwent surgical debridement and postoperatively developed life-threatening disseminated blastomycosis progressing to ARDS. With a high index of clinical suspicion, rapid diagnosis, and prompt therapy with amphotericin B (AmB), the patient recovered.  相似文献   

6.
Association between Body Composition and Frailty among Prevalent Hemodialysis Patients: A US Renal Data System Special Study     
Kirsten L. Johansen  Lorien S. Dalrymple  Cynthia Delgado  George A. Kaysen  John Kornak  Barbara Grimes  Glenn M. Chertow 《Journal of the American Society of Nephrology : JASN》2014,25(2):381-389
Studies of frailty among patients on hemodialysis have relied on definitions that substitute self-reported functioning for measures of physical performance and omit weight loss or substitute alternate criteria. We examined the association between body composition and a definition of frailty that includes measured physical performance and weight loss in a cross-sectional analysis of 638 adult patients receiving maintenance hemodialysis at 14 centers. Frailty was defined as having three of following characteristics: weight loss, weakness, exhaustion, low physical activity, and slow gait speed. We performed logistic regression with body mass index (BMI) and bioelectrical impedance spectroscopy (BIS)-derived estimates of intracellular water (ICW), fat mass, and extracellular water (ECW) as the main predictors, and age, sex, race, and comorbidity as covariates. Overall, 30% of participants were frail. Older age (odds ratio [OR], 1.31 per 10 years; 95% confidence interval [95% CI], 1.14 to 1.50), diabetes (OR, 1.65; 95% CI, 1.13 to 2.40), higher fat mass (OR, 1.18; 95% CI, 1.02 to 1.37), and higher ECW (OR, 1.33; 95% CI, 1.20 to 1.47) associated with higher odds of frailty. Higher ICW associated with lower odds of frailty (OR, 0.80 per kg; 95% CI, 0.73 to 0.87). The addition of BMI data did not change the area under the receiver operating characteristics curve (AUC; AUC=0.66 versus 0.66; P=0.71), but the addition of BIS data did change the AUC (AUC=0.72; P<0.001). Thus, individual components of body composition but not BMI associate strongly with frailty in this cohort of patients receiving hemodialysis.Patients on dialysis frequently experience protein energy wasting or loss of protein mass and energy stores, which is likely multifactorial.1 It has recently been appreciated that the same disorders that underlie protein energy wasting as well as muscle wasting itself are also commonly associated with frailty.24 Although frailty is generally considered to be a geriatric syndrome, individuals with chronic diseases, such as CKD, may be at risk for premature frailty.5,6 In fact, as many as two thirds to three quarters of patients new to dialysis may be frail by definitions that rely on patient self-report of physical functioning and omit weight loss or substitute alternate criteria for wasting (6,7

Table 1.

Definitions of frailty adapted from the Cardiovascular Health Study definition
Components of FrailtyUSRDS DMMS Wave 2 (Incident Dialysis)6CDS (Incident Dialysis)7REXDP (Prevalent Hemodialysis)8NHANES (CKD)9Seattle Kidney Study (CKD Stages 1–4)28
Slowness/weaknessRand-36 Physical Function Scale score<75SF-12 Physical Function score<75Slowness: gait speed over 6 m using cutpoints that correspond to the same speed as the cutpoints from the 15-ft walk in the CHSSlowness: gait speed over 8 ft with lowest quintile adjusted for sexSlowness: walking pace assessed over a 4-m course using cutpoints that correspond to the same speed as the cutpoints from the 15-ft walk in the CHS
The following items are about activities that you might do during a typical day: does your health now limit you in these activities? If so, how much?Weakness: patients were asked to stand up and sit down five times; the time for the slowest quartile of the SPPB chair stand based on community-dwelling elderly cohorts was used to define frailtyWeakness: based on self-report and defined as present if participants answered some difficulty, much difficulty, or unable to do when asked how much difficulty they have lifting or carrying something as heavy as 10 lbs (like a sack of potatoes or rice)Weakness: Grip strength using the same absolute cutoffs as in the CHS
Vigorous activities, such as running, lifting heavy objects, or participating in strenuous sports
Moderate activities, such as moving a table, pushing a vacuum cleaner, bowling, or playing golf
Lifting or carrying groceries
Climbing several flights of stairs
Climbing one flight of stairs
Bending, kneeling, or stooping
Walking more than 1 mi
Walking several blocks
Walking 1 block
Bathing or dressing yourself
Poor endurance/exhaustionRand-36 Vitality Scale score<55SF-12 Vitality score<55SF-36 Vitality score<55Defined as present if participants answered some difficulty, much difficulty, or unable to do when asked how much difficulty they have walking from one room to the other on the same levelSF-36 Vitality score<37.5
How much of the time during the last 30 days?
Did you feel worn out?
Did you feel tired?
Did you have a lot of energy?
Did you feel full of pep?
Physical inactivityHow often do you exercise (do physical activity during your leisure time)?Lowest quintile based on age- and sex-specific population norms for the Human Activity ProfilePatients who reported no activity beyond self-care and activities required for living were considered inactiveCompared with most (men/women) your age, would you say that you are more active, less active, or about the same?Self-reported exercise less than one time per week
Daily or almost dailyPatients answering less active were classified as inactive
Four to five times a week
Two to three times a week
About one time a week
Less than one time a week
Almost never or never
Patients answering almost never or never were classified as inactive
Unintentional weight loss or shrinkageUndernourished or cachectic (malnourished) as assessed by data abstractorNot includedBMI≤18.5 kg/m2BMI≤18.5 kg/m2Self-reported ≥10-lb unintentional weight loss in past 6 months
Open in a separate windowSee ref2 for Cardiovascular Health Study. USRDS, US Renal Data System; DMMS, Dialysis Morbidity and Mortality Study; CDS, Comprehensive Dialysis Study; REXDP, Renal Exercise Demonstration Project; NHANES, National Health and Nutrition Examination Survey; CHS, Cardiovascular Health Study; SPPB, short physical performance battery.Low Quételet’s (body mass) index (BMI), expressed in kilograms of body weight divided by height squared, has been substituted for the weight loss criterion of the frailty construct in several studies.8,9 However, BMI is a nonspecific metric of body composition, because the body weight component could reflect adipose tissue or intracellular (muscle) or extracellular (edema) water. The most commonly used definition of frailty, which includes direct measures of gait speed and grip strength (rather than self-reported functioning) as well as weight loss, exhaustion, and level of physical activity,2 has only recently been applied in an ESRD population,10 and the association between frailty and body composition has not been examined systematically in this population.11In this investigation, we sought to determine the extent to which body composition was associated with frailty in a prevalent hemodialysis cohort. We hypothesized that intracellular water (ICW) estimated by bioelectrical impedance spectroscopy (BIS) would be inversely associated and fat mass would be directly associated with frailty in a cohort of prevalent dialysis patients but that BMI would not be associated.  相似文献   

7.
Estimated GFR Reporting Influences Recommendations for Dialysis Initiation     
K. Scott Brimble  Rajnish Mehrotra  Marcello Tonelli  Carmel M. Hawley  Clare Castledine  Stephen P. McDonald  Vicki Levidiotis  Azim S. Gangji  Darin J. Treleaven  Peter J. Margetts  Michael Walsh 《Journal of the American Society of Nephrology : JASN》2013,24(11):1737-1742
Automated reporting of estimated GFR (eGFR) with serum creatinine measurement is now common. We surveyed nephrologists in four countries to determine whether eGFR reporting influences nephrologists’ recommendations for dialysis initiation. Respondents were randomly allocated to receive a survey of four clinical vignettes that included either serum creatinine concentration only or serum creatinine and the corresponding eGFR. For each scenario, the respondent was asked to rank his or her likelihood of recommending dialysis initiation on a modified 8-point Likert scale, ranging from 1 (“definitely not”) to 8 (“definitely would”). Analysis of the 822 eligible responses received showed that the predicted likelihood of recommending dialysis increased by 0.55 points when eGFR was reported (95% confidence interval, 0.33 to 0.76), and this effect was larger for eGFRs >5 ml/min per 1.73 m2 (P<0.001). Subgroup analyses suggested that physicians who had been in practice ≥13 years were more affected by eGFR reporting (P=0.03). These results indicate that eGFR reporting modestly increases the likelihood that dialysis is recommended, and physicians should be aware of this effect when assessing patients with severe CKD.Serum creatinine concentration is commonly used to calculate the estimated GFR (eGFR). The eGFR can identify patients with significant CKD even when the serum creatinine level is not markedly abnormal. As such, clinical practice guidelines recommend using eGFR, rather than serum creatinine, to identify, risk stratify, and manage patients with CKD.13Automatically reporting eGFR when serum creatinine is measured is becoming commonplace to enhance the identification of patients with CKD.4 Observational studies have demonstrated that eGFR reporting improved CKD detection and increased referrals of patients with early CKD to nephrologists.58 Furthermore, eGFR reporting is associated with increased appropriate treatment in patients with CKD.9 However, the effects of eGFR reporting on nephrologists’ practices are uncertain.Whether eGFR reporting influences the decision to initiate dialysis is important to patients, physicians, and health care systems. We conducted a randomized survey to determine whether eGFR reporting influences the likelihood that a nephrologist will recommend the initiation of dialysis. The survey randomly allocated half of the recipients to receive only serum creatinine information for clinical vignettes of CKD and half to receive both serum creatinine and eGFR information.Of the 5576 surveys distributed, 579 were returned to the sender, leaving 4997 potential respondents. Of the 938 surveys (19%) that were returned, 822 (16%) were analyzed (Figure 1). Respondents were similar between groups (Open in a separate windowFigure 1.Flow of surveys and respondents throughout the study.

Table 1.

Respondent characteristics
CharacteristicCreatinine (n=392)eGFR (n=430)
Years in practice13 (7–23)13 (6–23)
Country
 Australia76 (19.4)80 (18.6)
 Canada58 (14.8)61 (14.2)
 New Zealand16 (4.1)14 (3.3)
 United Kingdom33 (8.4)41 (9.5)
 United States185 (47.2)209 (48.6)
 Missing24 (6.1)25 (5.8)
Clinical practice
 CKD/ESRD275 (70.2)287 (66.7)
 Transplant11 (2.8)23 (5.4)
 Both82 (20.9)95 (22.1)
 Missing24 (6.1)25 (5.8)
Academic practice
 Yes222 (56.6)252 (62.2)
 No144 (36.7)153 (35.6)
 Missing26 (6.6)25 (5.8)
Open in a separate windowData are presented as the median (interquartile range) or n (%).  相似文献   

8.
Comparisons between Novel Oral Anticoagulants and Vitamin K Antagonists in Patients with CKD     
Ziv Harel  Michelle Sholzberg  Prakesh S. Shah  Katerina Pavenski  Shai Harel  Ron Wald  Chaim M. Bell  Jeffrey Perl 《Journal of the American Society of Nephrology : JASN》2014,25(3):431-442
Novel oral anticoagulants (NOACs) (rivaroxaban, dabigatran, apixaban) have been approved by international regulatory agencies to treat atrial fibrillation and venous thromboembolism in patients with kidney dysfunction. However, altered metabolism of these drugs in the setting of impaired kidney function may subject patients with CKD to alterations in their efficacy and a higher risk of bleeding. This article examined the efficacy and safety of the NOACs versus vitamin K antagonists (VKAs) for atrial fibrillation and venous thromboembolism in patients with CKD. A systematic review and meta-analyses of randomized controlled trials were conducted to estimate relative risk (RR) with 95% confidence interval (95% CIs) using a random-effects model. MEDLINE, Embase, and the Cochrane Library were searched to identify articles published up to March 2013. We selected published randomized controlled trials of NOACs compared with VKAs of at least 4 weeks’ duration that enrolled patients with CKD (defined as creatinine clearance of 30–50 ml/min) and reported data on comparative efficacy and bleeding events. Eight randomized controlled trials were eligible. There was no significant difference in the primary efficacy outcomes of stroke and systemic thromboembolism (four trials, 9693 participants; RR, 0.64 [95% CI, 0.39 to 1.04]) and recurrent thromboembolism or thromboembolism-related death (four trials, 891 participants; RR, 0.97 [95% CI, 0.43 to 2.15]) with NOACs versus VKAs. The risk of major bleeding or the combined endpoint of major bleeding or clinically relevant nonmajor bleeding (primary safety outcome) (eight trials, 10,616 participants; RR 0.89 [95% CI, 0.68 to 1.16]) was similar between the groups. The use of NOACs in select patients with CKD demonstrates efficacy and safety similar to those with VKAs. Proactive postmarketing surveillance and further studies are pivotal to further define the rational use of these agents.The introduction of the novel oral anticoagulants (NOACs) rivaroxaban (Xarelto, Bayer, Munich Germany), apixaban (Elequis, Pfizer, Bristol-Myers Squibb), and dabigatran (Pradax/Pradaxa/Prazaxa, Boehringer Ingelheim) as alternatives to vitamin K antagonists (VKAs) has been met with enthusiasm among clinicians. These agents are currently available for prophylaxis and treatment of venous thromboembolism (VTE) and for prophylaxis of stroke and systemic thromboembolism in the setting of atrial fibrillation. Furthermore, they have demonstrated similar or greater efficacy and safety in relation to conventional anticoagulants in large trials.16NOACs differ from traditional oral VKAs mechanistically and pharmacokinetically. Dabigatran directly inhibits the final effector of coagulation, thrombin (factor IIa), while rivaroxaban and apixaban directly inhibit the rate-limiting step of coagulation, factor Xa activation. Thrombin and factor Xa are targeted by the NOACs for anticoagulant therapy given their roles in clot formation.7,8 Advantages of the NOACs include their rapid onset of action, shorter half-lives, lack of requirement for regular laboratory monitoring, and absence of food interactions compared with VKAs.Although the NOACs differ in their degree of kidney excretion, their elimination is differentially impaired with worsening kidney function, with accumulating levels predisposing patients to an increased risk of bleeding events.9,10 CKD is increasing in prevalence and is associated with an increased risk of atrial fibrillation and venous thrombosis, both of which are indications for NOAC use.11,12 In North America, these agents have been approved by the US Food and Drug Administration and Health Canada for use in patients with varying degrees of kidney dysfunction. However, these agencies have extrapolated the efficacy and safety data from the NOAC trials and approved dabigatran and rivaroxaban for use in patients with more severe CKD, despite the exclusion of such patients from the trials (1318 Serious bleeding has been reported with the NOACs in patients with CKD.19,20

Table 1.

Regulatory agency recommendations for NOACs in patients with CKD
AgencyDrug
ApixabanDabigatranRivaroxaban
Health CanadaAtrial fibrillation and VTE:Atrial fibrillation:Atrial fibrillation:
 CrCl=30–50 ml/min: No dose adjustment required; i.e., 5 mg orally twice daily except for: CrCl=30–50 ml/min: 150 mg orally twice daily CrCl=30–49 ml/min: 15 mg orally once daily
 Cr>132 µmol/L Contraindicated if CrCl<30 ml/min Contraindicated if CrCl<30 ml/min
 Age>80 yr
  Weight <60 kgVTE:
 (if any of the above, use 2.5 mg orally twice daily) CrCl=30–49 ml/min: 15 mg orally twice daily×21 d, then 20 mg orally once daily
 Contraindicated if CrCl<25 ml/min Contraindicated if CrCl<30 ml/min
US Food and Drug AdministrationAtrial fibrillation and VTE:Atrial fibrillation and VTE:Atrial fibrillation:
 CrCl=30–50 ml/min: No dose adjustment required; i.e., 5 mg orally twice daily except for: CrCl>30 ml/min: 150 mg orally twice daily CrCl=30–49 ml/min: 15 mg orally once daily
 CrCl=15–30 ml/min: 75 mg orally twice daily CrCl=15–29 ml/min: 15 mg orally once daily
 Cr >132 µmol/L Contraindicated if CrCl<15 ml/min
 Age>80 yr Contraindicated if CrCl<15ml/min
 Weight<60 kgVTE:
 (if any of the above, use 2.5 mg orally twice daily) CrCl=30–49 ml/min: 15 mg orally twice daily×21 d then 20 mg orally once daily
 Contraindicated if CrCl<15 ml/min Contraindicated if CrCl<30 ml/min
Open in a separate windowThe increasing popularity of the NOACs, combined with the lack of a specific antidote in the face of hemorrhage, creates a potential “perfect storm” for adverse bleeding events in patients with CKD. As such, it is important to determine the efficacy and safety of these agents among patients with CKD in order to guide the rational use of these agents. We carried out a systematic review and meta-analysis comparing the efficacy and bleeding risk with the use of NOACs compared with conventional VKA therapy in patients with CKD.  相似文献   

9.
Evaluation of a Topical Anti-inflammatory/Antifungal Combination Cream in Mild-to-moderate Facial Seborrheic Dermatitis: An Intra-subject Controlled Trial Examining Treated vs. Untreated Skin Utilizing Clinical Features and Erythema-directed Digital Photography     
Federica Dall’Oglio  Aurora Tedeschi  Vincenzo Guardabasso  Giuseppe Micali 《The Journal of clinical and aesthetic dermatology》2015,8(9):33-38
  相似文献   

10.
Lichenoid Reactions in Association with Tumor Necrosis Factor Alpha Inhibitors: A Review of the Literature and Addition of a Fourth Lichenoid Reaction     
Morgan McCarty  Amy Basile  Brooke Bair  David Fivenson 《The Journal of clinical and aesthetic dermatology》2015,8(6):45-49
  相似文献   

11.
Linkage Disequilibrium Mapping of the Replicated Type 2 Diabetes Linkage Signal on Chromosome 1q     
《Diabetes》2009,58(7):1704-1709

OBJECTIVE

Linkage of the chromosome 1q21–25 region to type 2 diabetes has been demonstrated in multiple ethnic groups. We performed common variant fine-mapping across a 23-Mb interval in a multiethnic sample to search for variants responsible for this linkage signal.

RESEARCH DESIGN AND METHODS

In all, 5,290 single nucleotide polymorphisms (SNPs) were successfully genotyped in 3,179 type 2 diabetes case and control subjects from eight populations with evidence of 1q linkage. Samples were ascertained using strategies designed to enhance power to detect variants causal for 1q linkage. After imputation, we estimate ∼80% coverage of common variation across the region (r 2 > 0.8, Europeans). Association signals of interest were evaluated through in silico replication and de novo genotyping in ∼8,500 case subjects and 12,400 control subjects.

RESULTS

Association mapping of the 23-Mb region identified two strong signals, both of which were restricted to the subset of European-descent samples. The first mapped to the NOS1AP (CAPON) gene region (lead SNP: rs7538490, odds ratio 1.38 [95% CI 1.21–1.57], P = 1.4 × 10−6, in 999 case subjects and 1,190 control subjects); the second mapped within an extensive region of linkage disequilibrium that includes the ASH1L and PKLR genes (lead SNP: rs11264371, odds ratio 1.48 [1.18–1.76], P = 1.0 × 10−5, under a dominant model). However, there was no evidence for association at either signal on replication, and, across all data (>24,000 subjects), there was no indication that these variants were causally related to type 2 diabetes status.

CONCLUSIONS

Detailed fine-mapping of the 23-Mb region of replicated linkage has failed to identify common variant signals contributing to the observed signal. Future studies should focus on identification of causal alleles of lower frequency and higher penetrance.Genome-wide association (GWA) analysis has provided a powerful stimulus to the discovery of common variants influencing type 2 diabetes risk, and, to date, ∼20 susceptibility loci have been identified with high levels of statistical confidence (1). However, these known variants account for only a small proportion of the inherited component of disease risk (probably <10%), and the molecular basis of the majority of the genetic predisposition to type 2 diabetes has yet to be established (1).The success of the GWA approach contrasts with the slow progress that characterized previous efforts to map susceptibility loci through genome-wide linkage (2). However, now that many of the common variants of largest effect have been identified (in European-descent populations at least), there are cogent reasons to revisit regions previously identified through genome-wide linkage. First, variants within the genomic intervals representing replicated linkage signals can be considered to have raised prior odds for a susceptibility effect, and this information can be used to prioritize GWA signals (particularly those with only modest evidence of association) for targeted replication. Second, genuine linkage signals are likely to be driven by causal variants—particularly low-frequency SNPs or copy number variants not captured by the commodity GWA arrays—with effect sizes larger than those currently detectable by GWA (3). Because alleles with these characteristics will have a more marked impact on individual disease predisposition than the common variants found by GWA, identification of causal variants underpinning replicated linkage signals should accelerate efforts to obtain better predictors of disease (4).For type 2 diabetes, there appears to be only limited overlap between the regions identified by genome-wide linkage and those revealed by GWA (5). Although the discovery of TCF7L2 was prompted by a search for causal variants within a region of replicated type 2 diabetes linkage, neither the common variants in TCF7L2 nor those in HHEX and IDE (a second nearby GWA signal) account for that linkage signal (6). Thus, the discovery of TCF7L2 reflects either serendipity or the co-localization of common and rare causal variants in the same locus—the former driving the association and the latter the linkage. Similarly, whereas common variants in HNF4A have been reported to explain the chromosome 20 linkage signals seen in Finns and Ashkenazim (7,8), these associations have proved difficult to replicate (9).Chromosome 1q (in particular the 30-Mb stretch adjacent to the centromere) ranks alongside the regions on chromosomes 10 and 20 as among the strongest in terms of the replicated evidence for genome-wide linkage to type 2 diabetes. Linkage has been reported in samples of European (U.K., French, Amish, Utah), East Asian (Chinese, Hong Kong), and Native American (Pima) origin (summarized in Supplementary Table 1, which is available in the online-only appendix at http://diabetes.diabetesjournals.org/cgi/content/full/db09-0081/DC1; ref. (2). The region concerned is gene rich and contains a disproportionate share of excellent biological candidates (2). The homologous region has also emerged as a diabetes susceptibility locus from mapping efforts in several well-characterized rodent models (1013).The International 1q Consortium represents a coordinated effort by the groups with the strongest evidence for 1q linkage to identify variants causal for that signal. Here, we report efforts to map causal variants using a custom linkage-disequilibrium (LD) mapping approach, predominantly based around common SNP variants, applied to a well-powered set of multiethnic samples.To improve power, ascertainment of type 2 diabetes cases for this study aimed to enrich for 1q causal alleles through 1) a focus on populations and samples that had shown 1q linkage; 2) selection for positive family history; and 3) for some samples, preferential recruitment on the basis of patterns of identity by descent sharing in the 1q region. For each set of case subjects, we selected a control sample of individuals from the same population. Details of the recruitment have been reported previously (14) and are summarized in the supplementary material available in the online appendix. In all, the case-control part of the study included 2,198 samples (1,000 case subjects, 1,198 control subjects) of European descent, 281 (140 case subjects, 141 control subjects) of East Asian origin, and 285 (144 case subjects, 141 control subjects) who were Native American (Pima) (supplementary Table 2). We also included a small sample of individuals of African American origin (242 case subjects, 173 control subjects) and an additional 599 Pima individuals (520 affected, 79 nondiabetic after age 45 years) from 255 families who, after combination with the Pima case-control set, were used for family-based association analyses.These samples were submitted to dense-map SNP typing of the core region of interest (from 147.0 to 169.7 Mb [Build 35]) using a series of 1,536-plex BeadArray designs (Golden Gate, Illumina, San Diego, CA). Design of these arrays was contemporaneous with development of dbSNP and HapMap (15). Thus, whereas the first arrays were LD-agnostic and compiled using genomic localization as the primary consideration for inclusion, subsequent arrays used LD information from the CEU (European ancestry) and CHB/JPT (Asian ancestry) components of HapMap to guide SNP selection and maximize coverage of the region. In all, we designed assays for 6,023 SNPs, of which 5,290 provided reliable data in all populations after passing through our extensive quality control (see the supplementary material). We estimate that after imputation (using the appropriate set of HapMap data as a reference), coverage of the region (minor allele frequency >0.05; r2 > 0.8) reaches ∼80% in the European and ∼72% in the East Asian samples. Coverage is harder to estimate (and imputation likely to be less valuable) in Pima and African American samples, since reference data from these populations are not available, although we estimate ∼49% coverage in West Africans based on YRI data.Genotyping quality was generally good, with over 91% of SNPs passing quality control in each population (see the supplementary material) and <0.45% of SNPs failing (P < 10−4) tests of within-sample Hardy-Weinberg equilibrium. Significant departures from expectation in the distributions of test statistics observed in the Amish and Pima samples (as revealed by QQ plots; see supplementary material) likely reflect residual relatedness between subjects from those populations. We adjusted for this (and any population stratification effects) through genomic control methods (16). Association analyses treated each study as a separate stratum and used standard meta-analysis approaches to deliver estimates of joint effect size and statistical significance (see supplementary material). A series of nested meta-analyses were performed including 1) European-descent samples only (“4-way”); 2) non–African-descent samples only (“7-way”); and 3) all samples (“8-way”).Under an additive model with allele frequency of 0.25, our sample provides ∼80% power to detect per-allele odds ratios (ORs) of >1.36 (“8-way”) or >1.43 (“4-way”) for α = 5 × 10−6. Given that the region covers ∼1% of the genome, we consider this a reasonable benchmark for “region-wide” significance (equivalent to consensus genome-wide thresholds of 5 × 10−8). These power calculations are conservative: given the case ascertainment enrichment strategies used in this study, we would expect to detect variants with population-level effects in the 1.2–1.3 range. Under reasonable assumptions (three independent alleles contributing to a linkage signal with a locus-specific sibling relative risk of ∼1.15), we can expect the effect size of the variants we were seeking to detect (i.e., those responsible for the 1q linkage) to be substantially greater than this (e.g., allelic OR 1.6 for a variant with risk allele frequency of 25%). Our study was therefore well powered to detect putatively causal alleles within the European and/or combined datasets.Across these analyses, none of the SNPs showed an association with type 2 diabetes that withstood genome-wide correction (P < 5 × 10−8) (17). However, two clusters of SNPs showed association signals that approached or exceeded “region-wide” significance thresholds. The first of these, involving rs7538490 and nearby SNPs, mapped to a 51.4-kb interval (at ∼160.35 Mb) within the first intron of NOS1AP (nitric oxide synthase 1 [neuronal] adaptor protein) with an estimated per allele OR (in the 4-way, European-only analysis) of 1.38 (95% CI 1.21–1.57, P = 1.4 × 10−6, additive model, Fig. 1). Rs7538490 lies ∼5.6 kb from one of the SNPs (rs10494366) previously shown to influence cardiac repolarization and QT interval (18): the two SNPs are in modest LD (r2 = 0.47 in HapMap CEU), and rs10494366 shows some evidence for association with type 2 diabetes (P = 3.1 × 10−4) in the same 4-way meta-analysis.

TABLE 1

Association results for rs7538490 in the NOS1AP gene
Case subjects (n)Control subjects (n)Risk allele frequency in case subjectsRisk allele frequency in control subjectsAdditive model
OR (95% CI)P
U.K.4434430.310.241.38 (1.13–1.69)1.6 × 10−3
French2192390.340.291.21 (0.92–1.60)0.16
Utah1901610.300.201.68 (1.19–2.36)2.7 × 10−3
Amish*1473470.430.361.36 (0.97–1.91)0.071
Meta-analysis: 4 European descent populations9991,1901.38 (1.21–1.57)1.4 × 10−6
Shanghai77770.440.421.11 (0.71–1.72)0.66
Hong Kong63640.570.551.10 (0.68–1.78)0.70
Pima*1441410.460.441.09 (0.76–1.57)0.62
Meta-analysis: 7 non-African populations1,2831,4721.31 (1.17–1.46)4.3 × 10−6
African American2421730.250.280.86 (0.62–1.19)0.37
Meta-analysis: 8 1qC populations1,5251,6451.24 (1.12–1.39)5.4 × 10−5
Replication samples
    Independent WTCCC sample1,4952,9380.290.290.97 (0.88–1.07)0.57
    W2C vs. 58BC4721,9920.270.271.00 (0.85–1.17)0.93
    UKT2DGC3,9324,8180.290.281.03 (0.97–1.10)0.18
    Diabetes Genetics Initiative1,4641,4670.260.261.01 (0.87–1.16)0.82
    FUSION1,1611,1740.300.271.13 (0.97–1.29)0.063
Meta-analysis: all replication samples8,52412,3891.03 (0.98–1.07)0.29
Meta-analysis: all European descent populations9,52313,5791.06 (1.01–1.10)0.014
Meta-analysis: all data10,04914,0341.05 (1.01–1.10)0.015
Open in a separate window*P value for Amish and Pima was adjusted using estimated lambda for genomic control. WTCCC, Wellcome Trust Case Control Consortium.Open in a separate windowFIG. 1.Single-point type 2 diabetes associations within the 1q region. This plot shows the “4-way” (European-descent samples only) meta-analysis using the additive model (Cochran-Armitage trend test). Directly typed SNPs are shown in orange and imputed SNPs in blue. The pale blue track (and secondary y-axis) represents recombination rates (for HapMap CEU) across the region. Blue diamonds represent the strongest association P value in the two regions taken forward for replication. In the case of the PKLR/ASH1L region, the strongest association was seen for a dominant model (the equivalent additive model result is denoted with the red diamond). Only a small subset of genes within the region is denoted on the gene track.The second signal includes ∼10 SNPs in a 220-kb region of extensive LD at ∼152.1 Mb. This region includes the coding sequences of the genes encoding liver pyruvate kinase (PKLR) and ash1 (absent, small, or homeotic)-like (Drosophila) (ASH1L) among others. At the lead SNP (rs11264371), the estimated OR for the 4-way analysis was 1.36 (1.18–1.58) (P = 3.5 × 10−5) under the additive model. The effect size and significance were marginally greater (1.48 [1.18–1.76], P = 1.0 × 10−5), under a dominant model (Fig. 1).

TABLE 2

Association results for rs11264371 in the PKLR/ASH1L region
Case subjects (n)Control subjects (n)Risk allele frequency in case subjectsRisk allele frequency in control subjectsAdditive model
OR (95% CI)P*
U.K.4444430.250.201.33 (1.07–1.66)0.012
French2192440.250.231.17 (0.86–1.59)0.32
Utah1901620.260.211.34 (0.94–1.92)0.095
Amish*1473490.250.171.83 (1.19–2.81)5.8 × 10−3
Meta-analysis: 4 European descent populations1,0001,1981.36 (1.18–1.58)3.5 × 10−5
Shanghai77770.770.701.47 (0.87–2.49)0.15
Hong Kong63630.660.730.73 (0.42–1.23)0.24
Pima*1441410.580.600.91 (0.64–1.29)0.64
Meta-analysis: 7 non-African populations1,2841,4791.24 (1.10–1.42)5.6 × 10−4
African American2421730.370.341.18 (0.88–1.58)0.25
Meta-analysis: 8 1qC populations1,5261,6521.24 (1.10–1.39)2.9 × 10−4
Replication samples
    Independent WTCCC sample1,4952,9380.240.241.05 (0.95–1.17)0.33
    W2C vs. 58BC4861,9920.250.241.01 (0.86–1.19)0.56
    UKT2DGC3,9224,8190.240.241.04 (0.98–1.12)0.38
    Diabetes Genetics Initiative1,4641,4670.260.270.97(0.84–1.12)1.00
    FUSION1,1611,1740.250.270.90 (0.78–1.03)0.60
Meta-analysis: all replication samples8,52812,3901.02 (0.97–1.06)0.54
Meta-analysis: all European descent populations9,52813,5881.04 (1.00–1.09)0.083
Meta-analysis: all data10,05414,0421.04 (1.00–1.09)0.069
Open in a separate window*P value for Amish and Pima was adjusted using estimated lambda for GC. 58 BC, 1958 British Birth Cohort; UKT2DGC, United Kingdom Type 2 Diabetes Genetics Consortium; W2C, Warren 2 cases; WTCCC, Wellcome Trust Case Control Consortium.Both signals were most prominent in the European samples, and there was no evidence that an equivalent association signal extended to the East Asian, Native American, or African American samples. Though the association P values for these two signals remained strong in the 8-way meta-analysis of all data (5.4 × 10−5 for rs7538490 and 2.9 × 10−4 for rs11264371, and2),2), in each case they were driven by the larger European samples. Analyses in the larger Pima family-based association dataset also found no evidence of association (rs7538490, P = 0.59; rs11264372 [r 2 of one with rs11264371 in CEU and CHB/JPT HapMap], P = 0.79).Although neither signal was of sufficient effect size to be considered causal for the 1q linkage signal (the estimated sibling relative risk attributable to these loci in combination is only 1.045), we reasoned that these signals might nevertheless be pointers toward nearby causal variants (of lower frequency but higher penetrance) that were inadequately tagged by the SNPs we had typed. However, before proceeding to resequencing and fine-mapping, we first sought replication of our findings in independent datasets. Mindful that our case ascertainment strategies may have led to inflated estimates of effect size compared with those evident in unselected case subjects, we recognized that large sample sizes would be required to test the observed associations. Because the signals were clearest in European-descent samples, we focused replication on samples from Northern Europe.First, we used GWA data from the Wellcome Trust Case Control Consortium (19,20). After removing 429 overlapping case subjects, we examined 1,495 additional type 2 diabetes case subjects and 2,938 control subjects with Affymetrix 500k data (using imputation to test for association at the lead SNPs in each interval). No evidence of association was evident (rs7538490, P = 0.57; rs11264371, P = 0.33). Similarly, analysis of GWA data from the Diabetes Genetics Initiative (21) and FUSION (22) studies provided no corroboration of either signal. Furthermore, when analyzed jointly (4,549 case subjects, 5,579 control subjects), these three studies also failed to reveal any additional common variant signals of interest (P < 10−4) across the wider 1q region (23) and no corroboration of any of the lesser signals evident in the 1q consortium analyses.Finally, we genotyped the two lead SNPs (rs7538490, rs11264371) using fluorogenic 5′-nuclease (Taqman) assays in 4,572 case subjects and 6,941 control subjects from the U.K. (the UK Type 2 Diabetes Genetics Consortium and Warren 2 cases/1958 British Birth Cohort strata in and2).2). Once again, there was no evidence of replication. Taking into account all replication samples (∼8,500 case subjects, ∼12,400 control subjects), there was no significant association with type 2 diabetes (rs7538490, OR 1.03 [95% CI 0.98–1.07], P = 0.29; rs11264371, OR 1.02 [0.97–1.06], P = 0.54 [additive], 1.06 [0.99–1.13], P = 0.10 [dominant]). Nominal significance was retained when these replication data were combined with the original 1q consortium case-control data (rs7538490, P = 0.015; rs11264371, P = 0.069), but these associations are unimpressive in either the region-wide or genome-wide context. Even allowing for some heterogeneity of effect size between the primary and replication datasets (due to ascertainment differences and the “winner''s curse”), there seems to be no substantive evidence that the association signals observed in the NOS1AP and the PKLR/ASH1L region are genuinely associated with type 2 diabetes.In summary, we have undertaken a detailed survey of common variants across the region of replicated 1q linkage, achieving coverage that exceeds that of available GWA data for the region. Despite analysis of multiple ethnic groups in samples sufficiently powered (in the European-descent component at least) to have detected common variants causal for the linkage, we found no compelling signals.Should we conclude therefore that the original evidence for 1q linkage was false? Although this possibility cannot be discounted, it is worth considering that recent experience from GWA studies has shown that, for common susceptibility variants at least, effect sizes are modest and that none is of magnitude sufficient to generate a linkage signal detectable in achievable sample sizes. Efforts to explain the “missing heritability” for type 2 diabetes (that is, the disparity between the predisposition attributable to the known loci and independent estimates of overall heritability and familiality) are now shifting toward the search for low-frequency, medium-penetrance alleles. Alleles with these characteristics are likely to have escaped detection through the genome-wide approaches available so far, since they would be insufficiently penetrant to be detected with traditional linkage approaches applied to monogenic families and too infrequent to be reliably identified through GWA studies (4). Yet, low-frequency, medium-penetrance alleles could, particularly if several independent alleles map to the same locus, generate the kinds of linkage signals detectable in family-based studies (as is the case for NOD2/CARD15 and Crohn''s, for example) (24).Detection of low-frequency susceptibility variants will require new approaches based around next-generation resequencing and large-scale fine-mapping. Genome-wide resequencing of large case-control samples remains economically and logistically unfeasible, but targeted resequencing of selected regions is not, and the future plans of the 1q consortium include deep resequencing of the 1q region of interest, focusing at least initially on exons and conserved sequence.  相似文献   

12.
Adolescent Scalp Psoriasis: Update on Topical Combination Therapy     
Emily Osier  Barbara Gomez  Lawrence F. Eichenfield 《The Journal of clinical and aesthetic dermatology》2015,8(7):43-47
Plaque psoriasis can begin early in life and negatively affect quality of life. Topical agents are generally recommended as first-line therapy for plaque psoriasis. The synergy of a vitamin D analog and a steroid in a topical fixed-combination formulation provides more favorable effectiveness and tolerability as compared with either agent alone. The safety and effectiveness of a once-daily calcipotriene/betamethasone dipropionate topical suspension have been established in children 12 to 17 years of age with scalp plaque psoriasis. Combination topical formulations and once-daily dosing decrease regimen complexity and may increase adherence. Accommodation of vehicle preference may also improve adherence and real-life effectiveness.Psoriasis, a common dermatologic disorder affecting individuals of all ages, can begin early in life. Up to 35 percent of cases begin before 18 years of age, affecting 0.7 percent of children.1-3 This incidence doubled between 1970 and 1999 and is currently 40.8 cases per 100,000.4 The median age at diagnosis (10.6 years) has remained stable in the United States.4It is important for clinicians to appreciate the impact of psoriasis on children and teenagers. Even mild forms of psoriasis can affect childhood psychosocial functioning and quality of life (QoL).2,5-7 The scalp plaques and possible associated alopecia can be particularly troublesome during adolescence and detrimental to the individual’s sense of self during the transition to adulthood.8,9Topical therapy can be safe and effective in juvenile psoriasis. Topical treatments are a first-line option in adults, and some have been approved for use in adolescents (10-19 This article reviews the distinguishing features of psoriasis in younger patients and the considerations that enter into the choice of treatment.

TABLE 1

Approved topical corticosteroid formulations for adolescents10-14, 16-19
CORTICOSTEROIDAPPROVAL AGE (YEARS)
Betamethasone dipropionate 0.05%≥13
 Cream
 Ointment
 Lotion
Betamethasone dipropionate 0.05% Gel≥12
Clobetasol propionate 0.05%≥12
 Cream
 Ointment
 Foam
 Gel and Solution
Halobetasol 0.05%≥12
 Cream
 Ointment
Open in a separate window  相似文献   

13.
The renal mononuclear phagocytic system     
Nelson PJ  Rees AJ  Griffin MD  Hughes J  Kurts C  Duffield J 《Journal of the American Society of Nephrology : JASN》2012,23(2):194-203
  相似文献   

14.
Functional Effector Memory T Cells Enrich the Peritoneal Cavity of Patients Treated with Peritoneal Dialysis     
Gareth W. Roberts  Duncan Baird  Kathleen Gallagher  Rhiannon E. Jones  Christopher J. Pepper  John D Williams  Nicholas Topley 《Journal of the American Society of Nephrology : JASN》2009,20(9):1895-1900
The frequency and severity of episodes of peritonitis adversely affect the structure and function of the peritoneal membrane in patients treated with peritoneal dialysis (PD), but the underlying mechanisms are not well understood. Alterations in the phenotype and function of resident peritoneal cells may contribute. Because effector memory T cells play a pivotal role in maintaining peripheral tissue immunity, we hypothesized that these cells may initiate or perpetuate the peritoneal inflammatory response. Here, we characterized the phenotype and effector function of peritoneal memory T cells. We found that functional effector memory T cells capable of mounting long-term recall responses enrich the peritoneal cavity of PD patients. Peritoneal T cells were able to mount a Th1-polarized response to recall antigens, and these responses were greater in peritoneal T cells compared with T cells in the peripheral blood. We also observed that the peritoneal T cells had altered telomeres; some cells had ultrashort telomeres, suggesting a highly differentiated local population. In summary, we describe a resident population of memory T cells in the peritoneum of PD patients and speculate that these cells form part of the first line of defense against invading pathogens.Despite advances in treatment, peritoneal infection remains one of the main causes of technique failure in peritoneal dialysis (PD) patients. There is a strong association between peritonitis (frequency and severity) and the loss of membrane function.13 In view of this, there has been considerable interest in understanding the basic processes that regulate peritoneal early responses to infection. Most of these studies have focused on the contribution of peritoneal macrophages or mesothelial cells to these processes.49 Despite representing up to 25% of the resident peritoneal leukocyte population and forming a significant proportion of the leukocyte population present in resolving peritonitis, the function and phenotype of human peritoneal T cells is poorly defined.1012 Consequently we understand very little about the adaptive arm of the peritoneal immune responseRecent developments in the field of immunology have greatly enhanced our understanding of T cell phenotype, activation status, differentiation, and tissue homing capacity. Many studies have highlighted the important role of T cells in providing long-term immunological memory.1319 According to current definitions, memory T cells are distinguished from naïve T cells (which are yet to encounter their cognate antigen) by the expression of CD45RO (rather than CD45RA).20 Within the memory T cell population, distinct functional subsets have been characterized based on the expression the lymph node homing signal CCR7.1319 These subsets differ in both their tissue homing capability and in their response to antigenic stimulation.15,17,18 Central memory (TCM) cells (CD45RO/CCR7+) are thought to migrate through lymph tissue, whereas effector memory (TEM) cells (CD45RO /CCR7), which lack lymph node homing signals, are thought to reside primarily in peripheral tissue.16,17 The TEM subset rapidly produces effector cytokines such as IFN-γ and IL-4 and are thought to form a first line of defense in vulnerable peripheral tissues. In contrast, TCM cells lack immediate effector function but retain proliferative capacity and are capable of generating a secondary wave of antigen-specific effector T cells.1719 The memory subsets also differ in their replicative history and degree of differentiation. Within the T cell population, telomere lengths decrease from naïve through TCM through TEM cells, suggesting that the latter have undergone more cell divisions and are a more highly differentiated population.17,20Most of our current understanding of T cell memory is derived from murine data or from experiments performed on peripheral blood T cells. Because of the logistical difficulties involved in sample collection, there are very little data available regarding the phenotype and function of memory T cells in human peripheral tissue. The aim of our study was to characterize the phenotype, replicative history, and effector function of the peritoneal memory T cell population during steady-state (non-infected) PD.Our results demonstrate that as compared with peripheral blood, the peritoneal cavity is enriched in cells displaying a TEM phenotype, with very few intraperitoneal naïve T cells (Figure 1, A and B) (we found no significant difference in the proportion of TCM cells between blood and peritoneum). Subgroup analysis shows that neither time on PD nor recurrent peritonitis have a significant effect on the proportion of TEM within the peritoneal cavity, suggesting that TEM enrichment is a characteristic of the quiescent peritoneal cavity (Supplementary Figure 1). Further phenotypic analysis demonstrated increased expression of the proinflammatory chemokine receptor CCR5 on the TEM subset, but only low-level expression of the lymph node homing signal CD62L (Figure 1C).Open in a separate windowFigure 1.Paired samples of peripheral blood mononuclear cells (PBMCs) and peritoneal leukocytes were collected from PD patients. Cells were labeled with fluorescently conjugated monoclonal antibody directed against CD4/CD8/CCR7 and CD45RO. A combined gate was set on peritoneal T cells on the basis of CD4 or CD8 expression and their FSC:SSC profiles. (A) Representative example of CD4 data. (B) Summary of data obtained from paired blood and peritoneal T cells obtained from 20 patients. Differences between groups were tested using the Wilcoxon signed ranks test (*P < 0.05). (C) Peritoneal leukocytes were collected from ten separate PD patients. Cells were labeled with fluorescently conjugated monoclonal antibody directed against CD4/CCR7/CD45RO and CD62L or CCR5. A combined gate was set on peritoneal T cells on the basis of CD4 expression and their FSC:SSC profiles. Further gate was plotted on the TEM subset and percentage CD62L+/CCR5+ within this calculated subset. See and22 for patient demographics.

Table 1.

Patient demographics
PatientAge (yr)Peritonitis EpisodesDiabetes Mellitus (yes/no)Glucose Exposure (g/d)PD Vintage (mo)Icodextrin (yes/no)Etiology of End-Stage Renal Failure
A1560No1083NoHypertensive nephropathy
A2580No1084NoHypertensive nephropathy
A3660Yes99.817YesSystemic sclerosis
A4660Yes12737NoDiabetic nephropathy
A5420No154.25NoUnknown
A6590No12786NoChronic pyelonephritis
A7750No81.635YesReflux nephropathy
A8660No10813NoUnknown
B1601No81.622YesUnknown
B2651No81.611NoReflux nephropathy
B3621No1544NoRenovascular disease
B4642No12748NoUnknown
B5561No1084NoIgA GN
B6792Yes81.615YesDiabetic nephropathy
B7701Yes99.816YesDiabetic nephropathy
C1663No113.630YesIgA nephropathy
C2745No12772NoHypertensive nephropathy
C3715Yes81.624YesDiabetic nephropathy
C4575No9389YesObstructive nephropathy
C5646No163.460NoRenovascular disease
C6754No12734YesIgA nephropathy
Open in a separate window

Table 2.

Patients represented in each figure
FigurePatient
1AA2
1BA1 to A8, B1 to B7,C1 to C6
1CA2 to A4, A6, B3, B5, C3 to C6
2AA5
2BA5, A7, B2, C2
3AB1
3BC5, B1, A1
Open in a separate windowTo determine whether peritoneal memory cells retain effector function ex vivo, cells were stimulated for a short period with phorbol 12-myristate 13-acetate/ionomycin. As predicted from murine studies, the predominant early Th1 (IFN-γ-mediated) response to stimulation came from the TEM subset (Figure 2A). To examine the antigen specificity of this response, paired samples of peripheral blood and peritoneal T cells were exposed to the standardized recall antigens, including purified protein derivative from Mycobacterium tuberculosis, hemagglutinin antigen (HA) derived from influenza virus, and tetanus toxoid (TT). Our results demonstrate that peritoneal T cells are able to mount a Th1 polarized response to these recall antigens; moreover, these responses are increased in peritoneal T cells as compared with peripheral blood T cells (Figure 2B).Open in a separate windowFigure 2.(A) Peritoneal leukocytes were stimulated with phorbol 12-myristate 13-acetate (500 ng/ml)/ionomycin (50 ng/ml) for 2 h. Cells were labeled with fluorescently conjugated monoclonal antibody directed against CD4/CD45RO/CCR7. Cells were subsequently fixed and permeabilized then stained with anti-IFN-γ. A combined gate was set on peritoneal T cells on the basis of CD4 expression and their FSC:SSC profiles. Further gates were plotted on the CD4 memory subsets (naïve/TCM/TEM). Results show the percentage of IFN-γ+ cells within these memory subsets. Results are a representative example of experiments performed on four separate donors. (B) Paired samples of PBMCs and peritoneal leukocytes were collected from PD patients. ELISpot plates were coated with 50 μl anti-IFN-γ antibody in sterile PBS (10 μg/ml) and incubated at 4°C for 180 min. Peritoneal leukocytes and PBMCs were plated out at 2 × 105 cells/well. Cells were incubated at 37°C for 16 h in the presence of purified protein derivative (PPD) (10 μg/ml), TT (5 μg/ml), or HA (5 μg /ml). The ELISpot assay was developed according to the manufacturer''s instructions. Results show mean ± SEM of triplicate observations. See and22 for patient demographics.Because memory T cells are considered a highly differentiated population,17,20 we next examined the replicative history of the peritoneal T cells. Telomeres progressively shorten as a function of cell division, thus telomere length is a robust indicator of the replicative history of lymphocytes in vivo.20,21 To date, the two most widely applied methods for studying telomere length are terminal restriction fragment analysis and quantitative fluorescence in situ hybridization.21,22 Terminal restriction fragment analysis suffers from a low overall sensitivity and requires large cell numbers, whereas quantitative fluorescence in situ hybridization requires metaphase chromosomes, limiting analysis to cells that are actively proliferating.21 In view of these restrictions, such methods are of limited value in the analysis of peritoneal T cells (in which cell numbers and proliferative capacity are low). Furthermore, these hybridization-based technologies become increasingly less efficient as telomere length diminishes and fail to detect the very short telomeres capable of triggering replicative senescence.21The development of single telomere length analysis (STELA) has emerged, which overcomes many of these limitations and allows accurate measurement of the full spectrum of telomere lengths from individual chromosomes.2123 The robust nature of STELA allows detection of very short telomeres even when such telomeres are rare or present in a background of longer telomeres.21,22 This technology uniquely allows the accurate assessment of the mean telomere length (±SD) within a cellular population and can also identify subpopulations with longer or shorter telomeres. Using STELA, we were able to compare the telomere lengths of purified populations of peritoneal and peripheral blood T cells derived from the same patient. Our results show that as compared with peripheral blood from the same individual, peritoneal T cells have significantly shortened telomeres (mean 1 to 1.5 kb shorter, representing 15 to 20 population doublings, P < 0.05) with some telomere lengths approaching the senescent range (1–2kb) (Figure 3). Because telomere lengths progressively shorten from naïve through TCM to TEM,17,20 these data correlate with our phenotypic analysis, confirming that the peritoneal cavity is enriched in highly differentiated TEM cells. It is important to point out here that in addition to the mean telomere lengths being significantly shorter in the peritoneal cavity T cells, a population of T cells with very short telomeres not represented in the paired peripheral blood samples was evident. This provides compelling evidence for a distinct resident population of T cells in the peritoneal cavity.Open in a separate windowFigure 3.Samples of peritoneal leukocytes and PBMCs were obtained from PD patients. Further PBMC samples were obtained from healthy age-matched volunteers. Flow cytometry and cell sorting (FACS) were used to isolate highly purified (>98%) T cell populations. T cell telomere lengths were measured by STELA. (A) Representative example of STELA data from a PD patient. (B) Mean XpYp telomere lengths. The unpaired t test was used to compare means. See and22 for patient demographics.The above results collectively demonstrate that the peritoneal cavity is enriched in functional resident TEM cells. This observation is in agreement with the current paradigm of T cell memory that predicts that TEM cells reside primarily in peripheral tissues.16,17 The peritoneal homing of TEM cells is facilitated by the expression of proinflammatory chemokine receptors such as CCR5, the ligands for which (MiP-1 and RANTES) have been detected in the peritoneal effluent of PD patients.24,25 Our functional data support this “selective recruitment” hypothesis, because memory T cells formed in response to prior vaccination are present at an increased frequency within the peritoneal cavity. This peripheral homing of TEM cells may have evolved as a protective mechanism, ensuring that vulnerable peripheral tissues contain an abundance of “primed” effector cells.Further insight into the dynamics of memory T cell trafficking was obtained from the analysis of T cell telomere lengths. Although the term “resident peritoneal cell” is often used to describe cells obtained in PD effluent, this term is misleading because T cells recruited from the blood into the cavity then drained off minutes later are still classed as resident peritoneal cells. It could be argued that resident peritoneal T cells do not in fact exist, and that cells obtained in PD effluent are cells that are continually trafficking between the peripheral circulation, the peritoneal cavity, and peritoneal lymphatics. Until now, it has been very difficult to disprove this argument because cell-labeling studies are logistically difficult to perform in humans. Our telomere data allow us to argue that there is in fact a truly resident peritoneal memory T cell population that resides and divides locally within the peritoneum. The evidence supporting this comes from the finding that T cells with very short telomere lengths were found only in the peritoneum but not in the peripheral circulation. Had these cells been recruited from the blood, then we would have seen a corresponding telomere band in the peripheral blood, but this is not in fact the case. Indeed, more detailed analysis of the STELA data suggests that there are two distinct T cell populations. One population has similar telomere lengths to peripheral blood T cells and is likely to represent recirculating TCM and naïve T cells, the other population has shorter telomere lengths and are likely to represent truly resident TEM cells. We speculate that these resident cells avoid being drained off during PD by adhering to the peritoneal membrane or trafficking to peritoneal milky spots, thus enabling long-lived memory responses.2628 Some of these cells have ultrashort telomere lengths and may be approaching cellular senescence. Because senescent cells have a more catabolic, proinflammatory phenotype,21,22 the presence of even small numbers of such cells in the peritoneal cavity may contribute to the local proinflammatory milieuOf note, to standardize our telomere data we extended our study to include healthy age and sex-matched controls. We observed that (when compared with controls) PD patients had considerably shortened peripheral blood T cell telomeres (Figure 3). This telomere shortening may be secondary to the increased inflammatory burden that these patients have faced during the development of end-stage renal failure. This may have prognostic implications for PD patients, because previous population-based studies have shown that individuals with shortened peripheral blood telomeres have a 3-fold increase in cardiovascular mortality and an 8-fold higher mortality from infectious diseases.29In conclusion, we have shown for the first time that the peritoneal cavity hosts a population of functional resident TEM. Because these cells mount an immediate Th1 response, we speculate that they are important arbiters of the early immune response, aiding macrophage activation via the production of IFN-γ. Future work will focus on whether these cells can be primed to recognize components of the organisms that commonly cause peritonitis. Such work might allow the development of effective intraperitoneal vaccination conferring protection against recurrent peritonitis.  相似文献   

15.
LMX1B Mutations Cause Hereditary FSGS without Extrarenal Involvement     
Olivia Boyer  Stéphanie Woerner  Fan Yang  Edward J. Oakeley  Bolan Linghu  Olivier Gribouval  Marie-Josèphe Tête  José S. Duca  Lloyd Klickstein  Amy J. Damask  Joseph D. Szustakowski  Fran?oise Heibel  Marie Matignon  Véronique Baudouin  Fran?ois Chantrel  Jacqueline Champigneulle  Laurent Martin  Patrick Nitschké  Marie-Claire Gubler  Keith J. Johnson  Salah-Dine Chibout  Corinne Antignac 《Journal of the American Society of Nephrology : JASN》2013,24(8):1216-1222
  相似文献   

16.
Combined Cellular Blue Nevus and Trichoepithelioma     
Richard Martin  Patrick Emanuel 《The Journal of clinical and aesthetic dermatology》2013,6(8):35-38
  相似文献   

17.
Endothelin-A Receptor Antagonism Modifies Cardiovascular Risk Factors in CKD     
Neeraj Dhaun  Vanessa Melville  Scott Blackwell  Dinesh K. Talwar  Neil R. Johnston  Jane Goddard  David J. Webb 《Journal of the American Society of Nephrology : JASN》2012,24(1):31-36
Arterial stiffness and impaired nitric oxide (NO) bioavailability contribute to the high risk for cardiovascular disease in CKD. Both asymmetric dimethylarginine (ADMA), an endogenous inhibitor of NO production, and endothelin-1 (ET-1) oppose the actions of NO, suggesting that ET-1 receptor antagonists may have a role in cardiovascular protection in CKD. We conducted a randomized, double-blind, three-way crossover study in 27 patients with proteinuric CKD to compare the effects of the ETA receptor antagonist sitaxentan, nifedipine, and placebo on proteinuria, BP, arterial stiffness, and various cardiovascular biomarkers. After 6 weeks of treatment, placebo and nifedipine did not affect plasma urate, ADMA, or urine ET-1/creatinine, which reflects renal ET-1 production; in contrast, sitaxentan led to statistically significant reductions in all three of these biomarkers. No treatment affected plasma ET-1. Reductions in proteinuria and BP after sitaxentan treatment was associated with increases in urine ET-1/creatinine, whereas reduction in pulse-wave velocity, a measure of arterial stiffness, was associated with a decrease in ADMA. Taken together, these data suggest that ETA receptor antagonism may modify risk factors for cardiovascular disease in CKD.CKD is common, affecting 6%–11% of the population globally.1 It is strongly associated with incident cardiovascular disease (CVD).2 This increased cardiovascular risk is not adequately explained by conventional (Framingham) risk factors, such as hypertension, hypercholesterolemia, diabetes mellitus, and smoking, all of which are common in patients with CKD. Thus, emerging cardiovascular risk factors have been an area of intense investigation.3 Arterial stiffness4 makes an important independent contribution to CVD risk in CKD, and this is promoted by both conventional and emerging cardiovascular risk factors.Hyperuricemia and a shift in the balance of the vasodilator nitric oxide (NO) and vasoconstrictor endothelin (ET) systems have been identified as potential contributors to increased cardiovascular risk in patients with CKD.5 These are all common in a typical CKD population.3,6 Epidemiologic studies report a relationship between serum uric acid and a wide variety of cardiovascular conditions, including hypertension, diabetes mellitus, coronary artery disease, cerebrovascular disease, and CKD.7 Indeed, serum uric acid is considered by some to be an independent risk factor for both CVD8,9 and CKD.10 Others have noted that an elevated serum uric acid level predicts the development of hypertension and CKD.7 Of note, emerging clinical data show that decreasing serum uric acid levels has both cardiovascular and renal benefits.1113Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NO synthases. By inhibiting NO formation, ADMA causes endothelial dysfunction, vasoconstriction, elevation of BP, and progression of experimental atherosclerosis.14 ADMA concentrations are increased in patients with CKD,14 and clinical data support ADMA as an independent marker of CKD progression, cardiovascular morbidity, and overall mortality.1517 Studies have shown a reduction in ADMA after therapy in patients with hypertension and hypercholesterolemia,18,19 but not in patients with CKD.ET-1 is a potent endogenous vasoconstrictor produced within the vasculature. It is implicated in both the development and progression of CKD.20 Its effects are mediated via two receptors, the ETA and ETB receptors; the major pathologic effects are ETA receptor mediated.20 We have recently shown that long-term selective ETA receptor antagonist therapy using the orally active drug sitaxentan reduces proteinuria, BP, and arterial stiffness in patients with proteinuric CKD,21 effects that are potentially renoprotective. We hypothesized that in this same cohort of patients with CKD, sitaxentan would also reduce levels of serum uric acid, ADMA, and urine ET-1 (as a measure of renal ET-1 production) and so provide broader cardiovascular and renal protection. The current data show the effects of sitaxentan, as well as placebo and an active control agent, nifedipine, on these novel cardiovascular risk factors.As described elsewhere,21 after 6 weeks of dosing no significant differences were seen between sitaxentan and nifedipine in the reductions from baseline in BP measures. Despite this, sitaxentan reduced proteinuria to a significantly greater extent than did nifedipine. Pulse-wave velocity (PWV)—a measure of arterial stiffness—decreased to a similar degree with nifedipine as with sitaxentan. Placebo did not affect proteinuria, BP, or PWV (see VariablePlaceboSitaxentanNifedipineBaselineWeek 6BaselineWeek 6BaselineWeek 624-hr proteinuria (g/d)2.06±0.382.00±0.332.07±0.341.46±0.26a1.95±0.301.99±0.33Protein-to-creatinine ratio (mg/mmol)155±31153±27157±28114±23b155±27152±29Mean arterial pressure (mmHg)94.6±2.294.3±1.794.4±1.890.7±1.8c95.5±2.091.7±1.7cSystolic BP (mmHg)125.4±2.7124.2±1.9124.3±2.2120.7±1.9c125.7±2.4120.7±1.6cDiastolic BP (mmHg)77.9±1.577.5±1.277.9±1.374.3±1.3a78. 9±1.575.7±1.2aPWV (m/s)7.7±0.38.0±0.48.0±0.37.6±0.3c7.9±0.37.6±0.3cCentral augmentation index (%)20±220±220±215±2a19±217±2Plasma ET-1 (pg/ml)3.6±0.53.7±0.63.6±0.53.7±0.53.5±0.53.5±0.5Urine ET-1 (pg/ml)4.5±0.44.7±0.45.1±0.44.2±2.1c5.1±0.44.7±0.4Open in a separate windowValues are given as predosing baseline ± SEM.aP<0.01 for week 6 versus baseline.bP=0.01 for week 6 versus baseline.cP<0.05 for week 6 versus baseline.

Table 2.

Renal substudy data from clearance studies performed at baseline and week 6 of each study period
VariablePlaceboSitaxentanNifedipine
BaselineWeek 6BaselineWeek 6BaselineWeek 6
GFR (ml/min)56±754±857±848±8a59±858±9
Effective renal blood flow (ml/min)533±66552±65511±63543±73562±82530±72
Effective renal vascular resistance (mmHg/min per L)230±52206±39236±44232±48248±58254±56
Effective filtration fraction (%)19.1±1.117.9±1.320.8±1.016.6±0.7b20.3±1.120.5±1.4
Open in a separate windowValues are given as predosing baseline ± SEM.aP<0.05 for sitaxentan at week 6 versus sitaxentan at baseline.bP<0.01 for sitaxentan at week 6 versus sitaxentan at baseline.Baseline serum uric acid was in the frankly hyperuricemic range in all three phases of the study: placebo, 476±20 μmol/L; sitaxentan, 506±21 μmol/L; nifedipine, 479±19 μmol/L. Baseline serum uric acid was inversely related to baseline proteinuria (r2=0.19; P=0.02). Whereas placebo and nifedipine had no effect on serum uric acid, sitaxentan reduced serum uric acid by approximately 11% by study end (Figure 1A). This effect was similar at weeks 3 and 6 of the study. In multivariate analysis, the reduction in serum uric acid was not associated with changes in proteinuria, BP, or PWV (data not shown). The reduction in serum uric acid was matched by an increase in the fractional urinary excretion of uric acid (baseline versus week 6: 6.0%±0.6% versus 7.3%±0.7%; P=0.05).Open in a separate windowFigure 1.Selective endothelin-A receptor antagonism reduces serum urat, ADMA and urine ET-1/creat in CKD patients. Change from baseline in (A) serum uric acid, (B) ADMA, and (C) urine ET-1/creatinine after 3 and 6 weeks’ treatment with placebo (open bar), sitaxentan (speckled bar), and nifedipine (hashed bar). Values are expressed as mean ± SEM. *P<0.01 for sitaxentan versus placebo at 3 or 6 weeks; †P<0.05 for sitaxentan versus placebo at 3 weeks.Baseline ADMA concentrations were the same for all three phases of the study: placebo, 0.52±0.01 μmol/L; sitaxentan, 0.52±0.01 μmol/L; nifedipine, 0.52±0.02 μmol/L. Whereas placebo and nifedipine did not affect ADMA, 6 weeks of sitaxentan reduced ADMA by approximately 8% (Figure 1B). This reduction in ADMA was directly correlated with a reduction in PWV (Figure 2A; r=0.39; P<0.05), and in multivariate analysis, the change in ADMA (but not changes in proteinuria, BP, plasma ET-1, urine ET-1/creatinine, or serum uric acid) independently predicted the reduction in PWV after sitaxentan treatment.Open in a separate windowFigure 2.Changes in arterial stiffness, blood pressure and proteinuria following endothelin-A antagonism relate to changes in ADMA and urine ET-1/creat. Relationships between (A) percentage change in PWV against percentage change in ADMA and (B) percentage change in proteinuria and (C) percentage change in mean arterial pressure against percentage change in urine ET-1/creatinine. MAP, mean arterial pressure; uET-1/creat, urine ET-1/creatinine.Plasma ET-1 concentrations were similar at baseline in all three phases of the study—placebo, 3.57±0.50 pg/ml; sitaxentan, 3.60±0.49 pg/ml; nifedipine, 3.54±0.46 pg/ml—and were not affected by any of the interventions (Figure 1C). A similar effect was seen on urine ET-1 concentration (without correction to urine creatinine). Neither placebo nor nifedipine affected urine ET-1, whereas sitaxentan reduced this significantly (Figure 2, B and C; r=0.41; P<0.05 for both), and in multivariate analysis, the change in urine ET-1/creatinine independently predicted the changes in proteinuria and BP.In addition to the important evidence of potentially renoprotective effects on proteinuria, BP, and arterial stiffness, the current data show that ETA receptor antagonism selectively decreases serum uric acid, ADMA, and urinary ET-1 levels in patients with proteinuric CKD, independent of BP. These effects were seen in patients already receiving optimal treatment with angiotensin-converting enzyme inhibitors and angiotensin-receptor blockers. These findings suggest a potential role for ETA receptor antagonism in conferring additional longer-term cardiovascular and renal benefits in patients with CKD.Decreasing serum uric acid may reduce cardiovascular risk and CKD progression.7 Treatment of asymptomatic hyperuricemia improves renal function22 and delays disease progression11 in patients with early CKD (stage 3). In a different approach, withdrawal of the xanthine oxidase inhibitor allopurinol from a group of patients with stable CKD led to both worsening of hypertension and acceleration of renal dysfunction, although this occurred only in patients not taking an angiotensin-converting enzyme inhibitor.12 However, these studies suggesting benefits of reducing serum uric acid used allopurinol as the therapeutic agent. More recently, the angiotensin-receptor blocker losartan has been shown to decrease serum uric acid in a group of patients with type 2 diabetes and nephropathy, and this reduction was associated with a reduction in CKD progression.23 ET receptor antagonism offers a potentially novel approach to decreasing serum uric acid in patients with proteinuric CKD.Only two studies have shown that ET receptor antagonism reduces serum uric acid, neither of which included patients with CKD. Six months of treatment with the selective ETA receptor antagonist atrasentan reduced serum uric acid levels from 293 to 286 μmol/L in patients with early atherosclerosis.24 Change in serum uric acid was not a primary endpoint in this study, and although this was a statistically significant reduction it is not clinically meaningful. In another small open-label study (n=15) in patients with pulmonary arterial hypertension and no control group, Ulrich and colleagues showed that 6 months’ treatment with the mixed ETA/B receptor antagonist bosentan decreased serum uric acid from 353 to 305 μmol/L.25 The current data build on these studies by showing that in a randomized controlled trial of patients with proteinuric CKD, in which baseline serum uric acid levels were much higher, selective ETA receptor antagonism reduces serum uric acid by approximately 11% (more impressive reductions than seen in the previous two studies), independent of BP. Furthermore, as a mechanism for this we have shown an increase in the renal excretion of uric acid.There is increasing interest in the NO system and, in particular, ADMA in relation to both the development and progression of CKD. Many studies in patients with varying degrees of CKD have confirmed that ADMA is elevated in CKD.14,26 Of note, data suggest that ADMA is elevated independently of renal function in CKD,27 suggesting that mechanisms other than impaired clearance may contribute to the accumulation of ADMA in this setting. The ET system is upregulated in CKD.20 There is often reciprocal upregulation of the ET system20 in circumstances with downregulation of NO system activity.20 In the current study, baseline plasma ADMA and ET-1 did indeed correlate highly with each other (r2=0.56; P<0.01), confirming the reciprocal relationship between the NO and ET systems.Few interventional studies have shown a reduction in ADMA. These have not included patients with CKD and have suffered from being small or lacking in rigorous methods.18,19 To our knowledge, the current study is the first to show that ET receptor antagonism may reduce circulating ADMA concentrations. We have previously shown in a cross-sectional study that ADMA concentrations directly correlate with arterial stiffness—as measured by PVW—in a similar cohort of patients with CKD.26 The current study takes this observation further by showing for the first time that a decrease in ADMA correlates with an improvement in arterial stiffness, although we recognize this correlation to be weak. Additionally, it is not possible to separate independent effects of the ETA antagonist on arterial stiffness and ADMA in this limited number of patients. Because both increased ADMA and arterial stiffness independently contribute to CKD progression and its associated morbidity and mortality,14,20 ET receptor antagonism offers a potentially attractive novel therapy in CKD with benefits beyond those of lowering BP and proteinuria.Urinary ET-1 is a recognized measure of renal ET-1 production.28 Selective ETA receptor antagonism reduced renal ET-1 production at 6 weeks. Of note, the decrease in BP seen with sitaxentan at 6 weeks correlated inversely (albeit weakly) with urinary ET-1; that is, a greater decrease in BP was seen in patients who had less of a reduction, or even an increase, in renal ET-1 production. Renal ET-1 is involved in salt and water excretion29 and so part of the mechanism for the BP-lowering effect of ET receptor antagonism (in addition to their direct effects on the vasculature) may relate to an increase in renal ET-1 production to increase both natriuresis and diuresis. In this study there was no relationship between changes in salt excretion and urinary ET-1. A relationship similar to that seen with BP was also seen between the 6-week change in urinary ET-1 and proteinuria. The reduction in proteinuria with sitaxentan related to the decrease in BP (r2=0.16; P=0.04) and so this may explain part of this.The current data show for the first time that selective ETA receptor antagonism reduces novel cardiovascular risk factors in patients with proteinuric CKD established on optimal therapy. These data build on our earlier cross-sectional study, which showed that ADMA concentrations directly correlate with arterial stiffness, a powerful predictor of cardiovascular disease in patients with CKD. The mechanisms for these effects need to be further explored as a focus of future research. Certainly, reduction in BP is not sufficient because the active control agent nifedipine matched the decrease in BP seen with sitaxentan but did not reduce serum uric acid, ADMA, or urinary ET-1. Larger studies are needed to confirm these important findings in a group of patients at very high cardiovascular risk.  相似文献   

18.
A Practical Approach to Monitoring Patients on Biological Agents for the Treatment of Psoriasis     
Jason J. Emer  Amylynne Frankel    Joshua A. Zeichner 《The Journal of clinical and aesthetic dermatology》2010,3(8):20-26
Psoriasis is a chronic, systemic, inflammatory skin condition that manifests predominantly as well-demarcated, erythematous, scaly plaques on the elbows, knees, and scalp. While mild cases (minimal body surface) often respond to various topical treatments and light therapy, patients with extensive disease (larger body surface and possibly joint involvement) may require systemic medications for remission. The development of biological agents provides dermatologists valuable ways to help treat psoriatic disease quite efficiently, but literature regarding the monitoring of patients on biological treatments is sparse. Clinical practice varies widely since there is modest strong evidence to recommend or refute most tests currently recommended by the United States Food and Drug Administration. The purpose of this article is to present a practical approach to monitoring patients on biological therapy based on the most up-to-date literature.The use of biological treatments has grown significantly since their introduction and now account for a significant proportion of the systemic therapies used for the treatment of psoriasis. Biological therapies target precise segments of the immune system, offering the advantage of being less immunosuppressive compared to the traditional systemic therapies that broadly cause immunosuppression. Currently, five biological agents (e.g., alefacept, etanercept, infliximab, adalimumab, and ustekinumab) are approved by the United States Food and Drug Administration (FDA) for the treatment of psoriasis, and other newer agents (e.g., ABT-874) are in various stages of development and clinical trials (16 The biologicals at present are divided into either tumor necrosis factor alpha (TNF-α) or T-cell lymphocyte inhibitors. Recently, CD4+ T helper (Th) 17 cells and interleukins (IL)-12 and IL-23 have been important in the pathogenesis of T-cell mediated disorders, such as psoriasis, and have influenced the development of medications that specifically target these key immunological players. Both IL-12 and IL-23 stimulate differentiation of naive T-cells into Th1 and Th17 cells, key cells that regulate the production of other pro-inflammatory cytokines significant in the pathogenesis of psoriasis.7,8 Understanding of these immune cascade complexities has divulged this new class of biological agents that target cytokines (e.g., ustekinumab) important in the pathogenesis of inflammatory skin disease. Each drug class that is used in the treatment of psoriasis works by blocking different steps along the same immune-dysregulation pathway leading to psoriatic disease.

Table 1

Currently approved biological medications for the treatment of psoriasis14,6
DRUG NAMETRADE NAMEMECHANISM OF ACTIONDOSINGFDA-APPROVED INDICATIONSFDA APPROVAL FOR PSORIASIS
Anti-TNF-α
AdalimumabHumiraRecombinant human IgG1 monoclonal antibody80mg initial dose, followed by 40mg EOW starting one week after initial doseRA, JIA, PsA, Ps, AS, CD2008
EtanerceptEnbrelDimeric fusion protein linked to Fc portion of human IgG150mg SQ BIW for three months, followed by a reduction to a maintenance dose of 50mg per weekJIA, RA, PsA, AS, Ps2004
InfliximabRemicadeChimeric IgG1 monoclonal antibody5mg/kg IV infusion followed by additional doses at two and six weeks after the first infusion, then every eight weeks thereafterRA, PsA, CD, Ps, UC, AS2006
T-cell Inhibitor
AlefaceptAmeviveDimeric fusion protein of CD2/LFA-3 linked to Fc portion of human IgG115mg IM weekly for 12 weekly injectionsPs2003
Anti-IL*
UstekinumabStelaraHuman IgG1 monoclonal antibody specific to p40 protein subunit of interleukin-12 and -23 cytokines45mg or 90mg initially and four weeks later, followed by 45mg or 90mg every 12 weeksPs2009
Open in a separate window
TNF
tumor necrosis factor
mg
milligram
EOW
every other week
RA
rheumatoid arthritis
JIA
juvenile idiopathic arthritis
PsA
psoriatic arthritis
Ps
plaque psoriasis
AS
ankylosing spondylitis
CD
Crohn''s disease
SQ
subcutaneous
BIW
twice weekly
kg
kilogram
IV
intravenous
UC
ulcerative colitis
IM
intramuscular
IL
interleukin.
*The authors are categorizing ustekinumab and related medicines as a class called anti-IL for the purposes of this article.Biological agents have changed the treatment of psoriasis by giving dermatologists additional therapeutic options that are potentially less toxic to the liver, kidneys, and bone marrow, and are not teratogenic compared to the traditional systemic therapies for psoriasis, such as acitretin, methotrexate, and cyclosporine. Concerns of increased cholesterol, hair loss, and mucous membrane dryness seen with acitretin; liver and bone marrow toxicity, risk of lymphoma or cancers, and risk of serious infections seen with methotrexate; and increased blood pressure and increased cholesterol, electrolyte disturbance, risk of lymphoma and cancers, and risk of serious infections seen with cyclosporine, have essentially been shattered with the introduction of biological drugs. Even so, traditional systemic therapies continue to play an important role in the treatment of psoriasis with their oral route of administration and low cost, making them an important treatment option in the appropriate patient. Phototherapy is very efficacious, but requires a heavy time commitment and a phototherapy unit, may increase the risk of skin cancer, and involves the diligence of a physician who has experience making frequent use of this therapy. Biological agents have grown increasingly popular for the treatment of moderate-to-severe disease, as clinical studies have shown these agents to be free of the major organ toxicities of methotrexate and cyclosporine and successful in treating those who may have been unresponsive or unable to tolerate traditional therapies. Although the majority of patients on biological agents have few complications, associated side effects are of real concern, and cautious monitoring with frequent laboratory testing, pristine patient education, and regular office visits, are necessary.Several consensus statements and literature reviews have been published to reconcile differences among dermatologists and provide recommendations for the care of patients on biologicals.913 Current agreement mandates a diligent screening process prior to initiating any biological agent including a thorough medical history and physical examination, with particular attention to the review of systems; specifically, the neurological, cardiovascular, gastrointestinal, and musculoskeletal systems. Important information from the past medical history includes history of previous or current serious or opportunistic infection,16 malignancy including skin cancers and lymphomas,1423 demyelinating disorders such as multiple sclerosis,2431 heart disease such as congestive heart failure,32,33 liver disease such as hepatitis B13,3437 and C,3840 immunosuppressive disorder such as HIV,15,34,41,42 joint disease such as psoriatic arthritis, and vaccination status.10,13 A detailed social history should also be emphasized, specifically a past or current history of illicit substance and tobacco abuse, as well as pregnancy status.It has been established that psoriasis is associated with several comorbidities, including depression, psoriatic arthritis, and malignancy. Rapp et al43 reported that the impact of psoriasis on patient quality of life was comparable to that of other chronic conditions, such as heart failure, diabetes, and arthritis. Therefore, physicians should consider screening for these associated comorbidities including a screening for depression, particularly in patients with severe psoriasis. More recently, many publications have highlighted the link between psoriasis and conditions such as obesity, cardiovascular disease, diabetes, and metabolic syndrome. It is hypothesized that dysregulation of T-cells and over expression of pro-inflammatory cytokines such as TNF-α and IL-6, which leads to the hyperproliferation of keratinocytes and activation of neutrophils and endothelial cells within the skin, is also responsible for the increased prevalence of cardiovascular disease and metabolic syndrome in patients with psoriasis.44,45 In some cases the dermatologist may be the “first responder” and have a unique opportunity to evaluate for these associated conditions and subsequently refer patients to a primary care physician who can follow up with the crucial concomitant treatment. Only by approaching psoriasis as a potentially multisystem disorder can dermatologists facilitate optimal medical wellbeing.46,47Baseline laboratory studies should be performed and evaluated prior to initiating therapy with a biological agent, and these tests should include a comprehensive metabolic panel with liver function tests, a complete blood count, and a hepatitis panel. Baseline levels are important because hematological and metabolic disturbances have been reported (rarely) during biological therapy. Efalizumab, which was removed from the market in June of 2009 because of a potential risk to patients of developing progressive multifocal leukoencephalopathy (PML)—a rapidly progressing infection of the central nervous system that can lead to death or severe disability—has been shown to cause leukocytosis and possibly thrombocytopenia and hemolytic anemia5,4851; infliximab can cause elevated liver function tests3,52,53; and alefacept can cause a specific CD4+ leucopenia.4,54,55 Screening for antinuclear antibodies (ANA) prior to initiating a biological agent is controversial and should not preclude a patient from starting anti-TNF-α therapy.12,5662 The Centers for Disease Control and Prevention (CDC) recommends screening for tuberculosis (TB) prior to starting therapy with any TNF-α blocker and if positive, the patient is to begin prophylaxis TB therapy with isoniazid.6365  相似文献   

19.
New National Allocation Policy for Deceased Donor Kidneys in the United States and Possible Effect on Patient Outcomes     
Ajay K. Israni  Nicholas Salkowski  Sally Gustafson  Jon J. Snyder  John J. Friedewald  Richard N. Formica  Xinyue Wang  Eugene Shteyn  Wida Cherikh  Darren Stewart  Ciara J. Samana  Adrine Chung  Allyson Hart  Bertram L. Kasiske 《Journal of the American Society of Nephrology : JASN》2014,25(8):1842-1848
In 2013, the Organ Procurement and Transplantation Network in the United States approved a new national deceased donor kidney allocation policy that introduces the kidney donor profile index (KDPI), which gives scores of 0%–100% based on 10 donor factors. Kidneys with lower KDPI scores are associated with better post-transplant survival. Important features of the new policy include first allocating kidneys from donors with a KDPI≤20% to candidates in the top 20th percentile of estimated post-transplant survival, adding waiting time from dialysis initiation, conferring priority points for a calculated panel-reactive antibody (CPRA)>19%, broader sharing of kidneys for candidates with a CPRA≥99%, broader sharing of kidneys from donors with a KDPI>85%, eliminating the payback system, and allocating blood type A2 and A2B kidneys to blood type B candidates. We simulated the distribution of kidneys under the new policy compared with the current allocation policy. The simulation showed increases in projected median allograft years of life with the new policy (9.07 years) compared with the current policy (8.82 years). With the new policy, candidates with a CPRA>20%, with blood type B, and aged 18–49 years were more likely to undergo transplant, but transplants declined in candidates aged 50–64 years (4.1% decline) and ≥65 years (2.7% decline). These simulations demonstrate that the new deceased donor kidney allocation policy may improve overall post-transplant survival and access for highly sensitized candidates, with minimal effects on access to transplant by race/ethnicity and declines in kidney allocation for candidates aged ≥50 years.The current deceased donor kidney allocation policy has not changed substantially in >20 years.1 During this time, the gap between supply and demand has widened. Waiting time has become the dominant factor in allocation, and less emphasis has been placed on biologic criteria such as the degree of immune system sensitization or HLA matching. For minority candidates, such as African Americans, who have difficulty gaining access to the waiting list, delay in listing contributes to racial disparities in access to transplant.24 The current allocation system favors candidates who have waited the longest, but does not improve outcomes after transplant and discourages use of kidneys with a potentially shorter duration of functioning. These shortcomings have created inequities. Waiting times for blood type B candidates are much longer than waiting times for blood type A candidates.5,6 Kidneys with the potential to function longer may be allocated to candidates with shorter life expectancy; conversely, candidates with a longer estimated life span may be allocated kidneys with limited duration of functioning. These types of transplants result in high retransplant rates and increase the demand for donor kidneys. A new allocation policy was approved by the Organ Procurement and Transplantation Network (OPTN) on June 24, 2013.The new allocation policy risk-stratifies deceased donors using the kidney donor profile index (KDPI).7,8 The KDPI takes into account donor age, height, weight, ethnicity, history of hypertension and diabetes, cause of death, serum creatinine level, hepatitis C status, and donation after circulatory death status (Wait-Listed CandidatesKDPI≤0.20KDPI 0.21–0.34KDPI 0.35–0.85KDPI>0.85Local CPRA 100%Local CPRA 100%Local CPRA 100%Local CPRA 100%Regional CPRA 100%Regional CPRA 100%Regional CPRA 100%Regional CPRA 100%National CPRA 100%National CPRA 100%National CPRA 100%National CPRA 100%Local CPRA 99%Local CPRA 99%Local CPRA 99%Local CPRA 99%Regional CPRA 99%Regional CPRA 99%Regional CPRA 99%Regional CPRA 99%Local CPRA 98%Local CPRA 98%Local CPRA 98%Local CPRA 98%0 HLA mm top 200 HLA mm0 HLA mm0 HLA mmPrior living donorsPrior living donorsPrior living donorsLocal, regional adultLocal pediatricLocal pediatricLocalNational adultLocal top 20Local adultRegional0 HLA mm bottom 80Regional pediatricNationalLocal bottom 80Regional adultRegional pediatricNational pediatricRegional top 20National adultRegional bottom 80National pediatricNational top 20National bottom 80Open in a separate window0 HLA mm designates candidates with zero HLA mismatch at A, B, and DR loci; top 20 designates candidates in the top 20th percentile of survival; bottom 80 designates candidates not in the top 20th percentile of survival. Both the new and the current allocation policies give priority to candidates listed for simultaneous kidney and non-kidney organ transplants, including kidney-pancreas, kidney-liver, and kidney-heart transplants. This is not shown in the table above and is not included in the KPSAM modeling. Prior living donors represent a small number of candidates who are not included in the KPSAM modeling. SCr, serum creatinine; CVA, cerebrovascular accident.aKDPI is derived from the kidney donor risk index (KDRI) developed by Rao et al.7 The KDPI includes only the donor-specific elements of the KDRI, and is mapped to a reference population from the previous year, in order to yield percentiles. For KDRI, the reference population is all kidneys recovered for transplant between January 1, 2007, and December 31, 2009. The calculation is as follows: KDRI=exp(−0.0194×I[age<18 yr]×[age−18 yr]+0.0128×[age−40 yr]+0.0107×I[age>50 yr]+0.179×I[race=African American]+0.126×I [hypertensive]+0.130×I[diabetic]+0.220×[SCr−1 mg/dl] −0.209×I[SCr>1.5 mg/dl]×[SCr−1.5 mg/dl]+0.0881×I[cause of death=CVA]−0.0464×[{height−170 cm}/10]−0.0199×I[weight<80 kg]×[{weight–80 kg}/5]+0.133×I[donation after cardiac death] +0.240×I[hepatitis C]−0.0766, where I is equal to 1 if the condition is true and I is equal to 0 if the condition is false.bEPTS score=0.047×MAX (Age−25, 0)−0.015×Diabetes×MAX(Age–25,0)+0.398×Prior Organ Transplant−0.237×Diabetes×Prior Organ Transplant+0.315×log(Years on Dialysis+1)−0.099×Diabetes×log(Years on Dialysis+1)+0.130×(Years on Dialysis=0)−0.348×Diabetes×(Years on Dialysis=0)+1.262×Diabetes.As in the current system, points will be used to rank candidates in each category listed in and3).3). This scale was based inversely on the probability of receiving an organ offer. Other points will be awarded as in the current allocation policy (1 Other features of the current and new kidney allocation policies are compared in FactorPoints AwardedFor qualified time spent waiting1 per year (as 1/365 per day)Degree of sensitization (CPRA)0–202Prior living organ donor4Pediatric candidate if donor KDPI<0.351Pediatric candidate (age 0–10 yr at time of match) when offered a zero antigen mismatch4Pediatric candidate (age 11–17 yr at time of match) when offered a zero antigen mismatch3Share a single HLA-DR mismatch with donor1Share a zero HLA-DR mismatch with donor2Open in a separate windowThese points will be used to rank candidates in each of the categories listed in CPRA (%)Points0–19020–290.0830–390.2140–490.3450–590.4860–690.8170–741.0975–791.5880–842.4685–894.0590–946.719510.829612.179717.39824.49950.09100202.1Open in a separate window

Table 4.

Comparison of allocation concepts for current and new allocation policy
FeaturesPolicy
CurrentNew
SCD allocation (defined as KDPI≤0.85 for new policy)XX
DCD allocationX
ECD allocation (defined as KDPI>0.85 for new policy)XX
Payback systemX
Waiting time since listingX
Waiting time from dialysis initiationX
Waiting time points based on fractional yearsX
A2/A2B blood type donor to B candidates priority (local, regional, national)X
Highest scoring CPRA classificationX
Pediatric candidates cannot receive non-0 mm ECD offersX
Longevity matching (top 20th percentile survivors first offered kidneys with KDPI<0.20)X
Share KDPI<0.35 kidneys pediatric priority (donor age<35 yr for current policy)XX
Priority points for CPRA>19%X
Priority points for CPRA>79%X
National priority sharing for CPRA 100%, regional priority sharing for CPRA 99%, local priority for CPRA 98% candidatesX
Regional sharing for marginal kidneys (KDPI>0.85)X
Kidney pancreas/pancreas alone allocation policy: current (1)X
Kidney pancreas/pancreas alone allocation policy: future (1)X
Open in a separate windowSCD, standard criteria donor; DCD, donation after circulatory death; ECD, expanded criteria donor.In this study, we describe the final simulation models that were used to estimate how the current and new allocation systems allocate deceased donor kidneys. The results of these simulations were used to propose the new allocation system.  相似文献   

20.
Nonlinear Trajectory of GFR in Children before RRT     
Yichen Zhong  Alvaro Mu?oz  George J. Schwartz  Bradley A. Warady  Susan L. Furth  Alison G. Abraham 《Journal of the American Society of Nephrology : JASN》2014,25(5):913-917
GFR decline in patients with CKD has been widely approximated using linear models, but this linearity assumption is not well validated. We conducted a matched case-control study in children from the Chronic Kidney Disease in Children (CKiD) cohort ages 1–16 years with mild to moderate CKD to assess whether GFR decline follows a nonlinear trajectory as CKD approaches ESRD. Children (n=125) who initiated RRT (cases) during follow-up were individually matched by CKD stage at baseline and glomerular/nonglomerular diagnosis with children (n=125) who remained RRT-free when the corresponding case initiated RRT (controls). GFR trajectories were compared using log-linear and piecewise log-linear mixed effects models adjusted for baseline characteristics. From study entry to 18 months before RRT, GFR declined 7% faster among cases compared with controls. However, GFR declined 26% faster among cases compared with controls (P<0.001) during the 18 months before RRT. Nonlinearity in the rate of kidney function loss, which was shown in this cohort, may preclude accurate clinical prediction of the timing of RRT and adequate patient preparation. This study should prompt the characterization of predictive factors that may contribute to an acceleration of kidney function decline.GFR is a key measurement of kidney function, and the degree of GFR decline over time is a reflection of the severity of CKD progression. GFR decline has been approximated as linear or log-linear in most analyses of progression, an assumption that has been consistent with available data.14 However, many studies rely on relatively short follow-up periods and few repeated measures. Given the convenience of assuming a linear GFR trajectory, which results from the ease of modeling and interpreting linear slopes, few studies have sought to validate the linearity assumption and explore the possibility of nonlinear GFR decline. However, nonlinearity in GFR decline has been observed in some epidemiologic studies,57 and the implications on the risk for adverse outcomes have generated interest.8 A CKD cohort study in France found that about one half of its patients experienced nonlinear GFR decline during the last year before dialysis.5 A study by Li et al.9 used a flexible approach to model nonlinearity in GFR trajectories. Li et al.9 found evidence of nonlinear GFR trajectory behavior in adult patients with CKD, and furthermore, the probability of having nonlinear features in an individual trajectory was associated with known risk factors for CKD progression. O’Hare et al.10 found several distinct nonlinear patterns of GFR decline in the 2 years before dialysis initiation in Veterans Affairs patients.Clinical strategies and subsequent patient response to care could potentially benefit from new insights into the variable paths of progression in patients with CKD.10,11 The question of whether characterizing the nonlinearity in the GFR trajectory can assist the identification of risk groups for outcomes, such as ESRD, remains unexplored. The implications on future outcomes of an increased rate of GFR decline could inform clinical decisions about screening frequencies, treatment, or preparation for RRT.The Chronic Kidney Disease in Children (CKiD) study is an ongoing cohort study of children with CKD who, at baseline, had an eGFR between 30 and 90 ml/min per 1.73 m3 and were ages 1–16 years. An end point of the study is RRT defined as transplant or dialysis. To determine whether trajectories of GFR accelerate before RRT, we nested a case-control study, in which cases were children observed to have received RRT and controls were children with CKD who remained RRT-free at the time when the corresponding case initiated RRT.There were 147 children who experienced RRT during follow-up. Each case was matched individually to an eligible control at the time of the case occurrence. The matching factors included baseline CKD stage, glomerular/nonglomerular diagnosis, and, through design, the amount of follow-up time from study entry. Matching was done without replacement, and 22 cases were excluded from the analyses, because no appropriate control was available. We used a random sequence to determine the order of matching. The analysis was, thus, based on 125 matched case-control pairs. Demographic and clinical characteristics of cases and controls at baseline are shown in CharacteristicsCases (n=125)Controls (n=125)Age, yr12.64 (9.23–14.53)12.33 (8.71–14.74)Sex (girls), N (%)38 (30.4)57 (45.6)Race (nonwhite), N (%)51 (40.8)36 (28.8)Urine protein/creatinine ratio1.74 (0.48–4.04)0.60 (0.26–1.76)Proteinuria, N (%) 0.2≤protein/creatinine ratio<256 (46.7)71 (59.7) Protein/creatinine ratio≥251 (42.5)23 (19.3)Baseline GFRa32.21 (26.43–39.64)35.77 (27.86–43.78)Glomerular diagnosis, N (%)a47 (37.6)47 (37.6)Open in a separate windowMedian (interquartile range) unless otherwise indicated.aBaseline GFR and glomerular/nonglomerular diagnosis were matching factors.We compared the GFR trajectories using log-linear and piecewise log-linear mixed effects models, with the piecewise model specified to allow a change of the GFR slope at 18 months before RRT. Models were adjusted for baseline characteristics, including age, race, sex, and proteinuria status. and33 show the adjusted results from the mixed effects model analyses. The Akaike Information Criterion indicated that the piecewise log-linear model (including a spline or changing slope at 18 months before RRT) was a better fit to the data than the log-linear model that assumed a single slope across the entire period of observation. The GFR of cases declined at an adjusted rate of 6.8% per year (P <0.001) during the time before the spline in the earlier period of observation and 32.4% per year (P <0.001) after the spline within 18 months of RRT. The GFR of controls did not change significantly (P=1.00) before the spline and declined at an adjusted rate of 9.0% (P <0.001) after the spline. Although the rates of GFR decline comparing cases with controls differed by only 7% before the spline, the GFR of cases declined 26% faster (P <0.001) compared with controls within 18 months of RRT, suggesting an acceleration in the GFR decline during this period in the case group. This acceleration, which was quantified by the piecewise log-linear mixed effects model, could be clearly seen from the data and nonparametric smooth fits (Figure 1). The variability around the piecewise log-linear fit was assessed by the root mean square error (RMSE) and found to be similar between cases and controls (RMSE for controls=0.303; RMSE for cases=0.303), indicating an equally good fit. When a single slope was fit to the data, the GFR decline rate for cases was overestimated before the spline and considerably underestimated within 18 months of RRT. To assess whether the acceleration in decline was a function of the log scale, models were rerun with GFR in the natural scale. The results showed similar nonlinear patterns but a poorer model fit to the data.

Table 2.

The adjusted expected percent GFR change rates in the log-linear mixed effects model
Case GroupAdjusted % GFR Change per YearSEM (%)P Value
Controls−3.21.20.01
Cases−18.20.9<0.001
Cases-controls−15.51.3<0.001
AIC260.78
Open in a separate windowParameter estimates from the models are provided in Supplemental Appendix II. All results were adjusted for baseline characteristics, including age, race, sex, and proteinuria status. AIC, Akaike Information Criterion.

Table 3.

The adjusted expected percent GFR change rates in the piecewise log-linear mixed effects model
Case GroupBefore 18 mo before RRT of CasesAfter 18 mo before RRT of CasesDifference between Early and Late Slopesa
Adjusted % GFR Change per YearSEM (%)P ValueAdjusted % GFR Change per YearSEM (%)P ValueAdjusted % GFR Change per YearSEM (%)P Value
Controls0.31.50.87−9.02.5<0.0019.23.30.01
Cases−6.81.3<0.001−32.41.3<0.00127.42.0<0.001
Cases-controls−7.01.9<0.001−25.72.5<0.001
AIC149.14
Open in a separate windowParameter estimates from the models are provided in Supplemental Appendix II. All results were adjusted for baseline characteristics, including age, race, sex, and proteinuria status.aDifference resulting from the piecewise linear mixed effects model estimated in the log scale and then exponentiated.Open in a separate windowFigure 1.Nonlinear GFR decline before RRT can be approximated with a piece-wise log-linear model. A and B show the smooth fit of log GFR over time for cases of RRT and matched controls anchoring at the RRT onset time of cases. C and D show the fit from the adjusted log-linear and adjusted piecewise log-linear mixed effects models for cases of RRT and matched controls anchoring at the RRT onset time of cases. Models were adjusted for baseline characteristics including age, race, sex, and proteinuria status.Our results show that, although linear or log-linear GFR decline is a convenient assumption for longitudinal studies of CKD progression, individuals experience periods of accelerated decline. Li et al.9 showed that patients in the African American Study of Kidney Disease and Hypertension experienced a variety of nonlinear progression patterns. O’Hare et al.10 classified CKD patients who progressed to dialysis into four GFR trajectory categories and found evidence that patients with mild to moderate CKD experienced more rapid renal function deterioration in the 2 years before reaching long-term dialysis. In the current study assessing progression in children with CKD, we found similar results, indicating that RRT events are preceded by a period of accelerated decline in GFR. It is likely that this period of precipitous loss in kidney function is a key factor in the determination of the timing of RRT. An acceleration of GFR decline may be a primary feature of a worsening clinical profile that prompts a clinician to initiate dialysis or transplant. The question arises as to what contributes to accelerated kidney function loss. A primary epidemiologic challenge is to find predictors that antecede the acceleration and are amenable to intervention to prevent or delay such accelerated loss and RRT. Clearly, these results and the questions that they raise speak to a need for additional investigations of CKD progression in various populations, with care taken to appropriately characterize changing levels of factors that are known predictors of CKD progression. The timing of potential insults to the kidney (e.g., loss of control of BP) may hold important information concerning the patterns of CKD progression and nonprogression. O’Hare et al.10 found that rates of recommended pre-ESRD care were lower for those patients experiencing the most rapid progression before dialysis initiation. Ambrogi et al.5 suggested that nonlinear patterns in GFR decline might create difficulty in estimating the timing of dialysis.These results may also highlight the coarseness of current methods for assessing the impact of risk factors on CKD progression, which mainly rely on the assumption of linear decline in kidney function. Analyses assuming linear decline average over nonlinear patterns that speak to the true nature of the exposure–outcome relationships. More sensitive analyses may be needed to characterize the heterogeneity in the patterns that describe CKD progression and assess the impact of often changing values of the exposure. Improvements in how we characterize patterns of progression could lead to new approaches to clinical care, because accelerations in kidney function loss may complicate the timing of RRT and pre-ESRD care.7,10There are several strengths of this study. We drew from a well characterized cohort of children with CKD with directly measured GFR at the first two annual study visits and all even visits thereafter. The CKiD study also has an internally derived estimating equation for GFR to capture kidney function in odd visit years of the study, thereby providing regular GFR assessments for characterizing nonlinear patterns of GFR decline. The CKiD study has longitudinal data for up to 6 years of follow-up, and the multicenter setting with 43 clinical sites provides a sample of children highly representative of the pediatric CKD population in care in the United States. By adopting the case-control design, we were able to compare the nonlinearity of the GFR trajectory before RRT with the expected trajectory in comparable children who had not yet experienced RRT.There are also notable limitations to the current analysis. There were only 125 case-control pairs, and our GFR assessments were annual, limiting the degree to which heterogeneity in progression to RRT could be assessed among the case group. As has been reported previously, there is likely variation in GFR patterns before RRT.10 However, what is clear from the current study is that, on average, children approaching RRT experience acceleration in their loss of kidney function. Another consideration is the assumption of a break in linearity at 18 months before RRT, which provided sufficient data before and after the spline for our analyses but is an oversimplification of what is likely a more prolonged period of acceleration in GFR decline. However, our choice of 18 months before RRT to examine changes in the rate of GFR decline is consistent with other studies that have noted similar rapid declines in kidney function within 2 years of dialysis.10,12 Finally, it should be noted that, although cases and controls were matched, the models in and33 did not cluster on the matched pairs. Our final model provided practically identical results to a model including an additional random effect for case-control pair, and it had modestly higher precision.  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