首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
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   

2.
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   

3.
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   

4.
5.
6.
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   

7.
8.
Actinic keratosis (AK), the initial lesion in a disease continuum that may progress to squamous cell carcinoma, often begins with ultraviolet B light-induced photo damage and increases in prevalence with age. Topical 5-fluorouracil (5-FU) for the treatment of widespread multiple AK lesions has cure rates of more than 90 percent. The associated skin irritation, however, may lead patients to prematurely discontinue treatment. To improve treatment efficacy and tolerability, 5-FU cream 0.5%, a novel Microsponge®-based formulation, was developed. In the elderly population, 5-FU cream 0.5% may be preferable because of its convenient (once daily) administration and its lower potential for irritation and systemic absorption, which may impact adherence to therapy and, thus, possibly increase cure rates.Actinic keratoses (AKs), also called solar keratoses, occur more commonly in individuals with fair skin who have had extensive exposure to the sun.1,2 The prevalence of AK lesions in adults increases with age: less than 10 percent for 20- to 29-year-olds, approximately 80 percent for 60- to 69-year-olds, and greater than 80 percent for 70-year-olds and older.3,4 In the first five decades of life, the prevalence of AK lesions in men is almost twice that in women; however, after age 50, prevalence rates are similar between men and women.3,4 A study using data from the National Ambulatory Medical Care Survey from 1990 to 1999 found that 47 million visits to physicians involved an AK diagnosis, and more than 80 percent of patients with AK diagnoses were at least 50 years old.5 AK was the second most frequent diagnosis made by dermatologists.5 Because surveys and studies typically include only patients seen in clinics or by physicians, the actual number of patients with AK is probably higher.6 The incidence of AK is higher in individuals living close to the equator, but is increasing in men and women in general.7 8

Table 1

Risk factors for actinic keratoses8
Fair skin, with light-colored eyes and red or blonde hair
Inability to tan, tendency to sunburn (Fitzpatrick I-II)
Chronic, cumulative exposure to ultraviolet radiation
Advancing age
Outdoor occupation or leisure activities
Residence in sunbelt latitudes
Previous nonmelanoma skin cancer (SCC and BCC)*
Immunosuppression
Genetic predisposition
Open in a separate window*
SCC =
squamous cell carcinoma
BCC =
basal cell carcinoma
  相似文献   

9.
Objective: Following Mohs surgery, medium-to-large defects on the central forehead can often be complicated to surgically reconstruct. In this paper, the authors discuss possible central forehead reconstructions and report their successful experience employing a simple primary vertical linear closure with a special technique to demarcate forehead rhytides rather than performing an overly complicated flap or graft. Case report: The patient was a 57-year-old man who presented with a broad superficial basal cell carcinoma that required treatment with Mohs surgery. For the resulting defect, the authors elected to perform a complex linear repair taking advantage of substantial side-to-side laxity in the supraperiosteal plane and carefully labeling and matching each forehead rhytide across the defect as the wound was sutured. Conclusion: The findings of this case demonstrate that medium-to-large wounds of the central forehead can be aesthetically repaired with a simple primary vertical linear closure. Carefully mapping and labeling horizontal forehead rhytides with a sterile surgical marking pen prior to anesthesia ensures accurate approximation during wound closure.Due to the prominence and broad surface of the central forehead, aesthetic reconstruction of medium to large forehead defects is imperative. Commonly utilized flaps include the island pedicle flap, rotation flaps, A-to-T flap, and O-to-T flaps (1-3

TABLE 1

Commonly utilized flaps
TYPE OF REPAIRADVANTAGESDISADVANTAGES
Island pedicle flapRich vascularization; avoids medial eyebrow displacementDisruption of forehead anatomy; Possible large hypopigmented triangular scar
Rotation/advancement flapDraws from surrounding areas of skin laxityComplex incision lines/scars; extensive undermining required; poor motion of forehead
Full thickness skin graftObtains skin from distant areas of increased laxityBad color and texture match; leaves patchy scar
Z-plastyHides parts of the scar in horizontal relaxed skin tension lines“Mark of Zorro” scar
Vertical linear repairPreserves natural forehead anatomy and rhytides; simple repair with aesthetic resultLong scar due to dog ears
Open in a separate windowThe island pedicle flap can be considered as it is richly vascularized by the frontalis perforators and is useful when trying to avoid medial displacement of the eyebrows.4 The island pedicle flap is a better color and texture match than a graft on the central forehead; however, the natural forehead anatomy is often disrupted and a large hypopigmented triangular scar can remain.4Rotation and advancement flaps, such as the A-to-T flap and O-to-T flap can also be useful when it is difficult to achieve side-to-side movement at the immediate wound site.5 These flaps draw from surrounding areas of increased skin laxity, but involve complex incision lines and a risk of disturbing important neurovascular structures when undermining in the subcutaneous plane.3For certain defects, Z-plasties can be used to take advantage of the horizontal relaxed skin tension lines of the forehead by reorienting portions of vertical incision lines. However, a poor outcome is quite unaesthetic and when performed on longer incisions may leave the patient with a noticeable “mark of Zorro.”6For medium to large wounds located in the central third of the forehead, a primary vertical linear repair that preserves natural forehead anatomy provides a much simpler way to create equally if not better aesthetic results than local flaps and Z-plasties.5 The central forehead’s relative lack of neurovascular structures facilitates the deep plane undermining required for a vertical linear repair even for larger defects.2 Often, advancement, rotation, and pedicle flaps can prove to be overly complicated and time-consuming to perform due to the extensive undermining required, the vascularity of the forehead, and the lack of ease of movement of the forehead.  相似文献   

10.
11.
12.
Clinicians inevitably encounter patients with complaints and concerns about the quality of their care. This causes some to experience anxiety, fear, anger, resentment, guilt, and depression, especially when they believe they may have erred or caused harm. Lack of customer-service training and experience may contribute to these emotions. The “BLAST” technique is a complaint-resolution method that is useful in patient care and as a clinical teaching tool. The mnemonic stands for: Believe (what the patient is saying), Listen (actively, to assess and restate the patient’s unmet expectations), Apologize (for the patient’s unmet expectations), Satisfy (the patient), and Thank (the patient for expressing his/her concerns and providing a second chance to satisfy the patient). The technique appears to help clinicians become more at ease and confident when handling patient complaints. This may be especially helpful for clinicians who must routinely interact with post-treatment and post-procedure patients who commonly express surprise, concern, or complaints about their results and healing. BLAST may be an effective teaching tool enabling students, residents, and clinicians to become more comfortable and adept at working with displeased and concerned patients.Clinicians inevitably encounter patients with complaints and concerns about the quality of their care. Some experience anxiety, fear, anger, resentment, guilt, and depression when their care is criticized,1,2 especially when they believe they may have erred3 or caused harm.4 These reactions may, in part, be due to a lack of customer-service training5 and interfere with clinician effectiveness.1The BLAST technique is a complaint-resolution method developed by Albert Barneto. The mnemonic stands for Believe, Listen, Apologize, Satisfy, and Thank (6 This article describes its usefulness in patient care and as a clinical teaching tool. Anecdotal experience suggests its use helps clinicians remain more calm and effective when working with displeased patients.

TABLE 1

The BLAST technique is a complaint-resolution method
BBelieve
LListen
AApologize
SSatisfy
TThank
Open in a separate window  相似文献   

13.
14.
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
VariablePlaceboSitaxentanNifedipine
BaselineWeek 6BaselineWeek 6BaselineWeek 6
24-hr proteinuria (g/d)2.06±0.382.00±0.332.07±0.341.46±0.26a1.95±0.301.99±0.33
Protein-to-creatinine ratio (mg/mmol)155±31153±27157±28114±23b155±27152±29
Mean arterial pressure (mmHg)94.6±2.294.3±1.794.4±1.890.7±1.8c95.5±2.091.7±1.7c
Systolic BP (mmHg)125.4±2.7124.2±1.9124.3±2.2120.7±1.9c125.7±2.4120.7±1.6c
Diastolic BP (mmHg)77.9±1.577.5±1.277.9±1.374.3±1.3a78. 9±1.575.7±1.2a
PWV (m/s)7.7±0.38.0±0.48.0±0.37.6±0.3c7.9±0.37.6±0.3c
Central augmentation index (%)20±220±220±215±2a19±217±2
Plasma ET-1 (pg/ml)3.6±0.53.7±0.63.6±0.53.7±0.53.5±0.53.5±0.5
Urine ET-1 (pg/ml)4.5±0.44.7±0.45.1±0.44.2±2.1c5.1±0.44.7±0.4
Open 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   

15.
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   

16.
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   

17.
A Review of Epidermal Maturation Arrest: A Unique Entity or Another Description of Persistent Granulation Tissue?     
Maria C. Kessides  Amor Khachemoune 《The Journal of clinical and aesthetic dermatology》2014,7(12):46-50
  相似文献   

18.
High Prevalence of Sickle Cell Trait in African Americans with ESRD     
Vimal K. Derebail  Patrick H. Nachman  Nigel S. Key  Heather Ansede  Ronald J. Falk  Abhijit V. Kshirsagar 《Journal of the American Society of Nephrology : JASN》2010,21(3):413-417
Sickle cell trait (HbAS) associates with impaired urinary concentration, hematuria, and renal papillary necrosis, but its prevalence among African Americans with ESRD is unknown. We performed a cross-sectional study reviewing available hemoglobin phenotypes for 188 of 206 adult African-American patients receiving renal replacement therapy in four dialysis units. Results from the state newborn screening program in corresponding counties provided the local population prevalence of sickle trait among African Americans. Compared with the general African-American population, HbAS was twice as common among African Americans with ESRD (15% versus 7%, P < 0.001). Prevalence of hemoglobin C trait (HbAC) was similarly more common (5% versus 2%, P < 0.01). The higher prevalence of HbAS and HbAC in the ESRD population raises the possibility that these hemoglobinopathies contribute to a decline in kidney function, either alone or in conjunction with other known risk factors for renal disease. The potential effect of HbAS on the development and progression of CKD and its effect on the course and management of patients with ESRD deserve further study.Sickle cell trait (HbAS) is present in 7 to 9% of African Americans1,2 and has typically been described as a benign carrier state with little effect on the health of affected individuals. Although uncommon, several adverse effects of HbAS have been reported in settings of low oxygen tension or high oxygen demand, including splenic infarction at high altitude, sudden death with extreme physical exertion, venous thromboembolism, and glaucoma from anterior chamber hemorrhage.1,37 The low oxygen content of the renal medulla provides a propitious setting for intravascular sickling. HbAS has been associated with fewer and disrupted vessels of the vasa recta,8,9 which likely translates clinically to the highly prevalent impaired urinary concentration.3,6,7,10,11 Renal microvascular obstruction also occurs with HbAS, presenting most frequently as asymptomatic hematuria and most dramatically as renal papillary necrosis.3,6,7,10,11The rare renal medullary carcinoma is seen almost exclusively among HbAS patients.6,11,12In epidemiologic studies, the presence of HbAS has been associated with microalbuminuria and proteinuria, particularly among diabetic men,13,14 and African Americans with autosomal dominant polycystic kidney disease (ADPKD) and HbAS have been shown to progress to ESRD more rapidly than those without the trait.15 With its associated structural and physiologic changes, HbAS could adversely affect renal function, especially in the setting of comorbid disease, and may represent a potential risk factor for kidney disease.16,17 We postulated that HbAS may be more common among African Americans with ESRD and examined the prevalence of HbAS in a group of African Americans receiving renal replacement therapy.Hemoglobin phenotyping was performed on 188 of the 206 African-American patients receiving treatment through four of our affiliated dialysis centers, 172 of whom were receiving hemodialysis and 21 receiving peritoneal dialysis. We obtained newborn hemoglobinopathy screening results in the corresponding three North Carolina counties from the inception of the newborn screening program to the planned date of the study. This included 6729 African-American individuals born between January 1, 1994 and November 30, 2008 who were used to determine the local population prevalence.Among the tested African-American ESRD patients, 28 (14.9%) patients had HbAS, 9 (4.8%) were heterozygous for hemoglobin C [hemoglobin C trait (HbAC)], and 1 (0.5%) was heterozygous for β-thalassemia (β-thalassemia minor). In comparison, the local population prevalence among screened newborns was 7.1% (P < 0.001) for HbAS and 1.9% for HbAC (P < 0.01) (Figure 1).Open in a separate windowFigure 1.Prevalence of hemoglobin variants from North Carolina newborn screening data and among ESRD patients. North Carolina screening data from newborn screening of African-American live births (n = 6729) from January 1, 1994 to November 30, 2008 were obtained for the three counties in which African-American ESRD patients (n = 188) from four dialysis clinics were based. *Fisher''s exact test with Bonferroni correction for repeat testing. HbAS, sickle cell trait; HbAC, hemoglobin C trait.We also sought to determine if there were any major differences among the ESRD patients when separated by hemoglobin phenotype (CharacteristicHbAAb (%) (n = 150)HbAS (%) (n = 28)HbAC (%) (n = 9)PAge, years (SD)58.4 (14.8)59.8 (12.5)59.7 (16.8)0.9Gender, n (SD)0.9    female71 (47.3)13 (46.4)5 (55.6)    male79 (52.7)15 (53.6)4 (44.4)Age of ESRD onset, years (SD)54.1 (15.5)54.0 (13.8)54.5 (18.6)1.0Dialysis vintage, median years (interquartile range)3.0 (1.2 to 5.8)5.5 (2.2 to 8.5)5.5 (1.8 to 6.5)0.05Modality0.8    in-center hemodialysis137 (91.3)25 (89.3)9 (100)    peritoneal dialysis13 (8.7)3 (10.7)0Cause of ESRDc0.1    diabetes mellitus59 (39.3)10 (35.7)6 (66.7)    hypertension52 (34.7)15 (53.6)1 (11.1)    GN24 (16.0)1 (3.6)0    cystic disease3 (2.0)00    other12 (8.0)2 (7.1)2 (22.2)Open in a separate windowaOne patient heterozygous for β-thalassemia (β-thalassemia minor, β-thalassemia trait) is not included here. This patient was a 32 year-old female on hemodialysis with hypertension as cause of ESRD and onset of ESRD at age 22.bHbAA, normal adult hemoglobin phenotype.cAs reported in Medicare CMS-2728 form.Differences were noted in ascribed cause of ESRD, although these did not reach statistical significance (P = 0.1). Diabetes mellitus was the cause of ESRD in over one third of patients with normal adult hemoglobin phenotype and HbAS and two thirds of patients with HbAC. More patients with HbAS had hypertension as their cause of ESRD. Only one (4%) patient with HbAS had GN, compared with 24 (16%) from the portion of patients with normal adult hemoglobin phenotype. Only three patients in the entire cohort were identified as having cystic kidney disease, all of whom had normal hemoglobin phenotypes. The one 32 year-old female patient with β-thalassemia minor and hypertensive ESRD at age 22 was not included in the above analyses.In this study of 188 African-American patients receiving dialysis, we found that the prevalence of HbAS was more than twice that of the general population, present in one in seven ESRD patients. We also found that HbAC was more common in the dialysis patients.To our knowledge, this study is the first to evaluate the prevalence of these hemoglobinopathies among African-American ESRD patients relative to an appropriate reference population. Our analysis, which was based on an a priori hypothesis, is exploratory in nature. HbAS, as examined in a group of prevalent ESRD patients, could be a risk factor or risk indicator for ESRD or merely associated at a statistical level. Acknowledging that we cannot exclude residual confounding, we propose potential mechanisms for our observation.First, HbAS may directly lead to loss of renal function. Episodic sickling could lead to chronic parenchymal ischemia and eventually fibrotic changes as reported in autopsy and biopsy studies.18 Although not proven to be causative, the presence of HbAS has even been reported in the setting of GN.19,20 Second, HbAS could accelerate the effects of another process. Medullary structural and physiologic abnormalities provide a background pathology in which a primary disease would have an accelerated effect. Although HbAS may not be sufficient to cause ESRD by itself, it could contribute to progression of renal insufficiency to ESRD in the presence of additional factors, such as diabetes or hypertension. As noted previously, microalbuminuria and proteinuria have been reported in higher frequency among diabetic men with HbAS,14 further suggesting this possibility.If HbAS is a cofactor for renal disease, identifying HbAS in patients otherwise at risk for chronic kidney disease could be important for preventive measures. Overt sickle cell disease (HbSS) is associated with lesions of glomerular hypertension, and angiotensin converting enzyme inhibitors improve proteinuria in these patients.21 Similar pathology may occur in the HbAS state and identifying these patients would allow aggressive monitoring for albuminuria and early intervention with antiproteinuric agents.The presence of abnormal hemoglobin, particularly HbAS, may also affect the course and care of ESRD patients. HbAS may be an independent risk factor among African Americans for venous thromboembolism1,5 and could add to the already high risk for pulmonary embolism among ESRD patients.22 Additionally, this predisposition for thrombosis could affect arteriovenous fistula failure and access loss, already known to occur at higher rates among African Americans.23 African Americans with ESRD also appear to require larger doses of erythropoiesis stimulating agents (ESAs) to achieve their hemoglobin targets.24 Results of the Correction of Hemoglobin and Outcomes in Renal Insufficiency study suggested higher target hemoglobin may be associated with more cardiovascular outcomes,25 and adverse outcomes were seen primarily in those who failed to reach their target hemoglobin.26 The presence of HbAS and other hemoglobinopathies may play a role in this relative resistance, placing these patients at higher risk with increasing ESA exposure.Heterozygosity for HbAC was also twice as common in this cohort compared with the geographically matched African-American population and to the reported national prevalence.2 Hemoglobin C is more likely to precipitate, and when present with hemoglobin S it leads to a syndrome similar to but less severe than HbSS disease. Heterozygosity is thought to be clinically silent.27 The elevated prevalence of HbAC in our cohort is of unclear significance but may have similar consequences as those of HbAS.Of note, mean age of onset of ESRD in our study population was similar among all groups, whereas dialysis vintage was higher in the groups with either hemoglobinopathy by a median of 2.5 years. The small sample size and borderline statistical significance limit our ability to conjecture a potential explanation or draw meaningful conclusions from this difference. However, the greater length of vintage, in part, could influence our assessment of prevalence. The extended presence of those with hemoglobinopathies in this ESRD population could increase the prevalence of HbAS when surveyed in a cross-sectional manner.The findings of this study must be interpreted in the context of several limitations. Primarily, the assessment of population prevalence of hemoglobinopathies via newborn screening results may not be accurately reflective of the population from which our ESRD cohort is derived. North Carolina newborn screening results include births occurring after 1994, and our cohort is of a different era. Any large migration into or out of these regions would lead to prevalence discrepancies between the time of our cohort and the advent of newborn screening. However, because our measured prevalence is similar to that found in the general African-American population nationally,1,2 we do feel confident in our assessment of the baseline population prevalence.Our findings may not translate to all African-American ESRD populations. The aforementioned study of African-American ADPKD patients15 did include an assessment of HbAS in ESRD patients without ADPKD, finding it in only 6 of their 80 patients (7.5%). Although the local prevalence of HbAS was not evaluated, this discrepancy from our study does implicate that our findings should be confirmed in another cohort, perhaps larger and more geographically diverse.Lastly, the cross-sectional design prevents the determination of the exact nature of the observed associations. Initial cross-sectional findings are primarily useful for informing subsequent prospective studies. The size of the cohort is also relatively small and geographically compact, such that any familial clustering might provide an exaggeration of the prevalence.The prevalence of abnormal hemoglobin, HbAS and HbAC in this African-American ESRD cohort, was found to be over twice that of the baseline African-American population in the same geographic region. The high prevalence of these hemoglobinopathies suggests that they may contribute to progression to ESRD by providing a background of renal injury. Our findings also raise questions as to how the presence of HbAS or HbAC may affect management of ESRD patients. Longevity of hemodialysis access and response to ESAs may be altered. Although these results require confirmation, knowledge of a patient''s hemoglobin status may be important to the management of ESRD patients and before ESRD may even identify individuals who would benefit from aggressive interventions to attenuate progression of renal disease.  相似文献   

19.
Kidney Stones Associate with Increased Risk for Myocardial Infarction     
Andrew D. Rule  Veronique L. Roger  L. Joseph Melton  III  Eric J. Bergstralh  Xujian Li  Patricia A. Peyser  Amy E. Krambeck  John C. Lieske 《Journal of the American Society of Nephrology : JASN》2010,21(10):1641-1644
Kidney stones are a risk factor for chronic kidney disease (CKD), which, in turn, is a risk factor for myocardial infarction (MI). The objective of this study was to determine whether kidney stones associate with an increased risk for MI. We matched 4564 stone formers (1984 through 2003) on age and gender with 10,860 control subjects among residents in Olmsted County, Minnesota. We identified incident MI by diagnostic codes and validated events by chart review through 2006. We used diagnostic codes to determine incidence of kidney stones and presence of comorbidities (CKD, hypertension, diabetes, obesity, dyslipidemia, gout, alcohol dependence, and tobacco use). During a mean of 9 years of follow-up, stone formers had a 38% (95% confidence interval 7 to 77%) increased risk for MI, which remained at 31% (95% confidence interval 2% to 69%) after adjustment for CKD and other comorbidities. In conclusion, kidney stone formers are at increased risk for MI, and this risk is independent of CKD and other risk factors.Kidney stones have been identified as a risk factor for chronic kidney disease (CKD).1 Of great concern with the development of CKD is an increased risk for coronary heart disease. Reduction in both GFR and albuminuria is associated with coronary heart disease in the general population.2,3 It was hypothesized that the increased risk for CKD in individuals with kidney stones would lead to an increased risk for coronary heart disease. Alternatively, biological pathways that cause calcium kidney stones may also contribute to coronary artery calcification, a risk factor for coronary heart disease events. Myocardial infarction (MI) surveillance in Olmsted County, Minnesota, has been in place since 1979.47 We sought to determine whether kidney stone formers in Olmsted County were at an increased risk for MI and whether this risk is related to the development of CKD.A total of 5081 incident stone formers and 14,144 matched control subjects were identified from the Olmsted County general population between 1984 and 2003. The International Classification of Diseases, Ninth Revision (ICD-9) code used to identify stone formers was 592 (calculus of kidney and ureter) in 4467 (88%), 594 (calculus of lower urinary tract) in 613 (12%), and 274.11 (uric acid nephrolithiasis) in one (<1%). Prevalent MIs were similar between stone formers (n = 178 [3.5%]) and control subjects (n = 489 [3.5%]). After exclusion of individuals with prevalent MI and those who lacked clinic visits at least 90 days after the index date, 4564 stone formers and 10,860 control subjects were followed for incident MI. Only 2.1% of these individuals were nonwhite, consistent with the racial distribution of the community (96% white in 1990). As a consequence of the matching, stone formers and control subjects were similar with respect to age (mean 44.6 versus 44.4 year), gender (59 versus 58% male), length of medical record documentation before the index date (mean 18.4 versus 21.9 years), and length of follow-up to last clinic visit or death (mean 8.7 versus 9.0 years). As shown in ComorbidityStone Formers (n = 4564; n [%])Control Subjects (n = 10,860; n [%])PCKD116 (2.5)200 (1.8)0.0051Hypertension848 (18.6)1748 (16.1)0.0002Diabetes420 (9.2)771 (7.1)<0.0001Obesity1017 (22.3)2155 (19.9)0.0007Dyslipidemia860 (18.8)1802 (16.6)0.0007Gout135 (3.0)255 (2.4)0.028Alcohol dependence235 (5.2)777 (7.2)<0.0001Tobacco use619 (13.6)1598 (14.7)0.063Open in a separate windowThere were incident MIs in 96 stone formers and 166 control subjects. Stone formers were at increased risk for MI in analyses that were unadjusted (hazards ratio [HR] 1.43; 95% confidence interval [CI] 1.11 to 1.84; Figure 1), adjusted for age and gender (HR 1.38; 95% CI 1.07 to 1.77), further adjusted for CKD (HR 1.38; 95% CI 1.07 to 1.77), and fully adjusted for all comorbidities (HR 1.31; 95% CI 1.02 to 1.69). The risk for MI remained with adjustment for the number of comorbidities (excluding alcohol dependence), age, and gender (HR 1.35; 95% CI 1.05 to 1.73). By coding subgroup, the risk for MI adjusted for age and gender was statistically significant for ICD-9 code 592 (HR 1.40; 95% CI 1.07 to 1.84) but not for ICD-9 code 594 (HR 1.24; 95% CI 0.70 to 2.21).Open in a separate windowFigure 1.Increased risk for MI in stone formers than in controls among Olmsted County, Minnesota residents.After adjustment for age and gender, most comorbidities were associated with MI: CKD (HR 2.97; 95% CI 1.86 to 4.72), hypertension (HR 1.54; 95% CI 1.18 to 2.01), diabetes (HR 2.18; 95% CI 1.61 to 2.94), obesity (HR 1.77; 95% CI 1.38 to 2.27), dyslipidemia (HR 1.65; 95% CI 1.27 to 2.15), gout (HR 1.40; 95% CI 0.88 to 2.22), alcohol dependence (HR 1.22; 95% CI 0.76 to 1.95), and tobacco use (HR 2.23; 95% CI 1.67 to 2.96). After exclusion of individuals with these comorbidities at baseline, there remained 2366 stone formers with 25 incident MIs and 5818 control subjects with 42 incident MIs, and the risk for MI with kidney stones remained elevated but not statistically significant (HR 1.50; 95% CI 0.92 to 2.47). There was no detectable interaction between these comorbidities and the risk for MI with kidney stones (P ≥ 0.10 for each comorbidity × stone former interaction).We found stone formers to be at a 38% increased risk for MI. A consideration is that this association reflects shared risk factors for both MI and kidney stones, namely, hypertension, diabetes, obesity, and dyslipidemia8; however, the risk for MI in stone formers remained elevated with adjustment for these and other known risk factors for MI, including CKD. This finding adds to the literature that kidney stones should be viewed as a metabolic disorder with clinical relevance beyond symptomatic urinary tract obstruction.9A Medline (Ovid Technologies) search of English-language human studies on February 2010 with the terms “kidney/renal stone(s)/calculi or nephrolithiasis or urolithiasis” and “MI or coronary heart/artery disease” revealed 67 articles. Among these articles, there were six relevant studies, with most having small samples sizes.8,1014 Several studies showed increased risk for MI in stone formers,8,10,12,14 and other studies showed no association.11,13 Lack of effective calcification inhibitors may be a common mechanism linking coronary artery calcification to calcium kidney stones (80% of stone formers).15 High-dosage calcium supplements may overwhelm calcification inhibitors and have been associated with an increased risk for both MI16 and kidney stones.17The study strengths include general population cohorts with validated MI end points. Although there was likely some nondifferential misclassification of comorbidities by diagnostic codes, these comorbidities did have the expected associations with kidney stones and with MI. The risk for MI may vary by stone composition just as the risk for CKD may vary by stone composition.18 Limitations include lack of information on stone burden, stone composition, diet, medications, and laboratory test results. Furthermore, because study participants were mostly non-Hispanic white individuals, a population at increased risk for kidney stones,19 inferences to other ethnic groups are limited.In conclusion, the increased risk for MI with kidney stones suggests these two diseases share a common pathophysiologic pathway. This could be a target for future intervention strategies. A history of kidney stones may also be a useful addition in risk stratification algorithms for MI. Further studies are needed to assess the relevance of stone composition and stone burden to risk for MI.  相似文献   

20.
HLA-A2-matched peripheral blood mononuclear cells from type 1 diabetic patients, but not nondiabetic donors, transfer insulitis to NOD-scid/γc(null)/HLA-A2 transgenic mice concurrent with the expansion of islet-specific CD8+ T cells     
Whitfield-Larry F  Young EF  Talmage G  Fudge E  Azam A  Patel S  Largay J  Byrd W  Buse J  Calikoglu AS  Shultz LD  Frelinger JA 《Diabetes》2011,60(6):1726-1733

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   

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