Anda di halaman 1dari 8

BRIEF REVIEW

www.jasn.org

New Insights into Nephrogenic Systemic Fibrosis


Sundararaman Swaminathan and Sudhir V. Shah
Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas

ABSTRACT
Nephrogenic systemic fibrosis is a new disorder reported almost exclusively in patients who have renal insufficiency and are exposed to contrast media formulated with gadolinium. High morbidity and mortality are associated with this severely disabling and painful condition. The acute phase begins upon exposure to gadolinium contrast media, characterized by a systemic inflammatory response involving iron mobilization, and then as a progressive, chronic phase in which fibrosis develops. Proposed is a unifying model of cumulative risk factors in which the interplay of systemic inflammation and stimulated hematopoietic environment associated with hyperparathyroidism and erythropoietin may tie to a common pathogenic mechanism of fibrogenesis. Because there are no uniformly effective interventions to treat nephrogenic systemic fibrosis other than successful renal transplantation, prevention by avoiding gadolinium contrast media in patients with chronic kidney disease is vital. On the basis of suspected pathogenesis, it is also reasonable to limit erythropoietin and iron therapy to dosages ensuring recommended targets and adequately control hyperparathyroidism. Herein is reviewed what is currently known about this subject.
J Am Soc Nephrol 18: 2636 2643, 2007. doi: 10.1681/ASN.2007060645

Table 1. Components of NSF


Acute phase: Systemic inflammatory response fever hypotension acute kidney injury elevated D-dimer elevated lipase anemia, thrombocytopenia, or thrombocytosis leukoerythroblastic picture monocytosis high serum ferritin, low total iron-binding capacity high C-reactive protein Chronic phase: Edema and fibrosis edema woody induration scleral plaques

Nephrogenic systemic fibrosis (NSF) is a recently described systemic fibrosing disorder that has been reported almost exclusively in patients with renal insufficiency. It was first described by Cowper et al.1 in 2000 as a cutaneous scleromyxedema-like disorder in patients with ESRD and was initially called nephrogenic fibrosing dermopathy.2 Clinical and autopsy data now suggest the presence of a systemic fibrogenic reaction with evidence of involvement of muscle, tendons, diaphragm, testes, cardiac atrium, lungs, and duramater.3 The disease henceforth has been referred to as nephrogenic systemic fibrosis.4 More than 200 cases of NSF have been reported to the Food and Drug Administration and to the NSF registry at Yale University. The estimated prevalence of
2636
ISSN : 1046-6673/1810-2636

NSF, however, is likely to be inaccurate because it still is not a widely recognized clinical entity. In addition, subclinical forms of NSF that could be recognized only with a skin biopsy might remain undiagnosed. In this review, we describe novel clinical manifestations of NSF and discuss our views on its pathogenesis. Specifically, we present a general paradigm in which different risk factors may be tied to a common pathogenic mechanism.

features of NSF may include an acute phase that immediately follows exposure to gadolinium-based contrast media (GBCM) and an overlapping chronic phase characterized by progressive fibrosis (Table 1). Features in the acute phase are variably present and mimic systemic inflammatory response syndrome: Fever, hypotension, acute kidney injury, anemia, leukoerythroblastic picture, thrombocytopenia or thrombocytosis, leukocytosis, eosinophilia, monocytosis, elevated -glutamyl peptidase, elevated lipase,
Published online ahead of print. Publication date available at www.jasn.org.

CLINICAL AND LABORATORY FEATURES

NSF is a severely disabling systemic fibrosing condition associated with increased morbidity and mortality.57 Our observations suggest that clinical

Correspondence to: Dr. Sundararaman Swaminathan, Division of Nephrology, University of Arkansas for Medical Sciences, 4301 W Markham Street #501, Little Rock, AR 72205. Phone: 501-686-5295; Fax: 501-686-7878; E-mail: sswaminathan@ uams.edu Copyright 2007 by the American Society of Nephrology

J Am Soc Nephrol 18: 2636 2643, 2007

www.jasn.org

BRIEF REVIEW

and elevated D-dimer.8 Decreased total iron-binding capacity, elevated serum ferritin, low serum albumin, and elevated C-reactive protein are invariably present.7 Because many patients have critical illnesses such as sepsis, acute pancreatitis, hepatorenal syndrome, or acute graft dysfunction that precedes NSF, the acute phase of systemic fibrogenesis may go unrecognized. Onset of the chronic phase is also variable: As early as 4 d or as late as several months

after GBCM exposure.9 Early cutaneous manifestations in the chronic phase are usually limited to generalized edema followed closely by the development of a plaque-like skin rash with woody induration (Figure 1A). The condition tends to affect either dependent parts of the body, such as extremities or the presacral area, or high blood-flow areas, such as the skin overlying an arteriovenous dialysis fistula.5,6 Facial involvement is very un-

usual except in severe, advanced cases in which temporal regions of the face may be involved (S. Swaminathan, 2006, personal observation). Moderate to severe pain is more typical. Alopecia, yellowish scleral nodules, hyperpigmentation of extremities, and yellowish discoloration of facial skin are observed in some patients. Additional systemic findings that have been observed in patients with NSF include the development of acute pancreatitis, vas-

Figure 1. (A) NSF involving legs and hand. Note the induration, edema, plaque-like rash, and joint contractures. (B) Histopathologic appearance of NSF characterized by dermal spindle cell proliferation with extension into subcutaneous tissue.
J Am Soc Nephrol 18: 2636 2643, 2007 Nephrogenic Systemic Fibrosis

2637

BRIEF REVIEW

www.jasn.org

cular thrombosis, and frequent infection.5,10 The disease is severely disabling, and many patients become wheelchair dependent or bed bound. Severe depressive illness often ensues, and a significant number of patients quit dialysis. The diagnosis of NSF is established by performing a deep-skin biopsy from affected areas. Biopsy findings (Figure 1B) that confirm NSF include dermal spindle cell (fibroblast) proliferation (usually CD34 ) with frequent extension into subcutaneous tissue, presence of dermal mucin, variable infiltration of CD68 macrophages, and presence of broad collagen bundles with clefts and fragmented elastin. Osseous metaplasia, osteoclastlike giant cells, and calciphylaxis can be seen in some NSF biopsies.1113 On a technetium99-diphosphosphonate scan, areas of increased uptake can be observed in muscles.14,15 On magnetic resonance imaging, axial T1- and fat-suppressed T2-weighted images show symmetric skin thickening and soft tissue edema.15 The differential diagnoses of NSF include scleroderma, scleromyxedema, eosinophilic fasciitis, and graft-versus-host disease. An absence of facial involvement and circulating paraprotein, temporal relation to GBCM exposure, and the appropriate clinical context helps in differentiating NSF from these other conditions.

model proposed by us and detailed in the following section, patients with more cumulative risk (risk factor load) may only need low dosages of GBCM to trigger NSF or vice versa (Figure 2).

our ability to interpret these findings18 or arrive at a precise and safe cutoff of GFR above which the risk for NSF is negligible.

IMPAIRED RENAL FUNCTION

GADOLINIUM TOXICITY: ROLE OF IRON AND TRANSMETALLATION

RISK FACTORS FOR NSF

NSF is seen in patients both with acute kidney injury and with chronic kidney disease (CKD). No specific cause of kidney disease except hepatorenal syndrome15 has been associated with heightened risk. NSF is seen in patients receiving either hemodialysis or peritoneal dialysis as well as in patients with posttransplantation allograft dysfunction. Singularly, a low GFR is a major prerequisite for NSF to develop. In patients with CKD, on the basis of current reports as well as personal observations, the risk for NSF seems to be limited to those with stage 3 or worse CKD (GFR 60 ml/min). Recently, Sadowski et al. reported the risk for NSF after gadolinium exposure at various levels of estimated GFR (eGFR), and in their series, NSF developed predominantly in those with eGFR 30 ml/min.10 NSF also developed in two patients with an apparently higher eGFR in the setting of acute kidney injury, where formula-based estimates for GFR cannot be used. In our personal experience, we have observed, after GBCM exposure, that NSF can develop in patients with an eGFR as high as 40 ml/min. However, imprecision of formula-based eGFR in accurately predicting renal function in stage 3 CKD limits

In addition to several reports on the epidemiologic association of GBCM exposure and NSF, demonstration of gadolinium in NSF tissues has strengthened the causal link between the two.19 A higher dosage and multiple repeated exposures to GBCM increase the risk for NSF.10 The mechanisms through which administration of GBCM results in toxicity and NSF are still evolving. However, several important clues appear from current published literature. First, overt NSF develops in only 3 to 5% of patients after GBCM exposure.9,10,16 Second, some patients with NSF do not manifest any clinical signs, even when they were previously exposed to GBCM. Third, demonstration of significant quantities of insoluble gadolinium in the skin of patients with NSF, months after GBCM exposure and after extensive tissue processing, suggest that gadolinium might have undergone transmetallation in vivo. This finding is of significance because free gadolinium is toxic.20 Supporting the importance of transmetallation, all NSF cases reported thus far have been associated with linear magnetic resonance contrast agents9,16,21,22 that have inferior thermodynamic stability2326 and a kinetic or conditional sta-

The major risk factors associated with NSF include exposure to GBCM,9,16,17 high-dosage erythropoietin (EPO) therapy,7 elevated parathyroid hormone (PTH),7 hypothyroidism,17 and antiphospholipid antibodies.17 Significant vascular disease is also a risk factor.17 On the basis of current understanding, an interaction of low GFR; exposure to GBCM; and presence of additional risk factors such as high-dosage EPO, elevated PTH, vascular disease, and systemic inflammation seem necessary for the development of NSF in the majority of cases. In this cumulative risk factor
2638

Figure 2. Cumulative risk factor model of NSF: Conceptual inverse interaction between gadolinium dosage and risk factors in the pathogenesis of NSF.
J Am Soc Nephrol 18: 2636 2643, 2007

Journal of the American Society of Nephrology

www.jasn.org

BRIEF REVIEW

bility that favors transmetallation. The mechanism by which GBCM may undergo transmetallation in vivo or cause toxicity is still under investigation. In an initial report, acidosis was thought to induce gadoliniums dissociation from its chelate16; however, a subsequent report failed to confirm this.9 Administration of magnetic resonance contrast agents is widely known to cause transient (up to 72 h) elevations in serum iron, even in 15 to 30% of healthy volunteers.27,28 Because iron is tightly bound to ferritin and hemosiderin, it has been suggested that its free concentration is insufficient to induce transmetallation of GBCM25,29; however, we recently reported that GBCM administration in patients who have CKD and subsequently develop NSF results in a marked decrease in total iron-binding capacity, iron mobilization, profound transferrin oversaturation, and systemic inflammation.8 In patients with renal insufficiency, several factors may aggravate free iron release, including prolonged retention of GBCM (t 13.4 to 89.2 h versus 1.5 h controls),30 additional exogenous treatment with parenteral iron and low total iron-binding capacity secondary to malnutrition, urinary protein (transferrin) loss, sepsis, and chronic inflammation.3133 Available data suggest that iron is most potent in inducing transmetallation of GBCM because the thermodynamic stability of Fe3 -DTPA-BMA (1021.9) far exceeds the thermodynamic stability constant of gadolinium-DTPA-BMA (1016.9)34 (Table 2). Gadolinium-mobilized iron can also be directly toxic to tissues through the induction of oxidative stress by Fenton reaction (Figure 3).35 In addition, cata12

Figure 3. Transmetallation of gadolinium chelates by endogenous ions: The role of iron.

lytic iron released by systemic inflammation35 and gadolinium-mediated cellular acquisition of catalytic iron might contribute to the development of NSF.3538 Thus, a combination of free gadolinium, catalytic iron, systemic inflammation,10 and oxidative stress may result in initial injury and a subsequent systemic-fibrosing illness characteristic of NSF. Recent descriptions19,39 of significant iron and gadolinium deposition in NSF tissues support our findings. Collectively, these observations suggest the development of NSF after GBCM exposure needs not only a prolonged retention of GBCM but also a process by which GBCM induces toxicity, a process whereby iron may play an important role.

ROLE OF EPO, INFLAMMATION, AND VASCULAR INJURY

We and others7,17,40 recently reported that patients with NSF also receive higher dosages of EPO, suggesting that EPO might play a role in pathogenesis. On the basis of our clinical experience with 70 patients with NSF and other available large case series,7,8,17 it is uncommon that patients do not receive any EPO before NSF diagnosis. It is possible that endogenous EPO released during stress-in-

duced erythropoiesis, which could be several hundred-fold,41 or other growth factors that synergize with EPO42 might be important in patients who do not receive exogenous EPO before a diagnosis of NSF. In addition, in applying the cumulative risk model (Figure 1), exogenous EPO may not be a necessary prerequisite in all cases. There are many compelling reasons for why EPO might participate in the pathogenesis of NSF. Its pleiotropic biologic effects and/or systemic inflammation associated with an EPO-resistant state may be important (Table 3). Theoretically, EPO therapy influences all key elements of the pathogenesis of NSF: Endothelial dysfunction, inflammation, cell proliferation, and wound healing. EPO is a potent cytokine with stimulatory effects on vascular endothelium,43 smooth muscle cells,44,45 and platelets.46 EPO administration induces a release of vasoactive factors such as monocyte chemoattractant protein-1,45,47 endothelin-1,48 thromboxane A2, and selectin.46,49 It can also induce endothelial dysfunction by inhibiting dimethylarginine dimethylaminohydrolase, thereby increasing asymmetric dimethyl arginine.50 Furthermore, EPO is a potent stimulant of endothelial and progenitor cell proliferation,51,52 as well as wound-healing responses53 that are reminiscent of histologic
Table 3. Potential role of EPO in the pathogenesis of NSF
Endothelial dysfunction increases endothelin-1 asymmetric dimethyl arginine Proinflammatory increases monocyte chemoattractant protein-1 Cell proliferation increases endothelial cells smooth muscle cells endothelial progenitor cells

Table 2. Potency of various endogenous cations in inducing transmetallation of gadolinium-DTPA-BMAa


Cation-DTPA-BMA Calcium Zinc Copper Gadolinium Iron Log Ktherm 107.2 1012 1013 1016.9 1021.9 Transmetallation Potency

a Ktherm is a thermodynamic stability constant. The plus and minus symbols refer to the degree of potency. , mild; , moderate; , strong; , n/a.

J Am Soc Nephrol 18: 2636 2643, 2007

Nephrogenic Systemic Fibrosis

2639

BRIEF REVIEW

www.jasn.org

changes seen in NSF. EPO51,52,54 and PTH55 are also strong stimuli for a systemic release of CD34 progenitor cells, which are known to participate in wound healing.56 58 Vascular endothelial cell injury could similarly contribute to the pathogenesis of NSF by inducing the release of vasoactive factors and CD34 progenitors,41,59 and in this setting, EPOs detrimental effect on endothelia is further aggravated.60,61 In addition, chronic inflammation62 associated with an EPO-resistant state and highdosage EPO45,47 might also contribute to the fibrogenesis seen in NSF.

CELLULAR BASIS FOR THE PERSISTENCE OF FIBROSIS IN NSF

Figure 4. Cellular basis for fibrosis in NSF: Interaction of multiple risk factors and central role of hematopoietic stem cells.

As with any other systemic fibrosing condition,63 possible origins of fibroblasts in NSF include resident mesenchymal cell populations, epithelial-to-mesenchymal transition of local epithelia, and bone marrow derived cells. Cowper and Bucala64 demonstrated that the spindle cells in NSF skin lesions express CD34 and procollagen 1 and suggested that circulating fibrocytes mediate the fibrosis in NSF. Even though the presence of these markers suggests a contribution from circulating fibrocytes, it is important to note that these markers are not unique to fibrocytes65; fibrocytes contribution to fibrosis is highly variable in different organs66 and is affected by additional factors such as cell turnover rates. Supporting this, Fathke et al.,67 using crosstransplantation (bone marrow) experiments in an EGFP transgenic mouse model, nicely demonstrated that both CD45 and CD45 bone marrow derived stem cells contribute to dermal collagen deposition and wound repair. In addition, they observed a role for local dermal cells and endothelial progenitor cells in dermal wound healing. Similarly, using tissue reconstitution experiments with single hematopoietic stem cells (HSC), Ogawa et al.56 showed that HSC significantly contribute to myofibroblast population in tissues. EPO, PTH, and an oxidative stress-induced stimulated hematopoietic environment41,51,55,68,69
2640

may thus contribute to fibrosis through increased production of tissue myofibroblasts. It is unknown, however, whether fibrocytes are a necessary intermediate in this process of transformation of HSC into myofibroblasts in vivo; therefore, it is likely that both resident fibroblasts and diverse types of bone marrow derived cells that are released in response to injury and cytokines such as EPO, including HSC (likely to be the most significant contributor), endothelial progenitor cells, mesenchymal precursors,65 and monocyte-derived cell fibrocytes,64 contribute to fibrogenesis in NSF (Figure 4). Any contribution of epithelial-to-mesenchymal transition to fibrogenesis in NSF, although potentially possible, has not been evaluated. Like other chronic fibrosing disorders, persistent or progressive fibrosis, even after the original trigger is removed, may lead to NSF. This occurrence possibly relates to the concept of a persistently activated fibroblast phenotype, which could be mediated by factors such as ongoing chronic endothelial injury in uremia.70,71

TREATMENT

There are no good therapies for the cutaneous symptoms of NSF or, more important, to prevent its associated high

mortality; therefore, our first recommendation is prevention. On the basis of the available data and our personal experience, we recommend the following measures for NSF prevention: Renal function should be checked in all high-risk patients before GBCM administration (age 50 yr; infants; history of renal disease, diabetes, hypertension, or liver disease; and critically ill patients). Avoid GBCM in patients with an eGFR 40 ml/min. If clinically indicated and benefits outweigh risks, then use low dosages of GBCM (preferably macrocyclic agents) in patients with an eGFR of 40 to 60 ml/ min. GBCM is absolutely contraindicated in patients who are on peritoneal dialysis, because gadolinium is poorly cleared by the peritoneum. In patients who are on hemodialysis, GBCM should be administered only if the benefits of obtaining a contrast-enhanced magnetic resonance image substantially outweigh the risk. At the completion of imaging, patients should go directly to the hemodialysis unit for initiation of dialysis, and dialysis treatment should be administered for 3 consecutive days. Avoid intravenous iron immediately before or after GBCM exposure. Treatment of NSF involves a multidisciJ Am Soc Nephrol 18: 2636 2643, 2007

Journal of the American Society of Nephrology

www.jasn.org

BRIEF REVIEW

plinary approach. Aggressive physical therapy, pain control, and psychologic support are essential cornerstones of therapy in this severely disabling disease. The severity of pain usually necessitates opiate analgesics. Topical treatments include the use of moisturizers, emollients, and anti-inflammatory creams. Deep tissue massage and hydrotherapy may be helpful. On the basis of potential pathogenic mechanisms, control of PTH levels and limiting administration of EPO and intravenous iron enough to maintain recommended hemoglobin targets are likely to help. Limiting systemic inflammation by decreasing infection episodes such as catheter-related bacteremia may also be beneficial. Other treatment approaches that could result in variable and modest improvements include a combination of hydroxychloroquine and lowdosage steroids (S. Swaminathan, 2006, personal observation), extracorporeal photopheresis, and topical ultraviolet-A therapy. However, none of these options has been uniformly effective in patients with NSF. Renal transplantation that results in a well-functioning allograft usually leads to a substantial resolution of the disease, and maintaining lower cyclosporine and tacrolimus targets may also potentially help in limiting fibrogenesis.7274 Secondary worsening of NSF after initial improvement but without new GBCM exposure can occur, but the mechanisms involved in these relapses are unclear. No data are available on the benefit of using chelation therapy to remove gadolinium in NSF. A better understanding of the pathogenesis in this devastating disease may lead to new interventions, which may be relevant not only to NSF but other fibrosing conditions.

REFERENCES
1. Cowper SE, Robin HS, Steinberg SM, Su LD, Gupta S, LeBoit PE: Scleromyxoedema-like cutaneous diseases in renal-dialysis patients. Lancet 356: 1000 1001, 2000 2. Cowper SE, Su LD, Bhawan J, Robin HS, LeBoit PE: Nephrogenic fibrosing dermopathy. Am J Dermatopathol 23: 383393, 2001 3. Ting WW, Stone MS, Madison KC, Kurtz K: Nephrogenic fibrosing dermopathy with systemic involvement. Arch Dermatol 139: 903906, 2003 4. Cowper SE: Nephrogenic systemic fibrosis: The nosological and conceptual evolution of nephrogenic fibrosing dermopathy. Am J Kidney Dis 46: 763765, 2005 5. Cowper SE: Nephrogenic fibrosing dermopathy: The first 6 years. Curr Opin Rheumatol 15: 785790, 2003 6. Swaminathan S, Leung N: Nephrogenic fibrosing dermopathy: Lessons from the past. Int J Dermatol 45: 639 641, 2006 7. Swaminathan S, Ahmed I, McCarthy JT, Albright RC, Pittelkow MR, Caplice NM, Griffin MD, Leung N: Nephrogenic fibrosing dermopathy and high-dose erythropoietin therapy. Ann Intern Med 145: 234 235, 2006 8. Swaminathan S, Horn TD, Pellowski D, AbulEzz S, Bornhorst JA, Viswamitra S, Shah SV: Nephrogenic systemic fibrosis, gadolinium, and iron mobilization. N Engl J Med 357: 720 722, 2007 9. Marckmann P, Skov L, Rossen K, Dupont A, Damholt MB, Heaf JG, Thomsen HS: Nephrogenic systemic fibrosis: Suspected causative role of gadodiamide used for contrast-enhanced magnetic resonance imaging. J Am Soc Nephrol 17: 2359 2362, 2006 10. Sadowski EA, Bennett LK, Chan MR, Wentland AL, Garrett AL, Garrett RW, Djamali A: Nephrogenic systemic fibrosis: Risk factors and incidence estimation. Radiology 243: 148 157, 2007 11. Ruiz-Genao DP, Pascual-Lopez MP, Fraga S, Aragues M, Garcia-Diez A: Osseous metaplasia in the setting of nephrogenic fibrosing dermopathy. J Cutan Pathol 32: 172 175, 2005 12. Hershko K, Hull C, Ettefagh L, Nedorost S, Dyson S, Horn T, Gilliam AC: A variant of nephrogenic fibrosing dermopathy with osteoclast-like giant cells: A syndrome of dysregulated matrix remodeling? J Cutan Pathol 31: 262265, 2004 13. Edsall LC, English JC 3rd, Teague MW, Patterson JW: Calciphylaxis and metastatic calcification associated with nephrogenic fibrosing dermopathy. J Cutan Pathol 31: 247253, 2004 14. Gremmels JM, Kirk GA: Two patients with abnormal skeletal muscle uptake of Tc-99m hydroxymethylene diphosphonate following 15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

ACKNOWLEDGMENT
We thank Cindy Reid for technical editing and graphics assistance.

27.

DISCLOSURES
None.
J Am Soc Nephrol 18: 2636 2643, 2007

28.

liver transplant: Nephrogenic fibrosing dermopathy and graft vs host disease. Clin Nucl Med 29: 694 697, 2004 Broome DR, Girguis MS, Baron PW, Cottrell AC, Kjellin I, Kirk GA: Gadodiamide-associated nephrogenic systemic fibrosis: Why radiologists should be concerned. AJR Am J Roentgenol 188: 586 592, 2007 Grobner T: Gadolinium: A specific trigger for the development of nephrogenic fibrosing dermopathy and nephrogenic systemic fibrosis? Nephrol Dial Transplant 21: 1104 1108, 2006 Nephrogenic fibrosing dermopathy associated with exposure to gadolinium-containing contrast agentsSt. Louis, Missouri, 20022006. MMWR Morb Mortal Wkly Rep 56: 137141, 2007 Poggio ED, Wang X, Greene T, Van Lente F, Hall PM: Performance of the modification of diet in renal disease and Cockcroft-Gault equations in the estimation of GFR in health and in chronic kidney disease. J Am Soc Nephrol 16: 459 466, 2005 High WA, Ayers RA, Cowper SE: Gadolinium is quantifiable within the tissue of patients with nephrogenic systemic fibrosis. J Am Acad Dermatol 56: 710 712, 2007 Spencer AJ, Wilson SA, Batchelor J, Reid A, Rees J, Harpur E: Gadolinium chloride toxicity in the rat. Toxicol Pathol 25: 245255, 1997 Thomsen HS, Morcos SK, Dawson P: Is there a causal relation between the administration of gadolinium based contrast media and the development of nephrogenic systemic fibrosis (NSF)? Clin Radiol 61: 905906, 2006 Thomsen HS: Nephrogenic systemic fibrosis: A serious late adverse reaction to gadodiamide. Eur Radiol 16: 2619 2621, 2006 Puttagunta NR, Gibby WA, Smith GT: Human in vivo comparative study of zinc and copper transmetallation after administration of magnetic resonance imaging contrast agents. Invest Radiol 31: 739 742, 1996 Puttagunta NR, Gibby WA, Puttagunta VL: Comparative transmetallation kinetics and thermodynamic stability of gadoliniumDTPA bis-glucosamide and other magnetic resonance imaging contrast media. Invest Radiol 31: 619 624, 1996 Mann JS: Stability of gadolinium complexes in vitro and in vivo. J Comput Assist Tomogr 17[Suppl 1]: S19 S23, 1993 Kirchin MA, Runge VM: Contrast agents for magnetic resonance imaging: Safety update. Top Magn Reson Imaging 14: 426 435, 2003 VanWagoner M, OToole M, Worah D, Leese PT, Quay SC: A phase I clinical trial with gadodiamide injection, a nonionic magnetic resonance imaging enhancement agent. Invest Radiol 26: 980 986, 1991 Shellock FG, Kanal E: Safety of magnetic resonance imaging contrast agents. J Magn Reson Imaging 10: 477 484, 1999

Nephrogenic Systemic Fibrosis

2641

BRIEF REVIEW

www.jasn.org

29. Cacheris WP, Quay SC, Rocklage SM: The relationship between thermodynamics and the toxicity of gadolinium complexes. Magn Reson Imaging 8: 467 481, 1990 30. Joffe P, Thomsen HS, Meusel M: Pharmacokinetics of gadodiamide injection in patients with severe renal insufficiency and patients undergoing hemodialysis or continuous ambulatory peritoneal dialysis. Acad Radiol 5: 491502, 1998 31. Pupim LB, Caglar K, Hakim RM, Shyr Y, Ikizler TA: Uremic malnutrition is a predictor of death independent of inflammatory status. Kidney Int 66: 2054 2060, 2004 32. Kaysen GA, Eiserich JP: Characteristics and effects of inflammation in end-stage renal disease. Semin Dial 16: 438 446, 2003 33. Pupim LB, Evanson JA, Hakim RM, Ikizler TA: The extent of uremic malnutrition at the time of initiation of maintenance hemodialysis is associated with subsequent hospitalization. J Ren Nutr 13: 259 266, 2003 34. Idee JM, Port M, Raynal I, Schaefer M, Le Greneur S, Corot C: Clinical and biological consequences of transmetallation induced by contrast agents for magnetic resonance imaging: A review. Fundam Clin Pharmacol 20: 563576, 2006 35. Shah SV, Alam MG: Role of iron in atherosclerosis. Am J Kidney Dis 41: S80 S83, 2003 36. Olakanmi O, Stokes JB, Pathan S, Britigan BE: Polyvalent cationic metals induce the rate of transferrin-independent iron acquisition by HL-60 cells. J Biol Chem 272: 2599 2606, 1997 37. Olakanmi O, Stokes JB, Britigan BE: Gallium-inducible transferrin-independent iron acquisition is a property of many cell types: Possible role of alterations in the plasma membrane. J Investig Med 53: 143153, 2005 38. Olakanmi O, Rasmussen GT, Lewis TS, Stokes JB, Kemp JD, Britigan BE: Multivalent metal-induced iron acquisition from transferrin and lactoferrin by myeloid cells. J Immunol 169: 2076 2084, 2002 39. High WA, Ayers RA, Chandler J, Zito G, Cowper SE: Gadolinium is detectable within the tissue of patients with nephrogenic systemic fibrosis. J Am Acad Dermatol 56: 21 26, 2007 40. Marckmann P, Skov L, Rossen K, Heaf JG, Thomsen HS: Case-control study of gadodiamide-related nephrogenic systemic fibrosis. Nephrol Dial Transplant 17: 2359 2362, 2007 41. Socolovsky M: Molecular insights into stress erythropoiesis. Curr Opin Hematol 14: 215 224, 2007 42. Sigounas G, Steiner M, Anagnostou A: Synergism of hemopoietic growth factors on endothelial cell proliferation. Angiology 48: 141147, 1997 43. Anagnostou A, Lee ES, Kessimian N, Levinson R, Steiner M: Erythropoietin has a mito-

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

genic and positive chemotactic effect on endothelial cells. Proc Natl Acad Sci U S A 87: 5978 5982, 1990 Ammarguellat F, Llovera M, Kelly PA, Goffin V: Low doses of EPO activate MAP kinases but not JAK2-STAT5 in rat vascular smooth muscle cells. Biochem Biophys Res Commun 284: 10311038, 2001 Desai A, Zhao Y, Lankford HA, Warren JS: Nitric oxide suppresses EPO-induced monocyte chemoattractant protein-1 in endothelial cells: Implications for atherogenesis in chronic renal disease. Lab Invest 86: 369 379, 2006 Stohlawetz PJ, Dzirlo L, Hergovich N, Lackner E, Mensik C, Eichler HG, Kabrna E, Geissler K, Jilma B: Effects of erythropoietin on platelet reactivity and thrombopoiesis in humans. Blood 95: 29832989, 2000 De Marchi S, Cecchin E, Falleti E, Giacomello R, Stel G, Sepiacci G, Bortolotti N, Zanello F, Gonano F, Bartoli E: Long-term effects of erythropoietin therapy on fistula stenosis and plasma concentrations of PDGF and MCP-1 in hemodialysis patients. J Am Soc Nephrol 8: 11471156, 1997 Carlini RG, Dusso AS, Obialo CI, Alvarez UM, Rothstein M: Recombinant human erythropoietin (rHuEPO) increases endothelin-1 release by endothelial cells. Kidney Int 43: 1010 1014, 1993 Kahraman S, Yilmaz R, Kirkpantur A, Genctoy G, Arici M, Altun B, Erdem Y, Yasavul U, Turgan C: Impact of rHuEPO therapy initiation on soluble adhesion molecule levels in haemodialysis patients. Nephrology (Carlton) 10: 264 269, 2005 Scalera F, Kielstein JT, Martens-Lobenhoffer J, Postel SC, Tager M, Bode-Boger SM: Erythropoietin increases asymmetric dimethylarginine in endothelial cells: Role of dimethylarginine dimethylaminohydrolase. J Am Soc Nephrol 16: 892 898, 2005 Bahlmann FH, DeGroot K, Duckert T, Niemczyk E, Bahlmann E, Boehm SM, Haller H, Fliser D: Endothelial progenitor cell proliferation and differentiation is regulated by erythropoietin. Kidney Int 64: 1648 1652, 2003 George J, Goldstein E, Abashidze A, Wexler D, Hamed S, Shmilovich H, Deutsch V, Miller H, Keren G, Roth A: Erythropoietin promotes endothelial progenitor cell proliferative and adhesive properties in a PI 3-kinase-dependent manner. Cardiovasc Res 68: 299 306, 2005 Galeano M, Altavilla D, Bitto A, Minutoli L, Calo M, Lo Cascio P, Polito F, Giugliano G, Squadrito G, Mioni C, Giuliani D, Venuti FS, Squadrito F: Recombinant human erythropoietin improves angiogenesis and wound healing in experimental burn wounds. Crit Care Med 34: 1139 1146, 2006 Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S: Eryth-

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66.

67.

68.

ropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 102: 1340 1346, 2003 Ballen K: Targeting the stem cell niche: Squeezing blood from bones. Bone Marrow Transplant 39: 655 660, 2007 Ogawa M, LaRue AC, Drake CJ: Hematopoietic origin of fibroblasts/myofibroblasts: Its pathophysiologic implications. Blood 108: 28932896, 2006 Haroon ZA, Amin K, Jiang X, Arcasoy MO: A novel role for erythropoietin during fibrininduced wound-healing response. Am J Pathol 163: 9931000, 2003 Sayan H, Ozacmak VH, Guven A, Aktas RG, Ozacmak ID: Erythropoietin stimulates wound healing and angiogenesis in mice. J Invest Surg 19: 163173, 2006 Wojakowski W, Tendera M, Michalowska A, Majka M, Kucia M, Maslankiewicz K, Wyderka R, Ochala A, Ratajczak MZ: Mobilization of CD34/CXCR4 , CD34/CD117 , c-met stem cells, and mononuclear cells expressing early cardiac, muscle, and endothelial markers into peripheral blood in patients with acute myocardial infarction. Circulation 110: 32133220, 2004 Ando M, Iwata A, Ozeki Y, Tsuchiya K, Akiba T, Nihei H: Circulating platelet-derived microparticles with procoagulant activity may be a potential cause of thrombosis in uremic patients. Kidney Int 62: 17571763, 2002 Coleman TR, Westenfelder C, Togel FE, Yang Y, Hu Z, Swenson L, Leuvenink HG, Ploeg RJ, dUscio, LV, Katusic ZS, Ghezzi P, Zanetti A, Kaushansky K, Fox NE, Cerami A, Brines M: Cytoprotective doses of erythropoietin or carbamylated erythropoietin have markedly different procoagulant and vasoactive activities. Proc Natl Acad Sci U S A 103: 59655970, 2006 Meneghin A, Hogaboam CM: Infectious disease, the innate immune response, and fibrosis. J Clin Invest 117: 530 538, 2007 Wynn TA: Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Invest 117: 524 529, 2007 Cowper SE, Bucala R: Nephrogenic fibrosing dermopathy: Suspect identified, motive unclear. Am J Dermatopathol 25: 358, 2003 Kuwana M, Okazaki Y, Kodama H, Izumi K, Yasuoka H, Ogawa Y, Kawakami Y, Ikeda Y: Human circulating CD14 monocytes as a source of progenitors that exhibit mesenchymal cell differentiation. J Leukoc Biol 74: 833 845, 2003 Iredale JP: Models of liver fibrosis: Exploring the dynamic nature of inflammation and repair in a solid organ. J Clin Invest 117: 539 548, 2007 Fathke C, Wilson L, Hutter J, Kapoor V, Smith A, Hocking A, Isik F: Contribution of bone marrow-derived cells to skin: Collagen deposition and wound repair. Stem Cells 22: 812 822, 2004 Calvi LM, Adams GB, Weibrecht KW, Weber

2642

Journal of the American Society of Nephrology

J Am Soc Nephrol 18: 2636 2643, 2007

www.jasn.org

BRIEF REVIEW

JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT: Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425: 841 846, 2003 69. Liu J, Finkel T: Stem cell aging: What bleach can teach. Nat Med 12: 383384, 2006 70. Dou L, Bertrand E, Cerini C, Faure V, Sampol J, Vanholder R, Berland Y, Brunet P: The uremic solutes p-cresol and indoxyl sulfate inhibit endothelial proliferation and wound repair. Kidney Int 65: 442451, 2004

71. Laplante P, Raymond MA, Gagnon G, Vigneault N, Sasseville AM, Langelier Y, Bernard M, Raymond Y, Hebert MJ: Novel fibrogenic pathways are activated in response to endothelial apoptosis: Implications in the pathophysiology of systemic sclerosis. J Immunol 174: 5740 5749, 2005 72. McMorrow T, Gaffney MM, Slattery C, Campbell E, Ryan MP: Cyclosporine A induced epithelial-mesenchymal transition in human renal proximal tubular epithelial cells. Nephrol Dial Transplant 20:

22152225, 2005 73. Ceol M, Anglani F, Vianello D, Murer L, Del Prete D, Forino M, Favaro S, Gambaro G, DAngelo A: Direct effect of chronic cyclosporine treatment on collagen III mRNA expression and deposition in rat kidneys. Clin Nephrol 53[Suppl 8 9], 2000 74. Johnson DW, Saunders HJ, Johnson FJ, Huq SO, Field MJ, Pollock CA: Fibrogenic effects of cyclosporin A on the tubulointerstitium: Role of cytokines and growth factors. Exp Nephrol 7: 470 478, 1999

J Am Soc Nephrol 18: 2636 2643, 2007

Nephrogenic Systemic Fibrosis

2643

Anda mungkin juga menyukai