Crescentic Glomerulonephritis

Back

Background

Rapidly progressive glomerulonephritis (RPGN) is defined as any glomerular disease characterized by extensive crescents (usually >50%) as the principal histologic finding and by a rapid loss of renal function (usually a 50% decline in the glomerular filtration rate [GFR] within 3 mo) as the clinical correlate.

The image shown below depicts the histologic findings observed in patients with severe types of RPGN.



View Image

Glomerulonephritis, crescentic. Light microscopy (200x hematoxylin and eosin stain): The normal renal architecture is lost. The glomerulus (*) is soli....

Transient azotemia with oliguria is common in patients with acute glomerulonephritis.

Some patients have acute glomerulonephritis and present with rapidly progressive renal failure that develops within weeks to months and displays little tendency for spontaneous or complete recovery. Glomerular crescents can complicate any glomerulopathy, even noninflammatory glomerulopathy. In patients with noninflammatory glomerulopathies, the crescents tend to be fibrotic rather than cellular. Cellular crescents are a manifestation of a severe inflammatory process.

Classification

Idiopathic or primary crescentic glomerulonephritis is classified into the following types:

Pathophysiology

RPGN can develop in any of the following clinical settings:

Acute RPGN is mediated by antibody or cellular immunity or by interaction of the two arms of the immune system. Deposition of antibody along the basement membrane and/or glomerular deposition of preformed soluble immune complexes can result in glomerulonephritis. Lymphocytes and macrophages, along with deposited antibody, are important in the production of proliferation and proteinuria. The involved lymphocytes are identified as T cells; most are helper T cells with some suppressor T cells. Antibody- and cell-mediated immunity are together responsible for many lesions observed in patients with acute RPGN, and cell-mediated immunity without antibody may produce crescentic glomerulonephritis.

Crescents are defined as the presence of 2 or more layers of cells in the Bowman space. The presence of crescents in glomeruli is a marker of severe injury, and this may be seen with any form of severe glomerular injury.

The initiating event is the development of a physical disruption in the GBM. The lesions are mediated by processes involving macrophages and cell-mediated immunity. Following disruption of the glomerular capillary, circulating cells, inflammatory mediators, and plasma proteins pass through the capillary wall into the Bowman space. Cells and mediators from the interstitium enter the Bowman space with disruption of the Bowman capsule, which leads to development of crescents.

The major participants in crescent formation are coagulation proteins, macrophages, T cells, fibroblasts, and parietal epithelial cells. Activated macrophages contribute to the crescents by proliferating and releasing procoagulant tissue factor, interleukin-1 (IL-1) and tumor necrosis factor (TNF). T cells are not prominent components, but they play an important role in glomerular injury by antigen recognition and macrophage recruitment.

The crescents may be circumferential or noncircumferential, and the presence of circumferential crescents in over 80% of glomeruli portends a poor prognosis and often manifests as advanced kidney disease.

The reversibility of crescents correlates with relative predominance of cellular components. Whether crescents progress or resolve may depend upon the integrity of the Bowman capsule and resulting cellular composition of the crescent. Progression to fibrous crescents is more common when capsular rupture occurs and fibroblasts along with macrophages are prominent in the Bowman space. The presence of fibrous crescents usually correlates with glomerular sclerosis or irreversibility.

Epidemiology

Frequency

United States

Idiopathic crescentic glomerulonephritis accounts for fewer than 10% of all patients presenting with primary glomerulopathy. RPGN type III is more common than RPGN types I or II. More than 50% of patients with crescentic glomerulonephritis present with acute nephritic syndrome and rapidly deteriorating renal function; however, other modes do occur (eg, asymptomatic, 15%; nephrotic, 10%; chronic renal failure, 15%).

International

Peak incidence of anti-GBM disease occurs in spring and early summer. No seasonal predilection is observed in patients with non–anti-GBM disease.

Mortality/Morbidity

Renal failure at presentation carries an increased risk for end-stage renal disease and death despite immunosuppressive therapy.[1] Death or dialysis occurs in 73% of patients who are treated with conventional therapy and in 88% of patients if they are oligoanuric at time of presentation.

Race

No racial predilection exists.

Sex

For RPGN types I and III, a predilection for males exists.

Age

RPGN has a broad age distribution, as follows:

History

Clinical and laboratory presentations of all types of acute RPGN are quite similar.[2]

Physical

See the list below:

Causes

See the list below:

Laboratory Studies

See the list below:

Imaging Studies

See the list below:

Procedures

See the list below:

Histologic Findings

Idiopathic RPGN is classified as follows:

The normal renal architecture is lost, and the glomerulus is solid, hypercellular, and surrounded by severe interstitial mononuclear cell infiltrate (see the first image below) is observed in patients with severe types. The glomerular tuft is distorted by proliferation of epithelial cells crescents that occupy most of the Bowman space as shown in the second, third, and fourth images below.



View Image

Glomerulonephritis, crescentic. Light microscopy (200x hematoxylin and eosin stain): The normal renal architecture is lost. The glomerulus (*) is soli....



View Image

Glomerulonephritis, crescentic. Light microscopy (25x hematoxylin and eosin stain): Compression of the glomerular tuft with a circumferential cellular....



View Image

Glomerulonephritis, crescentic. Light microscopy (200x periodic acid-Schiff stain): Bowman capsule (arrow) surrounds each glomerulus. The glomerular t....



View Image

Glomerulonephritis, crescentic. Light microscopy (400x trichrome stain): The remnant of the glomerular tuft (*) is surrounded by the cellular crescent....

Anti-GBM disease is characterized by linear IgG deposits as depicted below. Endocapillary proliferation, if prominent, suggests the presence of infection. Segmental or diffuse endocapillary necrosis suggests underlying vasculitis.



View Image

Glomerulonephritis, crescentic. Immunofluorescence (25x): Anti–glomerular basement membrane characterized by the presence of linear immunoglobulin G d....

Medical Care

Early and aggressive treatment is warranted to preserve renal function. Principles of general therapy are listed in Medication.

For specific therapies, see the following related articles:

Consultations

A nephrologist should be involved early in the disease course. Rheumatologist consultation may be considered and may require hematologist involvement if plasma exchange is required.

Diet

See the list below:

Activity

No specific limitations are necessary other than limiting activity as required after renal biopsy.

Medication Summary

Principles of therapy include supportive and specific therapies. Supportive therapy involves control of infection (especially Pneumocystis jiroveci (PCP) infection with trimethoprim-sulfamethoxazole or Atovaquone), control of volume status (providing dialysis if required), and smoking cessation.

Goals of specific therapy

Induction of remission

The initial therapy is to induce remission and typically consists of glucocorticoids and cyclophosphamide, and this induces remission in 85-90% of patients in 2-6 months, with about 75% achieving complete remission. Recently, rituximab in RAVE[4, 5] and RITUXIVAS[6] trials was shown to be not inferior to cyclophosphamide and may be used in patients who cannot take or refuse to take cyclophosphamide. Methotrexate is an option in patients with mild disease, but the relapse rate was significantly higher with methotrexate compared with cyclophosphamide (70% vs 47%, respectively).[7] Etanercept in the WGET (Wegener's Granulomatosis Etanercept Trial) achieved remission in just over 90% of patients but had about a 50% relapse rate at 27 months.[8]

At present, the mainstay of therapy remains cyclophosphamide and steroids for induction of remission, with an option to consider rituximab in select patients.

Plasma exchange, although still controversial in patients with granulomatosis with polyangiitis, microscopic polyangiitis, or renal limited necrotizing glomerulonephritis, is useful in patients with advanced renal failure (serum creatinine >500 µmol/L or requiring dialysis),[1] in patients with severe pulmonary hemorrhage, and when associated with antiglomerular basement membrane antibody disease. Although short-term results with plasma exchange are encouraging, the long-term benefits remain unclear,[9] and the PEXIVAS (Plasma Exchange in VASculitis) trial is currently ongoing to better define the role of plasma exchange.[10]

Maintenance of remission to prevent relapse

As relapses are common, especially in granulomatosis with polyangiitis and microscopic polyangiitis, it is important to continue with immunosuppressive therapy, using less toxic agents, to maintain remission and prevent relapse. Most often, either azathioprine or methotrexate is used for maintenance therapy to reduce risk of relapse. Other agents that have been used for maintenance therapy include etanercept in WGET[8] and mycophenolate.[11]

In a recent pilot study, rituximab maintenance therapy was well tolerated but did not completely prevent relapses and persistent "grumbling" disease[12] ; however, in the interim analysis of an ongoing trial, rituximab at 500 mg every 6-months by the French Vasculitis Study Group in the MAINRISTAN trial show a remarkable reduction in relapse rate with rituximab versus azathioprine, at 28 months (5% vs 25%)[43] .

Methylprednisolone (Solu-Medrol)

Clinical Context:  Potent anti-inflammatory steroid with greater anti-inflammatory potency and fewer tendencies to induce retention of salt and water than prednisolone.

Prednisone (Deltasone, Orasone, Sterapred)

Clinical Context:  Decreases inflammation through multiple mechanisms. Reduced to its pharmacologically active form prednisolone. When used on a long-term basis, alternate-day therapy may elicit fewer adverse effects than daily therapy.

Class Summary

Pulses of intravenous methylprednisolone (5-20 mg/kg) followed by high-dose oral prednisone (2 mg/kg) daily or on alternate days for 2-3 months have shown improved 1-year renal survival rates of 40-70%.

Cyclophosphamide (Cytoxan)

Clinical Context:  Activated in the liver to its active metabolite, 4-hydroxycyclophosphamide, which alkylates the target sites in susceptible cells in an all-or-none–type reaction.

Azathioprine (Imuran)

Clinical Context:  Mechanism by which azathioprine affects autoimmune diseases is unknown. Slow acting, and its effects may persist after discontinuation.

Rituximab (Rituxan)

Clinical Context:  Rituximab is a genetically engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen found on surface of normal and malignant B lymphocytes. The antibody is an IgG1 kappa immunoglobulin containing murine light- and heavy- chain variable region sequences and human constant region sequences.

Class Summary

Addition of cytotoxic agents to corticosteroids has yielded varying success in treating patients with crescentic glomerulonephritis. Although pulse cyclophosphamide is often preferred in lupus nephritis, oral cyclophosphamide appears to have an advantage in Wegener granulomatosis.

Oral versus intravenous: Recently completed, prospectively randomized Cyclophosphamide daily oral versus PulSed (CYCLOPS) trial has shown very little difference in time to remission and time to relapse between daily oral or intermittent intravenous cyclophosphamide for induction therapy.

Cyclophosphamide 3 mg/kg/d for 12 weeks is a common recommendation, but the duration of therapy may be longer (4-6 mo) in patients with pauci-immune glomerulonephritis. This therapy should be followed by the administration of azathioprine (1.5-2 mg/kg/d) or methotrexate (5-20 mg qwk as a single dose) until the patient is in remission for at least 6-12 months. The duration of azathioprine therapy to prevent further relapses is unknown, but it should be at least for 2 years.

Continuing cyclophosphamide for longer than 6 months is not necessary, as recently shown by Cyclophosphamide versus Azathioprine during Remission (CYCAZAREM) trial, where the time to relapse was identical whether the patient was given cyclophosphamide for less than 6 months or more than 6 months.

Mycophenolate mofetil at 0.75-1 g twice daily versus azathioprine was compared for maintenance of remission in an open-label, randomized, controlled trial.[11] Both agents were withdrawn after 42 months and at a median follow-up of 39 months, but relapses were significantly higher in mycophenolate-treated group than in the azathioprine group (55% vs 38%).

Trimethoprim-sulfamethoxazole (Septra, Septra DS)

Clinical Context:  Long-term treatment with TMP (160 mg) and SMX (800 mg) bid has been reported in a prospective, controlled, double-blind trial to sustain remission of Wegener granulomatosis. The mechanism of this effect is not clear. Eradication of Staphylococcus aureus in the anterior nares of patients with Wegener granulomatosis has recently been reported to sustain remission of Wegener granulomatosis. TMP-SMX is also helpful as Pneumocystis carinii pneumonia (PCP) prophylaxis in patients who are on corticosteroids and other immunosuppressive agents.

Class Summary

Therapy must be comprehensive and should cover all likely pathogens in the context of this clinical setting.

Further Outpatient Care

See the list below:

Further Inpatient Care

See the list below:

Inpatient & Outpatient Medications

See the list below:

Transfer

See the list below:

Deterrence/Prevention

See the list below:

Complications

See the list below:

Prognosis

See the list below:

Patient Education

See the list below:

Author

Malvinder S Parmar, MBBS, MS, FRCPC, FACP, FASN, Professor of Medicine, Northern Ontario School of Medicine; Assistant Professor, Department of Medicine, University of Ottawa Faculty of Medicine; Consulting Physician, Timmins and District Hospital, Ontario, Canada

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Ajay K Singh, MB, MRCP, MBA, Associate Professor of Medicine, Harvard Medical School; Director of Dialysis, Renal Division, Brigham and Women's Hospital; Director, Brigham/Falkner Dialysis Unit, Faulkner Hospital

Disclosure: Nothing to disclose.

Chief Editor

Vecihi Batuman, MD, FASN, Huberwald Professor of Medicine, Section of Nephrology-Hypertension, Tulane University School of Medicine; Chief, Renal Section, Southeast Louisiana Veterans Health Care System

Disclosure: Nothing to disclose.

Additional Contributors

James H Sondheimer, MD, FACP, FASN, Associate Professor of Medicine, Wayne State University School of Medicine; Medical Director of Hemodialysis, Harper University Hospital at Detroit Medical Center; Medical Director, DaVita Greenview Dialysis (Southfield)

Disclosure: Nothing to disclose.

References

  1. Jayne DR, Gaskin G, Rasmussen N, et al. Randomized trial of plasma exchange or high-dosage methylprednisolone as adjunctive therapy for severe renal vasculitis. J Am Soc Nephrol. 2007 Jul. 18(7):2180-8. [View Abstract]
  2. Gupta R, Singh L, Sharma A, Bagga A, Agarwal SK, Dinda AK. Crescentic glomerulonephritis: a clinical and histomorphological analysis of 46 cases. Indian J Pathol Microbiol. 2011 Jul-Sep. 54(3):497-500. [View Abstract]
  3. Bollée G, Flamant M, Schordan S, Fligny C, Rumpel E, Milon M, et al. Epidermal growth factor receptor promotes glomerular injury and renal failure in rapidly progressive crescentic glomerulonephritis. Nat Med. 2011 Sep 25. 17(10):1242-50. [View Abstract]
  4. Stone JH, Merkel PA, Spiera R, Seo P, Langford CA, Hoffman GS, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. Jul 2010. 363(3):221-32. [View Abstract]
  5. Specks U, Merkel PA, Seo P, Spiera R, Langford CA, Hoffman GS, et al. Efficacy of remission-induction regimens for ANCA-associated vasculitis. N Engl J Med. Aug 2013. 369(5):417-27. [View Abstract]
  6. Jones RB, Tervaert JW, Hauser T, Luqmani R, Morgan MD, Peh CA, et al. Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis. N Engl J Med. Jul 2010. 363(3):211-20. [View Abstract]
  7. De Groot K, Rasmussen N, Bacon PA, Tervaert JW, Feighery C, Gregorini G, et al. Randomized trial of cyclophosphamide versus methotrexate for induction of remission in early systemic antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. Aug 2005. 52(8):2461-9. [View Abstract]
  8. Wegener's Granulomatosis Etanercept Trial (WGET) Research Group. Etanercept plus standard therapy for Wegener's granulomatosis. N Engl J Med. Jan 2005. 352(4):351-61. [View Abstract]
  9. Walsh M, Casian A, Flossmann O, Westman K, Höglund P, Pusey C, et al. Long-term follow-up of patients with severe ANCA-associated vasculitis comparing plasma exchange to intravenous methylprednisolone treatment is unclear. Kidney Int. Aug 2013. 84(2):397-402. [View Abstract]
  10. Walsh M, Merkel PA, Peh CA, Szpirt W, Guillevin L, Pusey CD, et al. Plasma exchange and glucocorticoid dosing in the treatment of anti-neutrophil cytoplasm antibody associated vasculitis (PEXIVAS): protocol for a randomized controlled trial. Trials. Mar 2013. 14:73. [View Abstract]
  11. Hiemstra TF, Walsh M, Mahr A, Savage CO, de Groot K, Harper L, et al. Mycophenolate mofetil vs azathioprine for remission maintenance in antineutrophil cytoplasmic antibody-associated vasculitis: a randomized controlled trial. JAMA. Dec 2010. 304(21):2381-8. [View Abstract]
  12. Roubaud-Baudron C, Pagnoux C, Méaux-Ruault N, Grasland A, Zoulim A, LE Guen J, et al. Rituximab maintenance therapy for granulomatosis with polyangiitis and microscopic polyangiitis. J Rheumatol. Jan 2012. 39(1):125-30. [View Abstract]
  13. European therapeutic trials in ANCA-associated systemic vasculitis: disease scoring, consensus regimens and proposed clinical trials. European Community Study Group on Clinical Trials in Systemic Vasculitis ECSYSVASTRIAL. Clin Exp Immunol. 1995 Jul. 101 Suppl 1:29-34. [View Abstract]
  14. Walsh M, Chaudhry A, Jayne D. Long-term follow-up of relapsing/refractory anti-neutrophil cytoplasm antibody associated vasculitis treated with the lymphocyte depleting antibody alemtuzumab (CAMPATH-1H). Ann Rheum Dis. 2008 Sep. 67(9):1322-7. [View Abstract]
  15. de Lind van Wijngaarden RA, Hauer HA, Wolterbeek R, et al. Chances of renal recovery for dialysis-dependent ANCA-associated glomerulonephritis. J Am Soc Nephrol. 2007 Jul. 18(7):2189-97. [View Abstract]
  16. Aoki S, Kotooka N, Yokoyama M, et al. Recurrence of rapidly progressive glomerulonephritis probably associated with two different kinds of drugs. Clin Nephrol. 2000 Sep. 54(3):249-51. [View Abstract]
  17. Bachmeyer C, Cadranel JF, Demontis R. Rituximab is an alternative in a case of contra-indication of cyclophosphamide in Wegener's granulomatosis. Nephrol Dial Transplant. 2005 Jun. 20(6):1274. [View Abstract]
  18. Bombassei GJ, Kaplan AA. The association between hydrocarbon exposure and anti-glomerular basement membrane antibody-mediated disease (Goodpasture's syndrome). Am J Ind Med. 1992. 21(2):141-53. [View Abstract]
  19. Booth AD, Pusey CD, Jayne DR. Renal vasculitis--an update in 2004. Nephrol Dial Transplant. 2004 Aug. 19(8):1964-8. [View Abstract]
  20. Cole E, Cattran D, Magil A, et al. A prospective randomized trial of plasma exchange as additive therapy in idiopathic crescentic glomerulonephritis. The Canadian Apheresis Study Group. Am J Kidney Dis. 1992 Sep. 20(3):261-9. [View Abstract]
  21. Couser WG. Idiopathic rapidly progressive glomerulonephritis. Am J Nephrol. 1982. 2(2):57-69. [View Abstract]
  22. Etanercept plus standard therapy for Wegener's granulomatosis. N Engl J Med. 2005 Jan 27. 352(4):351-61. [View Abstract]
  23. European Vasculitis Study Group. Clinical trial protocol: REMAIN. Randomised trial or prolonged remission-maintenance therapy in systemic vasculitis. June 2003.
  24. Falk RJ, Jennette JC. ANCA small-vessel vasculitis. J Am Soc Nephrol. 1997 Feb. 8(2):314-22. [View Abstract]
  25. Flossmann O, Jones RB, Jayne DR, et al. Should rituximab be used to treat antineutrophil cytoplasmic antibody associated vasculitis?. Ann Rheum Dis. 2006 Jul. 65(7):841-4. [View Abstract]
  26. Garcia-Canton C, Toledo A, Palomar R, et al. Goodpasture's syndrome treated with mycophenolate mofetil. Nephrol Dial Transplant. 2000 Jun. 15(6):920-2. [View Abstract]
  27. Goek ON, Stone JH. Randomized controlled trials in vasculitis associated with anti-neutrophil cytoplasmic antibodies. Curr Opin Rheumatol. 2005 May. 17(3):257-64. [View Abstract]
  28. Jarraya F, Abid M, Jlidi R, et al. Myeloperoxidase-antineutrophil cytoplasmic antibody-positive crescentic glomerulonephritis associated with benzylthiouracil therapy: report of the first case. Nephrol Dial Transplant. 2003 Nov. 18(11):2421-3. [View Abstract]
  29. Jayne D, Rasmussen N, Andrassy K, et al. A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med. 2003 Jul 3. 349(1):36-44. [View Abstract]
  30. Jayne DRW, Gaskin G, (EUVAS). Randomized trial of cyclophosphamide versus azathioprine during remission in ANCA-associated vasculitis (CYCAZAREM). J Am Soc Nephrol. 1999. 10:105A.
  31. Johnson JP, Moore J Jr, Austin HA 3rd, et al. Therapy of anti-glomerular basement membrane antibody disease: analysis of prognostic significance of clinical, pathologic and treatment factors. Medicine (Baltimore). 1985 Jul. 64(4):219-27. [View Abstract]
  32. Jordan SC. Treatment of systemic and renal-limited vasculitic disorders with pooled human intravenous immune globulin. J Clin Immunol. 1995 Nov. 15(6 Suppl):76S-85S. [View Abstract]
  33. Kallenbach M, Duan H, Ring T. Rituximab induced remission in a patient with Wegener's granulomatosis. Nephron Clin Pract. 2005. 99(3):c92-6. [View Abstract]
  34. Langford CA. How can relapses be detected and prevented in primary systemic small-vessel vasculitides?. Best Pract Res Clin Rheumatol. 2005 Apr. 19(2):307-20. [View Abstract]
  35. Masala A, Faedda R, Alagna S, et al. Use of testosterone to prevent cyclophosphamide-induced azoospermia. Ann Intern Med. 1997 Feb 15. 126(4):292-5. [View Abstract]
  36. Murray AN, Cassidy MJ, Templecamp C. Rapidly progressive glomerulonephritis associated with rifampicin therapy for pulmonary tuberculosis. Nephron. 1987. 46(4):373-6. [View Abstract]
  37. Nowack R, Gobel U, Klooker P, et al. Mycophenolate mofetil for maintenance therapy of Wegener's granulomatosis and microscopic polyangiitis: a pilot study in 11 patients with renal involvement. J Am Soc Nephrol. 1999 Sep. 10(9):1965-71. [View Abstract]
  38. Ogata H, Kubo M, Tamaki K, et al. Crescentic glomerulonephritis due to rifampin treatment in a patient with pulmonary atypical mycobacteriosis. Nephron. 1998. 78(3):319-22. [View Abstract]
  39. Pagniez D, Fortin F, Delvallez L, et al. [Favorable course under cotrimoxazole of excavated pneumopathy complicating rapidly progressive glomerulonephritis: Wegener's granulomatosis?]. Rev Med Interne. 1989 Mar-Apr. 10(2):143-6. [View Abstract]
  40. Stilmant MM, Bolton WK, Sturgill BC, et al. Crescentic glomerulonephritis without immune deposits: clinicopathologic features. Kidney Int. 1979 Feb. 15(2):184-95. [View Abstract]
  41. Thompson CH, Kalowski S. Anti-glomerular basement membrane nephritis due to hydralazine. Nephron. 1991. 58(2):238-9. [View Abstract]
  42. Walsh M, Jayne D. Rituximab in the treatment of anti-neutrophil cytoplasm antibody associated vasculitis and systemic lupus erythematosus: past, present and future. Kidney Int. 2007 Sep. 72(6):676-82. [View Abstract]
  43. Guillevin L, Pagnoux C, Karras A, Khouatra C, Aumaître O, Cohen P, et al. Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. N Engl J Med. Nov 2014. 371(19):1771-80. [View Abstract]

Glomerulonephritis, crescentic. Light microscopy (200x hematoxylin and eosin stain): The normal renal architecture is lost. The glomerulus (*) is solid and hypercellular and surrounded by severe interstitial inflammatory infiltrate. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Light microscopy (200x hematoxylin and eosin stain): The normal renal architecture is lost. The glomerulus (*) is solid and hypercellular and surrounded by severe interstitial inflammatory infiltrate. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Light microscopy (25x hematoxylin and eosin stain): Compression of the glomerular tuft with a circumferential cellular crescent that occupies most of the Bowman space. Image courtesy of Madeleine Moussa, MD, FRCPC, Department of Pathology, London Health Sciences Centre, London, Ontario, Canada.

Glomerulonephritis, crescentic. Light microscopy (200x periodic acid-Schiff stain): Bowman capsule (arrow) surrounds each glomerulus. The glomerular tuft (*) is distorted by a proliferation of epithelial cells (crescent), which replaces the urinary space. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Light microscopy (400x trichrome stain): The remnant of the glomerular tuft (*) is surrounded by the cellular crescent with abundant fibrin–red on trichrome stain. Interstitial edema separates the tubules, and scarce inflammatory cells are present. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Immunofluorescence (25x): Anti–glomerular basement membrane characterized by the presence of linear immunoglobulin G deposit along the glomerular basement membrane. Image courtesy of Madeleine Moussa, MD, FRCPC, Department of Pathology, London Health Sciences Centre, London, Ontario, Canada.

Glomerulonephritis, crescentic. Light microscopy (25x hematoxylin and eosin stain): Compression of the glomerular tuft with a circumferential cellular crescent that occupies most of the Bowman space. Image courtesy of Madeleine Moussa, MD, FRCPC, Department of Pathology, London Health Sciences Centre, London, Ontario, Canada.

Glomerulonephritis, crescentic. Light microscopy (200x hematoxylin and eosin stain): The normal renal architecture is lost. The glomerulus (*) is solid and hypercellular and surrounded by severe interstitial inflammatory infiltrate. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Light microscopy (200x periodic acid-Schiff stain): Bowman capsule (arrow) surrounds each glomerulus. The glomerular tuft (*) is distorted by a proliferation of epithelial cells (crescent), which replaces the urinary space. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Light microscopy (400x trichrome stain): The remnant of the glomerular tuft (*) is surrounded by the cellular crescent with abundant fibrin–red on trichrome stain. Interstitial edema separates the tubules, and scarce inflammatory cells are present. Image courtesy of Suzanne Meleg-Smith, MD, Department of Pathology, Tulane University School of Medicine, New Orleans.

Glomerulonephritis, crescentic. Immunofluorescence (25x): Anti–glomerular basement membrane characterized by the presence of linear immunoglobulin G deposit along the glomerular basement membrane. Image courtesy of Madeleine Moussa, MD, FRCPC, Department of Pathology, London Health Sciences Centre, London, Ontario, Canada.