Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Lupus ; 28(9): 1091-1100, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31291846

ABSTRACT

BACKGROUND: The study aimed to investigate whether HLA-G antigen is expressed in the kidneys of patients affected by lupus nephritis (LN) and whether its detection in renal biopsies could be adopted as a marker of treatment response and prognosis. METHODS: Thirty renal biopsies from patients with LN were selected and analyzed through immunohistochemistry. Laboratory and clinical data were retrospectively collected at baseline, 6 and 12 months and at the latest clinical appointment. A number of patients (63.3%) were treated with rituximab (RTX) +/- methylprednisolone in the induction phase. The expression of HLA-G in glomeruli, tubules and infiltrating cells was examined and compared between lupus patients who achieved either complete or partial renal response and those who did not respond to treatment. RESULTS: HLA-G staining was observed in the glomeruli of 20 of 30 samples from patients with LN. The expression of the antigen was detected in podocytes, along glomerular capillary walls, on parietal glomerular epithelial cells and within the juxtaglomerular apparatus. Seventy per cent of patients whose glomeruli expressed HLA-G achieved partial or complete response at 6 months and 75% at the latest available follow up compared with 30% and 40%, respectively, of those who did not show any expression. The pattern of staining in tubules and infiltrating cells was highly variable precluding any clinical correlation. CONCLUSION: This study demonstrates that HLA-G is expressed in renal tissue in LN. Our retrospective data suggest that its expression could correlate with response to treatment.


Subject(s)
HLA-G Antigens/immunology , Lupus Nephritis/drug therapy , Methylprednisolone/administration & dosage , Rituximab/administration & dosage , Adult , Anti-Inflammatory Agents/administration & dosage , Biopsy , Female , Follow-Up Studies , Humans , Immunologic Factors/administration & dosage , Lupus Nephritis/immunology , Male , Middle Aged , Pilot Projects , Retrospective Studies , Treatment Outcome , Young Adult
2.
Scand J Immunol ; 88(1): e12671, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29706017

ABSTRACT

Factor H is an important regulator of complement activation in plasma and on cell surfaces in both humans and mice. If FH function is compromised, inappropriate complement activation on self-surfaces can have disastrous effects as seen in the kidney diseases atypical haemolytic uremic syndrome (aHUS) and C3 glomerulopathy. As FH constructs have been proposed to be used in treatment for these diseases, we studied the distribution of exogenous FH fragments in mice. Full-length mFH, mFH1-5 and mFH18-20 fragments were radiolabelled, and their distribution was examined in WT, FH-/- and FH-/- C3-/- mice in vivo. Whole body scintigraphy revealed accumulation of radioactivity in the abdominal part of the mice, but also to the thyroid gland and urinary bladder. At organ level in WT mice, some full-length FH accumulated in internal organs, but most of it remained in the circulation. Both of the mFH fragments accumulated in the kidneys and were excreted in urine. For mFH1-5, urinary secretion is the likely cause for the accumulation. Concentration of mFH18-20 to kidneys was slower, and at tissue level, mFH18-20 was localized at the proximal tubuli in WT and FH-/- C3-/- mice. No C3-independent binding to glomeruli was detected. In conclusion, these results show that glomerular glycosaminoglycans and sialic acids alone do not collect FH in kidneys. Deposition of C3 fragments is also needed, which implies that in aHUS, the problem is in simultaneous recognition of C3 fragments and glycosaminoglycans or sialic acids by FH, not just the inability of FH to recognize glomerular endothelium as such.


Subject(s)
Complement Factor H/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/metabolism , Tissue Distribution
3.
Clin Exp Immunol ; 192(3): 337-347, 2018 06.
Article in English | MEDLINE | ID: mdl-29405270

ABSTRACT

Hyperlipidaemia accompanies chronic renal disease either as a consequence of the renal dysfunction or as part of generalized metabolic derangements. Under both situations, the lipid profile is characterized by accumulation of triglyceride-rich lipoproteins (TGRLs). This lipid profile is recognized as a risk factor for cardiovascular complications. Whether it may pose a risk for renal injury as well remains unclear. A hyper-TGRL state was generated in C57BL/6 mice using poloxamer-407 (P-407) and immune complex-mediated renal injury was triggered using the accelerated nephrotoxic nephritis (ANTN) model. The hyper-TGRL animals were hypersensitive to ANTN demonstrated by greater haematuria and glomerular cellularity. These changes were accompanied by increased glomerular accumulation of CD68+ macrophages. The hypersensitive response to ANTN was not seen in low-density lipoprotein receptor knock-out mice fed with a high fat diet, where triglyceride levels were lower but cholesterol levels comparable to those obtained using P-407. These data indicate that a hyper-TGRL state might be more detrimental to the kidneys than low-density lipoprotein-driven hypercholesterolaemia during immune complex-mediated nephritis. We speculate that the hyper-TGRL environment primes the kidney to exacerbated renal damage following an inflammatory insult with increased accumulation of macrophages that may play a key role in mediating the injurious effects.


Subject(s)
Acute Kidney Injury/pathology , Hypercholesterolemia/pathology , Lipoproteins/metabolism , Macrophages/immunology , Nephritis/pathology , Triglycerides/metabolism , Acute Kidney Injury/chemically induced , Animals , Complement C3/metabolism , Disease Models, Animal , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/chemically induced , Poloxamer/toxicity
4.
Clin Exp Immunol ; 176(1): 84-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24279761

ABSTRACT

Uncontrolled activation of the complement alternative pathway is associated with complement-mediated renal disease. Factor B and factor D are essential components of this pathway, while factor H (FH) is its major regulator. In complete FH deficiency, uncontrolled C3 activation through the alternative pathway results in plasma C3 depletion and complement-mediated renal disease. These are dependent on factor B. Mannan-binding lectin-associated serine proteases 1 and 3 (MASP-1, MASP-3) have been shown recently to contribute to alternative pathway activation by cleaving pro-factor D to its active form, factor D. We studied the contribution of MASP-1 and MASP-3 to uncontrolled alternative pathway activation in experimental complete FH deficiency. Co-deficiency of FH and MASP-1/MASP-3 did not ameliorate either the plasma C3 activation or glomerular C3 accumulation in FH-deficient mice. Our data indicate that MASP-1 and MASP-3 are not essential for alternative pathway activation in complete FH deficiency.


Subject(s)
Complement C3/immunology , Complement Factor H/deficiency , Complement Factor H/immunology , Kidney Diseases/immunology , Mannose-Binding Protein-Associated Serine Proteases/immunology , Animals , Blotting, Western , Complement Activation/immunology , Complement C3/metabolism , Complement C5/immunology , Complement C5/metabolism , Complement Factor B/immunology , Complement Factor B/metabolism , Complement Factor D/immunology , Complement Factor D/metabolism , Complement Factor H/genetics , Complement Pathway, Alternative/genetics , Complement Pathway, Alternative/immunology , Enzyme-Linked Immunosorbent Assay , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/metabolism , Hereditary Complement Deficiency Diseases , Kidney Diseases/blood , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Mannose-Binding Protein-Associated Serine Proteases/deficiency , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Clin Exp Immunol ; 174(2): 326-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23919682

ABSTRACT

Dengue disease is a mosquito-borne infection caused by Dengue virus. Infection may be asymptomatic or variably manifest as mild Dengue fever (DF) to the most severe form, Dengue haemorrhagic fever (DHF). Mechanisms that influence disease severity are not understood. Complement, an integral component of the immune system, is activated during Dengue infection and the degree of activation increases with disease severity. Activation of the complement alternative pathway is influenced by polymorphisms within activation (factor B rs12614/rs641153, C3 rs2230199) and regulatory [complement factor H (CFH) rs800292] proteins, collectively termed a complotype. Here, we tested the hypothesis that the complotype influences disease severity during secondary Dengue infection. In addition to the complotype, we also assessed two other disease-associated CFH polymorphisms (rs1061170, rs3753394) and a structural polymorphism within the CFH protein family. We did not detect any significant association between the examined polymorphisms and Dengue infection severity in the Thai population. However, the minor allele frequencies of the factor B and C3 polymorphisms were less than 10%, so our study was not sufficiently powered to detect an association at these loci. We were also unable to detect a direct interaction between CFH and Dengue NS1 using both recombinant NS1 and DV2-infected culture supernatants. We conclude that the complotype does not influence secondary Dengue infection severity in the Thai population.


Subject(s)
Complement Factor H/genetics , Complement Pathway, Alternative/genetics , Dengue Virus , Dengue/genetics , Dengue/immunology , Adolescent , Adult , Cells, Cultured , Child , Child, Preschool , Complement C3/genetics , Complement Factor B/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Genetic , Thailand , Young Adult
6.
Am J Transplant ; 11(1): 152-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21114651

ABSTRACT

Complement factor H-related protein 5 (CFHR5) nephropathy is a familial renal disease endemic in Cyprus. It is characterized by persistent microscopic hematuria, synpharyngitic macroscopic hematuria and progressive renal impairment. Isolated glomerular accumulation of complement component 3 (C3) is typical with variable degrees of glomerular inflammation. Affected individuals have a heterozygous internal duplication in the CFHR5 gene, although the mechanism through which this mutation results in renal disease is not understood. Notably, the risk of progressive renal failure in this condition is higher in males than females. We report the first documented case of recurrence of CFHR5 nephropathy in a renal transplant in a 53-year-old Cypriot male. Strikingly, histological changes of CFHR5 nephropathy were evident in the donor kidney 46 days post-transplantation. This unique case demonstrates that renal-derived CFHR5 protein cannot prevent the development of CFHR5 nephropathy.


Subject(s)
Complement System Proteins/genetics , Glomerulonephritis/genetics , Aged , Complement Factor H/genetics , Cyprus , Female , Humans , Kidney Diseases/genetics , Kidney Transplantation , Male , Middle Aged , Recurrence
7.
Am J Transplant ; 10(1): 168-72, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19951285

ABSTRACT

Factor H (CFH) autoantibodies are associated with atypical hemolytic uremic syndrome (aHUS). Peritransplantation plasma exchange therapy and intensification of immunosuppression, with adjuvant use of anti-CD20 monoclonal antibodies has recently been advocated for cases of CFH-autoantibody associated aHUS. In this report, we describe successful deceased donor renal transplantation in a case of CFH-autoantibody associated aHUS with combined CFHR1 and 3 deficiency in addition to the CFH sequence variant, (cG2850T, pGln950His). CFH-autoantibodies were detected 2 weeks prior to transplantation. Disease recurrence was not observed using basiliximab, an IL2-receptor antagonist and high-dose corticosteroids with mycophenolate mofetil. Adjuvant therapies such as Rituximab nor intensification of plasma therapy were employed. Consequently, careful consideration needs to be given to the use of additional immunosuppression in certain cases of CFH-autoantibody associated aHUS. Serial measurement of CFH-autoantibodies is required in the immediate pre- and posttransplantation period to further clarify their role as a factor in the recurrence of aHUS posttransplantation. Furthermore, delineation of the functional significance of CFH-autoantibodies is warranted in individual cases.


Subject(s)
Autoantibodies/blood , Blood Proteins/deficiency , Complement C3b Inactivator Proteins/deficiency , Complement Factor H/genetics , Complement Factor H/immunology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/surgery , Kidney Transplantation , Amino Acid Substitution , Child , Female , Genetic Variation , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , Humans , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Kidney Transplantation/pathology , Kidney Transplantation/physiology , Polymorphism, Single Nucleotide
8.
Clin Exp Immunol ; 155(2): 339-47, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19040606

ABSTRACT

Haemolytic uraemic syndrome (HUS) is characterized by microangiopathic haemolytic anaemia, thrombocytopenia and renal failure because of thrombotic microangiopathy (TMA). It may be caused by infection with Shiga toxin-producing enteropathic bacteria (Stx-associated HUS) or with genetic defects in complement alternative pathway (CAP) regulation (atypical HUS). We hypothesized that defective complement regulation could increase host susceptibility to Stx-associated HUS. Hence, we studied the response of mice with heterozygous deficiency of the major CAP regulator, factor H, to purified Stx-2. Stx-2 was administered together with lipopolysaccharide to wild-type and Cfh(+/-) C57BL/6 animals. Forty-eight hours after administration of the first Stx-2 injection all animals developed significant uraemia. Renal histology demonstrated significant tubular apoptosis in the cortical and medullary areas which did not differ between wild-type or Cfh(+/-) Stx-2-treated mice. Uraemia and renal tubular apoptosis did not develop in wild-type or Cfh(+/-) animals treated with lipopolysaccharide alone. No light microscopic evidence of TMA or abnormal glomerular C3 staining was demonstrable in the Stx-2 treated animals. In summary, Stx-2 administration did not result in TMA in either Cfh(+/-) or wild-type C57BL/6 mice. Furthermore, haploinsufficiency of factor H did not alter the development of Stx-2-induced renal tubular injury.


Subject(s)
Complement Factor H/deficiency , Hemolytic-Uremic Syndrome/chemically induced , Kidney Tubules/drug effects , Shiga Toxin 2/toxicity , Animals , Apoptosis/drug effects , Female , Hemolysis/drug effects , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Spleen/drug effects , Spleen/pathology , Uremia/chemically induced
10.
Clin Exp Immunol ; 151(2): 210-30, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18190458

ABSTRACT

Factor H is the major regulatory protein of the alternative pathway of complement activation. Abnormalities in factor H have been associated with renal disease, namely glomerulonephritis with C3 deposition including membranoproliferative glomerulonephritis (MPGN) and the atypical haemolytic uraemic syndrome (aHUS). Furthermore, a common factor H polymorphism has been identified as a risk factor for the development of age-related macular degeneration. These associations suggest that alternative pathway dysregulation is a common feature in the pathogenesis of these conditions. However, with respect to factor H-associated renal disease, it is now clear that distinct molecular defects in the protein underlie the pathogenesis of glomerulonephritis and HUS. In this paper we review the associations between human factor H dysfunction and renal disease and explore how observations in both spontaneous and engineered animal models of factor H dysfunction have contributed to our understanding of the pathogenesis of factor H-related renal disease.


Subject(s)
Complement Factor H/deficiency , Glomerulonephritis, Membranoproliferative/immunology , Hemolytic-Uremic Syndrome/immunology , Adolescent , Adult , Animals , Autoantibodies/analysis , Child , Child, Preschool , Complement Factor H/genetics , Complement Factor H/immunology , Complement Pathway, Alternative , Female , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/pathology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , Male , Middle Aged
11.
Kidney Int ; 71(11): 1142-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17410102

ABSTRACT

Complement factor D is a serine protease essential for the activation of the alternative pathway and is expressed in the kidney, adipocytes, and macrophages. Factor D is found at relatively high levels in glomeruli suggesting that this component of the complement cascade could influence renal pathophysiology. In this study, we utilize mice with a targeted deletion of the activating complement factor D gene and compare these results to mice with targeted deletion of the inhibitory complement factor H gene. Eight-month-old mice with a deleted factor D gene spontaneously develop albuminuria and have reduced creatinine clearance due to mesangial immune complex glomerulonephritis. These mesangial deposits contain C3 and IgM. In contrast to the mesangial location of the immune deposits in the factor D-deficient mice, age-matched factor H-deficient mice develop immune deposits along the glomerular capillary wall. Our observations suggest that complement factor D or alternative pathway activation is needed to prevent spontaneous accumulation of C3 and IgM deposits within the mesangium. Our studies show that the complement factor D gene knockout mice are a novel model of spontaneous mesangial immune complex glomerulonephritis.


Subject(s)
Complement Factor D/deficiency , Glomerulonephritis/immunology , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology , Albuminuria , Animals , Antigens, Differentiation/metabolism , Capillaries/immunology , Capillaries/metabolism , Capillaries/ultrastructure , Complement C3/immunology , Complement C3/metabolism , Complement Factor D/genetics , Complement Factor H/deficiency , Complement Factor H/genetics , Creatinine/blood , Creatinine/urine , Female , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Genotype , Glomerular Mesangium/chemistry , Glomerular Mesangium/immunology , Glomerular Mesangium/pathology , Glomerular Mesangium/ultrastructure , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Histocytochemistry , Immune Complex Diseases/genetics , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Immunohistochemistry , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction
12.
Proc Natl Acad Sci U S A ; 103(25): 9649-54, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16769899

ABSTRACT

Membranoproliferative glomerulonephritis (MPGN) type II (dense deposit disease) is an inflammatory renal disease characterized by electron-dense deposits and complement C3 on the glomerular basement membrane. There is no effective therapy. We investigated the role of C5 activation in a model of MPGN that develops spontaneously in complement factor H-deficient mice (Cfh(-/-)). At 12 months there was a significant reduction in mortality, glomerular cellularity, neutrophil numbers, and serum creatinine levels in Cfh(-/-) mice deficient in C5. Excessive glomerular neutrophil numbers, frequently seen in patients with MPGN during disease flares, were also observed in Cfh(-/-) mice after the administration of an antiglomerular basement membrane antibody. This exaggerated injurious phenotype was absent in Cfh(-/-) mice deficient in C5 but not in Cfh(-/-) mice deficient in C6, indicating a key role for C5 activation in the induction of renal lesions. Importantly, the renal injury was completely reversed in Cfh(-/-) mice pretreated with an anti-murine C5 antibody. These results demonstrate an important role for C5 in both spontaneous MPGN and experimentally induced nephritis in factor H-deficient mice and provide preliminary evidence that C5 inhibition therapy might be useful in human MPGN type II.


Subject(s)
Complement Activation , Complement C5/immunology , Complement Factor H/deficiency , Complement Factor H/metabolism , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Complement C5/deficiency , Complement C5/genetics , Complement C6/deficiency , Complement C6/genetics , Complement C6/metabolism , Complement Factor H/genetics , Disease Models, Animal , Glomerulonephritis/genetics , Glomerulonephritis/therapy , Mice , Mice, Knockout , Neutrophils/cytology
13.
J Exp Med ; 194(6): 747-56, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11560991

ABSTRACT

There is evidence that the classical complement pathway may be activated via a "C1-tickover" mechanism, analogous to the C3-tickover of the alternative pathway. We have quantitated and characterized this pathway of complement activation. Analysis of freshly collected mouse and human plasma revealed that spontaneous C3 activation rapidly occurred with the generation of C3 fragments in the plasma. By the use of complement- and Ig-deficient mice it was found that C1q, C4, C2, and plasma Ig were all required for this spontaneous C3 activation, with the alternative complement pathway further amplifying C3 fragment generation. Study of plasma from a human with C1q deficiency before and after therapeutic C1q infusion confirmed the existence of a similar pathway for complement activation in humans. Elevated levels of plasma C3 were detected in mice deficient in complement components required for activation of either the classical or alternative complement pathways, supporting the hypothesis that there is continuous complement activation and C3 consumption through both these pathways in vivo. Blood stasis was found to stimulate C3 activation by classical pathway tick-over. This antigen-independent mechanism for classical pathway activation may augment activation of the complement system at sites of inflammation and infarction.


Subject(s)
Complement Activation/immunology , Complement C3/immunology , Animals , Complement C1q/immunology , Complement C4/immunology , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Tumor Cells, Cultured
14.
J Invest Dermatol ; 117(1): 52-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11442749

ABSTRACT

Exposure to ultraviolet B radiation is an important trigger of both systemic and cutaneous disease flares in individuals with systemic lupus erythematosus. More than 90% of individuals with homozygous C1q deficiency develop a systemic-lupus-erythematosus-like illness, which is typically associated with a severe photosensitive rash. Apoptotic, human keratinocytes have been shown in vitro to bind C1q, in the absence of antibody. These observations, together with the hypothesis that a major source of the autoantigens driving the immune response in systemic lupus erythematosus comes from apoptotic cells, led us to investigate the effects of murine C1q deficiency on ultraviolet-radiation-induced keratinocyte apoptosis in vivo. In this work, we demonstrated C1q binding to apoptotic murine keratinocytes in vitro and showed for the first time that C1q is also present on sunburn cells in vivo. In addition to C1q, we detected C3 deposition on sunburn cells in both wild-type and C1q-deficient mice, suggesting activation of the alternative pathway. Following acute ultraviolet exposure in vivo, no difference in the rate of clearance of sunburn cells was found in C1q-deficient mice from three different genetic backgrounds, compared with strain-matched wild-type controls. Furthermore, chronic ultraviolet exposure did not result in the production of autoantibodies or the development of glomerulonephritis. Our findings suggest that C1q does not play a critical role in the physiologic clearance of apoptotic murine keratinocytes in vivo.


Subject(s)
Apoptosis/radiation effects , Complement C1q/genetics , Keratinocytes/pathology , Ultraviolet Rays , Acute Disease , Animals , Autoantibodies/blood , Autoimmunity/immunology , Autoimmunity/radiation effects , Chronic Disease , Complement C1q/metabolism , Complement C3/metabolism , Epidermis/immunology , Epidermis/pathology , Epidermis/radiation effects , Female , Glomerulonephritis/immunology , In Vitro Techniques , Keratinocytes/metabolism , Keratinocytes/radiation effects , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sunburn/immunology , Sunburn/pathology
16.
J R Coll Physicians Lond ; 34(2): 169-77, 2000.
Article in English | MEDLINE | ID: mdl-10816874

ABSTRACT

We report our clinical experience of 230 patients referred to the Hammersmith hospital with a working diagnosis of Behçet's syndrome. The pathogenesis, diagnosis and management of the syndrome are discussed.


Subject(s)
Behcet Syndrome , Adolescent , Adult , Aged , Algorithms , Behcet Syndrome/classification , Behcet Syndrome/complications , Behcet Syndrome/diagnosis , Behcet Syndrome/etiology , Behcet Syndrome/physiopathology , Behcet Syndrome/therapy , Eye Diseases/drug therapy , Eye Diseases/etiology , Female , Humans , Inflammation , Male , Middle Aged
20.
Curr Opin Rheumatol ; 10(6): 504-10, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9812210

ABSTRACT

Eosinophilic infiltration into skeletal muscle, although rare, has been described in a diverse group of conditions. It most commonly occurs in parasitic infection as focal eosinophilic myositis but can be a feature of systemic hypereosinophilic conditions such as eosinophilia-myalgia syndrome and idiopathic hypereosinophilic syndrome. The majority of cases have no discernible etiological factor. Eosinophilic myopathies should be distinguished from the commoner idiopathic inflammatory myopathies such as polymyositis and dermatomyositis. This report describes the various conditions in which eosinophilic myopathy occurs and reviews the current state of knowledge of eosinophilic myopathy.


Subject(s)
Eosinophilia , Muscular Diseases/immunology , Eosinophilia/chemically induced , Eosinophilia/complications , Eosinophilia-Myalgia Syndrome/immunology , Eosinophils , Humans , Hypereosinophilic Syndrome/immunology , Muscular Diseases/parasitology , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...