ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Hyperimmunoglobulin D syndrome: Pathophysiology

Hyperimmunoglobulin D syndrome: Pathophysiology
Literature review current through: Jan 2024.
This topic last updated: Apr 15, 2022.

INTRODUCTION — Hyperimmunoglobulin D syndrome (HIDS) is a rare genetic disorder characterized by recurrent febrile episodes typically associated with lymphadenopathy, abdominal pain, and an elevated serum polyclonal immunoglobulin D (IgD) level (MIM #260920). The syndrome can be further categorized into classic and variant forms. The genetic defect is known in the classic form, which makes up 75 percent of cases. The variant form has similar clinical manifestations, although its genetic basis is unknown [1].

This topic reviews the genetics and pathophysiology of HIDS. The clinical manifestations, diagnosis, and management of this disorder are discussed in detail separately. (See "Hyperimmunoglobulin D syndrome: Clinical manifestations and diagnosis" and "Hyperimmunoglobulin D syndrome: Management".)

GENETICS — Classic HIDS is caused by mevalonate kinase (MVK) deficiency and is inherited as an autosomal-recessive trait [2-6]. Most patients are compound heterozygous for two different mutations in the MVK gene [7-9]. However, some patients may be homozygous for the same defect on both alleles [10]. Patients with similar clinical features but lacking mutant MVK genes are referred to as having variant HIDS. (See 'Variant HIDS' below.)

Mevalonate kinase — The MVK gene (MIM *251170) is located on the long arm of chromosome 12 (12q24) [11]. The gene product, MVK, is a cytosolic protein that is localized to the peroxisome [12,13]. It is an enzyme in the cholesterol synthesis pathway. This pathway is responsible for the synthesis of sterol products, such as cholesterol and its derivatives, as well as nonsterol isoprenoids, including prenylated proteins, heme A, dolichol, and ubiquinone 10 [14,15]. MVK is one enzyme downstream of the highly regulated hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase enzyme (figure 1).

Compound heterozygous MVK mutations in HIDS patients usually consist of one allele with a mutation that causes mevalonic aciduria when present in a homozygous state (eg, H20P, I268T, or A334T mutation) [16] (see "Hyperimmunoglobulin D syndrome: Clinical manifestations and diagnosis", section on 'Mevalonic aciduria'). The other allele has a mutation that is not typical for mevalonic aciduria. The V377I mutation is the most common mutation in patients with HIDS (52 to >90 percent) [11,17-19]. In the remaining HIDS patients without the V377I mutation, the second mutation probably causes a similar enzymatic defect, leading to residual MVK activity [2].

The V377I mutation and others like it probably prevent appropriate folding of newly formed MVK proteins at body temperature (37°C), thereby causing decreased in vivo enzyme activity [14,19]. A mouse model of HIDS was created via the deletion of a single MVK allele, supporting diminished MVK activity as the probable cause of HIDS [20].

Other genetic factors — The estimated incidence of HIDS is 1:5000 to 1:50,000 in the general population, although the observed incidence is much lower, suggesting decreased penetrance of the disease [16,21]. Autosomal-recessive homozygous genotypes, especially of the V377I mutation, are encountered with less frequency than expected, assuming these mutations are in Hardy-Weinberg equilibrium within the population, suggesting incomplete penetrance of this genotype specifically [4,16].

Homozygous V377I mutations would theoretically allow for double the enzymatic activity found in compound heterozygous HIDS patients with a V377I and a nonfunctional allele (eg, H20P, I268T, or A334T mutations), with a range of 1 to 12 percent of residual enzyme activity expected. This configuration may or may not result in sufficient reduction of enzymatic activity to yield the HIDS phenotype, thereby contributing to the observed incomplete penetrance [16]. As an example, of two sisters with identical homozygous V377I mutations, only one was symptomatic [22]. In addition, other minor mutations may be present in downstream enzymes in such homozygous patients who demonstrate the HIDS phenotype [23].

Variant HIDS — Defects in downstream enzymes have been proposed to account for variant HIDS, given that the phenotypes are essentially the same [24]. Such mutations would also create a lack of nonsterol isoprenoids, one of the mechanisms proposed to cause febrile episodes in classic HIDS. It is likely that these conditions will take on other names as the gene mutations underlying variant HIDS are identified. It is also possible that regulatory region mutations for MVK itself exist that evaded identification via standard sequencing of the gene. (See 'Nonsterol synthesis pathway products' below and "Hyperimmunoglobulin D syndrome: Clinical manifestations and diagnosis", section on 'Variant HIDS'.)

Overlap syndromes — Mutations found in other rare periodic fever syndromes have been identified in some HIDS patients [25,26]. These "overlap syndromes" may account for clinical features that are unusual in HIDS patients, such as amyloidosis [27-29]. One patient with HIDS and systemic amyloidosis was homozygous for the alpha/alpha genotype of the serum amyloid A 1 (SAA1) gene and was also a compound heterozygote for HIDS mutations (H20P and V250I) [29]. Such coincidental mutations may also account for atypical responses to medical treatment [30]. Further genetic and clinical studies on patients with hereditary periodic fever syndromes are required to support the concept of overlap syndromes. (See "Hyperimmunoglobulin D syndrome: Clinical manifestations and diagnosis", section on 'Overlap syndromes'.)

PATHOPHYSIOLOGY — A successful explanatory model of HIDS pathophysiology needs to account for the periodicity of fevers, the frequent presence of increased serum levels of immunoglobulin D (IgD) and/or immunoglobulin A (IgA), and the HIDS phenotype in patients with no identifiable mutations in the MVK gene. Although reduced MVK enzyme activity that results from mutations in the MVK gene is responsible for many cases of HIDS, the definitive pathophysiologic mechanisms through which the genetic defect causes the HIDS phenotype remain unknown.

One explanatory model begins with triggering events that cause transient elevations in body temperature. Temperature sensitivity of the MVK enzyme leads to the decreased production of mevalonate 5-phosphate and downstream nonsterol isoprenoids, particularly those with high turnover (ubiquinone 10 in patients with mevalonic aciduria and antiinflammatory isoprenoids in HIDS patients), promoting a proinflammatory phenotype with more fever and inflammation [14,19]. In HIDS patients, this cascade leads to increased 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR) activity that increases mevalonate levels and drives production of nonsterol end-products despite persistently decreased MVK enzyme activity at higher temperatures. Eventually, homeostasis is restored, and the febrile episode ends. This theory explains why events such as vaccinations, minor trauma, surgery, or stress can trigger an attack in HIDS patients and is in general agreement with the observed phenotype and the above in vitro data. More clinical and molecular studies are required, though, before this theory can become widely accepted.

Serum immunoglobulin D — Some in vitro experimental data suggest that IgD is part of the autoinflammatory process rather than an epiphenomenon. This proinflammatory effect was illustrated when peripheral blood mononuclear cells (PBMCs) from normal patients were exposed in vitro to IgD purified from an HIDS patient [31]. Large quantities of interleukin (IL) 1, IL-6, IL-1 receptor antagonist (IL-1Ra), and tumor necrosis factor alpha (TNF-alpha) were detected. IgD also enhanced cytokine production in response to lipopolysaccharide (LPS), a potent inducer of inflammatory cytokines.

The degree of O-glycosylation of IgD and IgA1 may alter the inflammatory nature of these immunoglobulins. The T cell IgD receptor is a lectin that has a binding affinity for the O-glycans of IgD and IgA1. Patients with HIDS have increased O-galactosylation of IgD and decreased O-sialylation of both IgD and IgA1 compared with normal controls [32]. This may have a significant effect on basal immunoglobulin synthesis, as well as myeloid cytokine release. However, serum IgD and its O-galactosylation are elevated between, as well as during, attacks. In addition, IgD levels do not appear to correlate with the frequency or severity of attacks [33].

Mevalonate — In patients with HIDS, increased excretion of mevalonate (mevalonic aciduria) is transient and only occurs during febrile episodes. Studies suggest that elevated mevalonate (mevalonic acid) levels more likely help to prevent attacks than to promote them [34]. As an example, treatment of two patients with the HMGR inhibitor lovastatin, which should have reduced mevalonate synthesis, resulted in severe crises in both cases [35].

Supplying metabolic products downstream of mevalonate may have a favorable antiinflammatory effect. This was illustrated in an in vitro study using PBMCs from patients with HIDS [36]. Production of IL-1 beta was increased relative to that from PBMCs of healthy controls. A further increase in IL-1 beta was noted following blockade of HMGR using lovastatin but was partially reversed by adding farnesol (FOH) and geranylgeraniol (GGOH), two downstream intermediates in the cholesterol synthetic pathway. FOH decreased IL-1 beta production by 62 percent in cells from HIDS patients. Adding mevalonic acid caused a smaller effect but also decreased, rather than increased, IL-1 beta release.

Sterol synthesis pathway products — A deficiency of cholesterol or its derivative seems unlikely to be important in causing fever or other symptoms of HIDS. Patients with HIDS appear to have normal cholesterol levels [37] and continued cholesterol synthesis in cultured cell lines [34].

Nonsterol synthesis pathway products — Disruption of cellular signaling caused by decreased production of nonsterol pathway products, cytokine derangements, and an inflammatory process seem plausible results of partial MVK enzyme deficiency. However, this does not explain the limited and periodic nature of HIDS attacks.

Molecular studies in a mouse model, cell lines, and primary human cells further support this theory [38-44]. A mouse model, where the isoprenoid pathway is inhibited by bisphosphonates and inflammation is further stimulated by bacterial muramyl dipeptide, showed an increased inflammatory response [44]. This response was attenuated by the addition of exogenous isoprenoids (geraniol, farnesol, and geranylgeraniol). RAW264.7 cell lines and primary monocytes from healthy controls demonstrated similar effects, with lipopolysaccharide replacing muramyl dipeptide as the additional stimulus [41]. In cellular and animal models, inhibition of the mevalonate pathway with a bisphosphonate (alendronate or pamidronate) increased expression of NLRP3 (nacht domain-, leucine-rich repeat-, and pyrin domain-containing 3), a key inflammasome component and mediator of innate immunity linked to IL-1 beta secretion [38,42].

The inflammation in these models was also reduced by the addition of manumycin A, an inhibitor of farnesyl transferase, which prevented the consumption of farnesyl pyrophosphate and increased the production of geranylgeranyl pyrophosphate (figure 1). Newer farnesyl transferase inhibitors, tipifarnib and lonafarnib, demonstrated similar effects [43]. It is therefore proposed that the inflammatory response is linked to a shortage of geranylgeranyl pyrophosphate, which is in agreement with other studies linking geranylgeranylation with IL-1 beta activation [45-47].

Some cholesterol biosynthetic pathway intermediates, such as polyisoprenyl phosphates (PIPP), presqualene diphosphate (PSDP), and presqualene monophosphate (PSMP) may serve as potent signaling molecules [48]. The nonsterol end-products, the isoprenoids, also play important cellular roles, including as components of prenylated proteins (cytokine signaling, cell cycling, cell development), heme A (electron transport), dolichol (glycoprotein synthesis), ubiquinone 10 (electron transport, antioxidant), and isopentenyl tRNAs [14,15]. Some small G-proteins, which are involved in cellular signaling, are prenylated proteins.

One such G-protein of the Rho family, Rac-1, plays a critical role in caspase 1 activation through inflammasome oligomerization [34,49]. The NLRP3 inflammasome is linked to familial Mediterranean fever (FMF) and cryopyrin-associated periodic syndromes (CAPS). Rac-1 has increased activity in the presence of simvastatin, an HMGR inhibitor [45,50]. Other studies have shown that inhibition of the mevalonate pathway with a geranylgeranyl-transferase inhibitor and with simvastatin leads to a caspase 1-dependent release of IL-1 beta and IL-18 by activated monocytes [45,50,51]. Farnesyl transferase inhibitors block this effect [50]. Additionally, dermal fibroblasts cultured from healthy donors increased production of IL-1 beta to levels matching that of dermal fibroblasts cultured from mevalonate kinase-deficient (MKD) donors when exposed to simvastatin. Combining simvastatin with mevalonate reduces IL-1 beta production in fibroblasts from healthy donors but not in those from MKD donors [52].

Secondary mediators — Regardless of the primary cause, cytokine activation is thought to contribute to the symptoms experienced by patients with HIDS during febrile episodes. Elevated circulating IL-6 and TNF-alpha levels are present during febrile episodes compared with asymptomatic intervals [53]. IL-6 levels correlate significantly with C-reactive protein (CRP) levels. Antiinflammatory molecules, such as IL-1Ra and soluble TNF receptor superfamily 1A (sTNFRSF1A: sTNFr p55 and p75) are also elevated during attacks. Inflammatory IL-1 alpha, IL-1 beta, and the antiinflammatory cytokine, IL-10, levels were not elevated during febrile episodes.

Persistently elevated alpha1-acid glycoprotein (AGP) in patients with HIDS compared with normal controls points to a continued inflammatory state even between attacks [54]. Finally, increased urinary neopterin levels in HIDS patients during febrile episodes, which normalize between attacks, represent products of monocytes and macrophages that have been stimulated by interferon gamma [55].

SUMMARY

Hyperimmunoglobulin D syndrome (HIDS) is characterized by febrile episodes that are typically associated with lymphadenopathy, abdominal pain, and an elevated serum polyclonal immunoglobulin D (IgD) level. (See "Hyperimmunoglobulin D syndrome: Clinical manifestations and diagnosis".)

Mutations of the mevalonate kinase (MVK) gene are responsible for the autosomal-recessive pattern of inheritance seen in many cases of the classic HIDS. Impaired folding of the MVK protein at body temperature may play a role in decreased enzyme activity. A similar phenotype may occur in the absence of mutations of the MVK gene and is referred to as variant HIDS. (See 'Genetics' above.)

The pathophysiology of HIDS and mevalonic aciduria are incompletely understood. Although IgD levels are typically elevated in HIDS, it is unlikely that elevated serum levels of IgD are responsible for the periodic fevers and other symptoms of HIDS. (See 'Serum immunoglobulin D' above.)

Neither the increased production nor the increased excretion of mevalonate (mevalonic acid) appears to be the proximate cause of the inflammatory phenotype. Attempts to decrease mevalonate production using statin drugs in patients with HIDS have led to worsening of disease.

Decreased availability of nonsterol biosynthetic pathway intermediates (which may have antiinflammatory properties) may be of pathophysiologic importance. In vitro studies suggest that supplying exogenous precursors (farnesol and geranylgeraniol) for downstream products may reduce the production of proinflammatory cytokines from cultured cells from patients with HIDS. Additionally, inhibition of farnesyl transferase may also reduce proinflammatory cytokines by increasing the availability of geranylgeranyl-PP. (See 'Nonsterol synthesis pathway products' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges E Richard Stiehm, MD, who contributed as a Section Editor to earlier versions of this topic review.

  1. Simon A, Cuisset L, Vincent MF, et al. Molecular analysis of the mevalonate kinase gene in a cohort of patients with the hyper-igd and periodic fever syndrome: its application as a diagnostic tool. Ann Intern Med 2001; 135:338.
  2. Hospach T, Lohse P, Heilbronner H, et al. Pseudodominant inheritance of the hyperimmunoglobulinemia D with periodic fever syndrome in a mother and her two monozygotic twins. Arthritis Rheum 2005; 52:3606.
  3. Frenkel J, Kuis W. Overt and occult rheumatic diseases: the child with chronic fever. Best Pract Res Clin Rheumatol 2002; 16:443.
  4. Simon A, Mariman EC, van der Meer JW, Drenth JP. A founder effect in the hyperimmunoglobulinemia D and periodic fever syndrome. Am J Med 2003; 114:148.
  5. Drenth JP, Cuisset L, Grateau G, et al. Mutations in the gene encoding mevalonate kinase cause hyper-IgD and periodic fever syndrome. International Hyper-IgD Study Group. Nat Genet 1999; 22:178.
  6. Houten SM, Kuis W, Duran M, et al. Mutations in MVK, encoding mevalonate kinase, cause hyperimmunoglobulinaemia D and periodic fever syndrome. Nat Genet 1999; 22:175.
  7. Sinha A, Waterham HR, Sreedhar KV, Jain V. Novel mutations causing hyperimmunoglobulin D and periodic fever syndrome. Indian Pediatr 2012; 49:583.
  8. Mizuno T, Sakai H, Nishikomori R, et al. Novel mutations of MVK gene in Japanese family members affected with hyperimmunoglobulinemia D and periodic fever syndrome. Rheumatol Int 2012; 32:3761.
  9. Schlabe S, Schwarze-Zander C, Lohse P, Rockstroh JK. Hyper-IgD and periodic fever syndrome (HIDS) due to compound heterozygosity for G336S and V377I in a 44-year-old patient with a 27-year history of fever. BMJ Case Rep 2016; 2016.
  10. Santos JA, Aróstegui JI, Brito MJ, et al. Hyper-IgD and periodic fever syndrome: a new MVK mutation (p.R277G) associated with a severe phenotype. Gene 2014; 542:217.
  11. Houten SM, Koster J, Romeijn GJ, et al. Organization of the mevalonate kinase (MVK) gene and identification of novel mutations causing mevalonic aciduria and hyperimmunoglobulinaemia D and periodic fever syndrome. Eur J Hum Genet 2001; 9:253.
  12. Biardi L, Sreedhar A, Zokaei A, et al. Mevalonate kinase is predominantly localized in peroxisomes and is defective in patients with peroxisome deficiency disorders. J Biol Chem 1994; 269:1197.
  13. Wanders RJ, Romeijn GJ. Differential deficiency of mevalonate kinase and phosphomevalonate kinase in patients with distinct defects in peroxisome biogenesis: evidence for a major role of peroxisomes in cholesterol biosynthesis. Biochem Biophys Res Commun 1998; 247:663.
  14. Houten SM, Frenkel J, Waterham HR. Isoprenoid biosynthesis in hereditary periodic fever syndromes and inflammation. Cell Mol Life Sci 2003; 60:1118.
  15. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature 1990; 343:425.
  16. Houten SM, van Woerden CS, Wijburg FA, et al. Carrier frequency of the V377I (1129G>A) MVK mutation, associated with Hyper-IgD and periodic fever syndrome, in the Netherlands. Eur J Hum Genet 2003; 11:196.
  17. Houten SM, Frenkel J, Kuis W, et al. Molecular basis of classical mevalonic aciduria and the hyperimmunoglobulinaemia D and periodic fever syndrome: high frequency of 3 mutations in the mevalonate kinase gene. J Inherit Metab Dis 2000; 23:367.
  18. Cuisset L, Drenth JP, Simon A, et al. Molecular analysis of MVK mutations and enzymatic activity in hyper-IgD and periodic fever syndrome. Eur J Hum Genet 2001; 9:260.
  19. Houten SM, Frenkel J, Rijkers GT, et al. Temperature dependence of mutant mevalonate kinase activity as a pathogenic factor in hyper-IgD and periodic fever syndrome. Hum Mol Genet 2002; 11:3115.
  20. Hager EJ, Tse HM, Piganelli JD, et al. Deletion of a single mevalonate kinase (Mvk) allele yields a murine model of hyper-IgD syndrome. J Inherit Metab Dis 2007; 30:888.
  21. Grose C. Periodic fever in children with hyperimmunoglobulinemia D and mevalonate kinase mutations. Pediatr Infect Dis J 2005; 24:573.
  22. Messer L, Alsaleh G, Georgel P, et al. Homozygosity for the V377I mutation in mevalonate kinase causes distinct clinical phenotypes in two sibs with hyperimmunoglobulinaemia D and periodic fever syndrome (HIDS). RMD Open 2016; 2:e000196.
  23. Hammoudeh M. Hyperimmunoglobulinemia D syndrome in an Arab child. Clin Rheumatol 2005; 24:92.
  24. Frenkel J, Houten SM, Waterham HR, et al. Clinical and molecular variability in childhood periodic fever with hyperimmunoglobulinaemia D. Rheumatology (Oxford) 2001; 40:579.
  25. Stojanov S, Lohse P, Lohse P, et al. Molecular analysis of the MVK and TNFRSF1A genes in patients with a clinical presentation typical of the hyperimmunoglobulinemia D with periodic fever syndrome: a low-penetrance TNFRSF1A variant in a heterozygous MVK carrier possibly influences the phenotype of hyperimmunoglobulinemia D with periodic fever syndrome or vice versa. Arthritis Rheum 2004; 50:1951.
  26. Hoffmann F, Lohse P, Stojanov S, et al. Identification of a novel mevalonate kinase gene mutation in combination with the common MVK V377I substitution and the low-penetrance TNFRSF1A R92Q mutation. Eur J Hum Genet 2005; 13:510.
  27. Obici L, Manno C, Muda AO, et al. First report of systemic reactive (AA) amyloidosis in a patient with the hyperimmunoglobulinemia D with periodic fever syndrome. Arthritis Rheum 2004; 50:2966.
  28. van der Hilst JC, Simon A, Drenth JP. Hereditary periodic fever and reactive amyloidosis. Clin Exp Med 2005; 5:87.
  29. Siewert R, Ferber J, Horstmann RD, et al. Hereditary periodic fever with systemic amyloidosis: is hyper-IgD syndrome really a benign disease? Am J Kidney Dis 2006; 48:e41.
  30. Arkwright PD, McDermott MF, Houten SM, et al. Hyper IgD syndrome (HIDS) associated with in vitro evidence of defective monocyte TNFRSF1A shedding and partial response to TNF receptor blockade with etanercept. Clin Exp Immunol 2002; 130:484.
  31. Drenth JP, Göertz J, Daha MR, van der Meer JW. Immunoglobulin D enhances the release of tumor necrosis factor-alpha, and interleukin-1 beta as well as interleukin-1 receptor antagonist from human mononuclear cells. Immunology 1996; 88:355.
  32. de Wolff JF, Dickinson SJ, Smith AC, et al. Abnormal IgD and IgA1 O-glycosylation in hyperimmunoglobulinaemia D and periodic fever syndrome. Clin Exp Med 2009; 9:291.
  33. Drenth JP, Haagsma CJ, van der Meer JW. Hyperimmunoglobulinemia D and periodic fever syndrome. The clinical spectrum in a series of 50 patients. International Hyper-IgD Study Group. Medicine (Baltimore) 1994; 73:133.
  34. Houten SM, Schneiders MS, Wanders RJ, Waterham HR. Regulation of isoprenoid/cholesterol biosynthesis in cells from mevalonate kinase-deficient patients. J Biol Chem 2003; 278:5736.
  35. Hoffmann GF, Charpentier C, Mayatepek E, et al. Clinical and biochemical phenotype in 11 patients with mevalonic aciduria. Pediatrics 1993; 91:915.
  36. Frenkel J, Rijkers GT, Mandey SH, et al. Lack of isoprenoid products raises ex vivo interleukin-1beta secretion in hyperimmunoglobulinemia D and periodic fever syndrome. Arthritis Rheum 2002; 46:2794.
  37. Simon A, Bijzet J, Voorbij HA, et al. Effect of inflammatory attacks in the classical type hyper-IgD syndrome on immunoglobulin D, cholesterol and parameters of the acute phase response. J Intern Med 2004; 256:247.
  38. Pontillo A, Paoluzzi E, Crovella S. The inhibition of mevalonate pathway induces upregulation of NALP3 expression: new insight in the pathogenesis of mevalonate kinase deficiency. Eur J Hum Genet 2010; 18:844.
  39. De Leo L, Marcuzzi A, Decorti G, et al. Targeting farnesyl-transferase as a novel therapeutic strategy for mevalonate kinase deficiency: in vitro and in vivo approaches. Pharmacol Res 2010; 61:506.
  40. Marcuzzi A, Decorti G, Pontillo A, et al. Decreased cholesterol levels reflect a consumption of anti-inflammatory isoprenoids associated with an impaired control of inflammation in a mouse model of mevalonate kinase deficiency. Inflamm Res 2010; 59:335.
  41. Marcuzzi A, Tommasini A, Crovella S, Pontillo A. Natural isoprenoids inhibit LPS-induced-production of cytokines and nitric oxide in aminobisphosphonate-treated monocytes. Int Immunopharmacol 2010; 10:639.
  42. Marcuzzi A, Crovella S, Pontillo A. Geraniol rescues inflammation in cellular and animal models of mevalonate kinase deficiency. In Vivo 2011; 25:87.
  43. Marcuzzi A, De Leo L, Decorti G, et al. The farnesyltransferase inhibitors tipifarnib and lonafarnib inhibit cytokines secretion in a cellular model of mevalonate kinase deficiency. Pediatr Res 2011; 70:78.
  44. Marcuzzi A, Pontillo A, De Leo L, et al. Natural isoprenoids are able to reduce inflammation in a mouse model of mevalonate kinase deficiency. Pediatr Res 2008; 64:177.
  45. Kuijk LM, Beekman JM, Koster J, et al. HMG-CoA reductase inhibition induces IL-1beta release through Rac1/PI3K/PKB-dependent caspase-1 activation. Blood 2008; 112:3563.
  46. Mandey SH, Schneiders MS, Koster J, Waterham HR. Mutational spectrum and genotype-phenotype correlations in mevalonate kinase deficiency. Hum Mutat 2006; 27:796.
  47. Stoffels M, Simon A. Hyper-IgD syndrome or mevalonate kinase deficiency. Curr Opin Rheumatol 2011; 23:419.
  48. Levy BD, Petasis NA, Serhan CN. Polyisoprenyl phosphates in intracellular signalling. Nature 1997; 389:985.
  49. Mandey SH, Kuijk LM, Frenkel J, Waterham HR. A role for geranylgeranylation in interleukin-1beta secretion. Arthritis Rheum 2006; 54:3690.
  50. Massonnet B, Normand S, Moschitz R, et al. Pharmacological inhibitors of the mevalonate pathway activate pro-IL-1 processing and IL-1 release by human monocytes. Eur Cytokine Netw 2009; 20:112.
  51. Kuijk LM, Mandey SH, Schellens I, et al. Statin synergizes with LPS to induce IL-1beta release by THP-1 cells through activation of caspase-1. Mol Immunol 2008; 45:2158.
  52. Normand S, Massonnet B, Delwail A, et al. Specific increase in caspase-1 activity and secretion of IL-1 family cytokines: a putative link between mevalonate kinase deficiency and inflammation. Eur Cytokine Netw 2009; 20:101.
  53. Drenth JP, van Deuren M, van der Ven-Jongekrijg J, et al. Cytokine activation during attacks of the hyperimmunoglobulinemia D and periodic fever syndrome. Blood 1995; 85:3586.
  54. Havenaar EC, Drenth JP, van Ommen EC, et al. Elevated serum level and altered glycosylation of alpha 1-acid glycoprotein in hyperimmunoglobulinemia D and periodic fever syndrome: evidence for persistent inflammation. Clin Immunol Immunopathol 1995; 76:279.
  55. Drenth JP, Powell RJ, Brown NS, Van der Meer JW. Interferon-gamma and urine neopterin in attacks of the hyperimmunoglobulinaemia D and periodic fever syndrome. Eur J Clin Invest 1995; 25:683.
Topic 5956 Version 16.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟