Next Article in Journal
Quantification of Anterior Chamber Particles Using Anterior Segment Optical Coherence Tomography in Angle-Closure Glaucoma Patients after Laser Iridotomy
Next Article in Special Issue
Colocalization of IgG and IgA Heavy Chains with Kappa and Lambda Light Chains in Glomerular Deposits of IgA Nephropathy Patients Using High-Resolution Confocal Microscopy and Correlation with Oxford MEST-C Scores
Previous Article in Journal
Characteristics of Computed Tomography for Identifying Patients at High Risk of Endogenous Endophthalmitis Due to Klebsiella pneumoniae-Related Pyogenic Liver Abscess
Previous Article in Special Issue
Crescents and IgA Nephropathy: A Delicate Marriage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue: New Insights into the Pathogenesis and Therapies of IgA Nephropathy

1
Department of Nephrology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan
2
Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(15), 4378; https://doi.org/10.3390/jcm11154378
Submission received: 19 July 2022 / Accepted: 24 July 2022 / Published: 28 July 2022
(This article belongs to the Special Issue New Insights into the Pathogenesis and Therapies of IgA Nephropathy)
IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis worldwide [1]. Up to 40% of IgAN patients progress to kidney failure within 20 years following diagnosis [2]. Moreover, life expectancy is reduced by a decade in patients with IgAN [3]. IgA vasculitis with nephritis (IgAVN), formerly known as Henoch–Schönlein purpura nephritis, is a systemic form of vasculitis with renal manifestations similar to IgAN and with variable clinical outcome [4]. Thus, IgAN and IgAVN are significant public health problems.
The pathologic assessment of a renal biopsy specimen is the current “gold standard” for diagnosis of IgAN (for review, see [5,6,7,8,9]). Routine immunofluorescence reveals presence of glomerular immunodeposits consisting of IgA, with variable IgG and/or IgM. Complement C3 is present in most cases, but C1q is absent. Light microscopy provides an assessment of disease severity and prognosis based on the Oxford classification system. Its expanded version, termed MEST-C score, provides an evaluation of five different features: Mesangial cellularity, Endothelial hypercellularity, Segmental sclerosis, Tubular atrophy/interstitial fibrosis, and Crescents. IgAVN presents, in addition to kidney vasculitis features, with a greater frequency of severe lesions, such as glomerular necrosis and crescents. However, the pathologic findings for patients with IgAN as well as IgAVN may be impacted by the time between disease onset, diagnostic renal biopsy, and prior medications. Thus, a renal biopsy provides a snap shot in time since the inherent risks associated with renal biopsy hinder the use of a repeat biopsy. Minimally invasive approaches, such as those based on liquid biopsy biomarkers (e.g., blood, urine), are needed for monitoring disease progression, responses to treatments, and the identification of patients at risk of disease progression who may benefit from participation in new clinical trials.
In the last two decades, extensive studies, using genetic, cellular, biochemical, and immunologic approaches, have enabled the formulation of a multi-hit pathogenesis model for IgAN [10], which has been expanded to IgAVN [11]. This hypothesis postulates that glomerular immunodeposits originate from circulating immune complexes consisting of aberrantly glycosylated IgA1 bound by IgG autoantibodies. This IgA1 has some of the 3 to 6 clustered hinge-region O-glycans deficient in galactose (galactose-deficient IgA1). These IgA1-IgG circulating immune complexes have been found in patients with IgAN as well as IgAVN. Moreover, glomerular immunodeposits of patients with IgAN are enriched in galactose-deficient IgA1 glycoforms and the corresponding IgG autoantibodies [12,13,14]. Furthermore, the elevated serum levels of galactose-deficient IgA1 and corresponding IgG autoantibodies predict disease progression. The pathogenic role of immune complexes consisting of galactose-deficient IgA1 and IgG autoantibodies was recently confirmed in an experimental animal model [15,16]. Moreover, there are genetic elements involved in disease susceptibility and progression that can be associated with this multi-step mechanism [17,18,19,20,21,22,23].
Despite the progress since the disease was initially described in 1968 [24], disease-specific therapy to slow or prevent the progression of kidney injury is still not available. To develop a curative treatment, new strategies for early diagnosis, disease-specific targets, and methods for the assessment of clinical responses in clinical trials need to be identified and developed.
This Special Issue presents a collection of reviews and clinical and experimental studies focused on specific aspects of the current research of the diagnosis, prognosis, disease pathogenesis, determination of disease activity, and emerging strategies for the treatment of IgAN and IgAVN. Of the twelve papers in this Special Issue, two are focused on IgAVN, nine on IgAN, and one makes a comparison between IgAN and IgAVN. Two papers are related to pathology, specifically, the prognostic significance of crescents in IgAN (Trimarchi et al. [25]) and the light microscopic features observed at different times since disease onset in adult patients with IgAVN (Lai et al. [26]). Another paper compares features of IgAN and IgAVN (Pillebout et al. [27]). Two reviews focus on the two key factors in IgAN, the autoantigen (galactose-deficient IgA1) and the corresponding autoantibodies (Ohyama et al. and Knoppova et al. [28,29]), whereas an experimental study postulates that urinary galactose-deficient IgA1 may be a disease-specific biomarker useful for the assessment of disease activity in IgAN (Fukao et al. [30]). Two additional studies consider the role of complement in disease pathogenesis as it relates to therapeutic approaches or the diagnosis and prognosis of disease progression (Poppelaars et al. and Mizerska-Wasiak et al. [31,32]). Another study postulates that obesity may play a role in mesangial lesions in IgAN (Hong et al. [33]). In the last three papers, the authors review the characteristics of various murine models of IgAN (Wehbi et al. [34]) and provide updates on the current clinical trials in IgAN (Cheung et al. [35] and Maixnerova et al. [36]).
Although the papers in this Special Issue do not cover all current activities in the field, we found these publications to be inspirational and informative. We hope that the readers of this Special Issue will reach the same conclusion.

Author Contributions

H.S. and J.N. worked together on the initial draft and its further editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

H.S. and J.N. are co-inventors for a US patent application 14/318,082 (assigned to UAB Research Foundation). J.N. is a co-founder and co-owner of and consultant for Reliant Glycosciences, LLC.

References

  1. Lai, K.N.; Tang, S.C.W.; Schena, F.P.; Novak, J.; Tomino, Y.; Fogo, A.B.; Glassock, R.J. IgA nephropathy. Nat. Rev. Dis. Prim. 2016, 2, 16001. [Google Scholar] [CrossRef]
  2. Wyatt, R.J.; Julian, B.A. IgA nephropathy. N. Engl. J. Med. 2013, 368, 2402–2414. [Google Scholar] [CrossRef] [Green Version]
  3. Hastings, M.C.; Bursac, Z.; Julian, B.A.; Baca, E.V.; Featherston, J.; Woodford, S.Y.; Bailey, L.; Wyatt, R.J. Life expectancy for patients from the southeastern United States with IgA nephropathy. Kidney Int. Rep. 2017, 3, 99–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Davin, J.-C.; Ten Berge, I.J.; Weening, J.J. What is the difference between IgA nephropathy and Henoch-Schönlein purpura nephritis? Kidney Int. 2001, 59, 823–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Clarkson, A.R.; Woodroffe, A.J.; Aarons, I.; Hiki, Y.; Hale, G. IgA nephropathy. Annu. Rev. Med. 1987, 38, 157–168. [Google Scholar] [CrossRef]
  6. Jennette, J.C. The Immunohistology of IgA nephropathy. Am. J. Kidney Dis. 1988, 12, 348–352. [Google Scholar] [CrossRef]
  7. Roberts, I.S.D. Pathology of IgA nephropathy. Nat. Rev. Nephrol. 2014, 10, 445–454. [Google Scholar] [CrossRef] [PubMed]
  8. Rodrigues, J.C.; Haas, M.; Reich, H.N. IgA nephropathy. Clin. J. Am. Soc. Nephrol. 2017, 12, 677–686. [Google Scholar] [CrossRef] [PubMed]
  9. Fogo, A.B.; Lusco, M.A.; Najafian, B.; Alpers, C.E. AJKD atlas of renal pathology: IgA nephropathy. Am. J. Kidney Dis. 2015, 66, e33–e34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Suzuki, H.; Kiryluk, K.; Novak, J.; Moldoveanu, Z.; Herr, A.B.; Renfrow, M.B.; Wyatt, R.J.; Scolari, F.; Mestecky, J.; Gharavi, A.G.; et al. The pathophysiology of IgA nephropathy. J. Am. Soc. Nephrol. 2011, 22, 1795–1803. [Google Scholar] [CrossRef] [Green Version]
  11. Hastings, M.C.; Rizk, D.V.; Kiryluk, K.; Nelson, R.; Zahr, R.S.; Novak, J.; Wyatt, R.J. IgA vasculitis with nephritis: Update of pathogenesis with clinical implications. Pediatr. Nephrol. 2021, 37, 719–733. [Google Scholar] [CrossRef] [PubMed]
  12. Hiki, Y.; Odani, H.; Takahashi, M.; Yasuda, Y.; Nishimoto, A.; Iwase, H.; Shinzato, T.; Kobayashi, Y.; Maeda, K. Mass spectrometry proves under-O-glycosylation of glomerular IgA1 in IgA nephropathy. Kidney Int. 2001, 59, 1077–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Allen, A.C.; Bailey, E.M.; Brenchley, P.E.; Buck, K.S.; Barratt, J.; Feehally, J. Mesangial IgA1 in IgA nephropathy exhibits aberrant O-glycosylation: Observations in three patients. Kidney Int. 2001, 60, 969–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rizk, D.V.; Saha, M.K.; Hall, S.; Novak, L.; Brown, R.; Huang, Z.-Q.; Fatima, H.; Julian, B.A.; Novak, J. Glomerular immunodeposits of patients with IgA nephropathy are enriched for IgG autoantibodies specific for galactose-deficient IgA1. J. Am. Soc. Nephrol. 2019, 30, 2017–2026. [Google Scholar] [CrossRef]
  15. Moldoveanu, Z.; Suzuki, H.; Reily, C.; Satake, K.; Novak, L.; Xu, N.; Huang, Z.-Q.; Knoppova, B.; Khan, A.; Hall, S.; et al. Experimental evidence of pathogenic role of IgG autoantibodies in IgA nephropathy. J. Autoimmun. 2021, 118, 102593. [Google Scholar] [CrossRef] [PubMed]
  16. Makita, Y.; Suzuki, H.; Nakano, D.; Yanagawa, H.; Kano, T.; Novak, J.; Nishiyama, A.; Suzuki, Y. Glomerular deposition of galactose-deficient IgA1-containing immune complexes via glomerular endothelial cell injuries. Nephrol. Dial. Transplant. 2022; Online ahead of print. [Google Scholar] [CrossRef]
  17. Julian, B.A.; Quiggins, P.A.; Thompson, J.S.; Woodford, S.Y.; Gleason, K.; Wyatt, R.J. Familial IgA nephropathy. N. Engl. J. Med. 1985, 312, 202–208. [Google Scholar] [CrossRef]
  18. Gharavi, A.G.; Moldoveanu, Z.; Wyatt, R.; Barker, C.V.; Woodford, S.Y.; Lifton, R.P.; Mestecky, J.; Novak, J.; Julian, B.A. Aberrant IgA1 glycosylation is inherited in familial and sporadic IgA nephropathy. J. Am. Soc. Nephrol. 2008, 19, 1008–1014. [Google Scholar] [CrossRef] [PubMed]
  19. Kiryluk, K.; Li, Y.; Sanna-Cherchi, S.; Rohanizadegan, M.; Suzuki, H.; Eitner, F.; Snyder, H.J.; Choi, M.; Hou, P.; Scolari, F.; et al. Geographic differences in genetic susceptibility to IgA nephropathy: GWAS replication study and geospatial risk analysis. PLoS Genet. 2012, 8, e1002765. [Google Scholar] [CrossRef]
  20. Gharavi, A.G.; Kiryluk, K.; Choi, M.; Li, Y.; Hou, P.; Xie, J.; Sanna-Cherchi, S.; Men, C.J.; Julian, B.A.; Wyatt, R.; et al. Genome-wide association study identifies susceptibility loci for IgA nephropathy. Nat. Genet. 2011, 43, 321–327. [Google Scholar] [CrossRef]
  21. Kiryluk, K.; Li, Y.; Scolari, F.; Sanna-Cherchi, S.; Choi, M.; Verbitsky, M.; Fasel, D.; Lata, S.; Prakash, S.; Shapiro, S.; et al. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat. Genet. 2014, 46, 1187–1196. [Google Scholar] [CrossRef] [PubMed]
  22. Kiryluk, K.; Li, Y.; Moldoveanu, Z.; Suzuki, H.; Reily, C.; Hou, P.; Xie, J.; Mladkova, N.; Prakash, S.; Fischman, C.; et al. GWAS for serum galactose-deficient IgA1 implicates critical genes of the O-glycosylation pathway. PLoS Genet. 2017, 13, e1006609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gale, D.P.; Molyneux, K.; Wimbury, D.; Higgins, P.; Levine, A.P.; Caplin, B.; Ferlin, A.; Yin, P.; Nelson, C.P.; Stanescu, H.; et al. Galactosylation of IgA1 is associated with common variation in C1GALT1. J. Am. Soc. Nephrol. 2017, 28, 2158–2166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Berger, J.; Hinglais, N. Intercapillary deposits of IgA-IgG. J. Urol. Nephrol. 1968, 74, 694–695. [Google Scholar]
  25. Trimarchi, H.; Haas, M.; Coppo, R. Crescents and IgA nephropathy: A delicate marriage. J. Clin. Med. 2022, 11, 3569. [Google Scholar] [CrossRef] [PubMed]
  26. Lai, L.; Liu, S.; Azrad, M.; Hall, S.; Hao, C.; Novak, J.; Julian, B.A.; Novak, L. IgA vasculitis with nephritis in adults: Histological and clinical assessment. J. Clin. Med. 2021, 10, 4851. [Google Scholar] [CrossRef] [PubMed]
  27. Pillebout, E. IgA vasculitis and IgA nephropathy: Same disease? J. Clin. Med. 2021, 10, 2310. [Google Scholar] [CrossRef]
  28. Ohyama, Y.; Renfrow, M.B.; Novak, J.; Takahashi, K. Aberrantly glycosylated IgA1 in IgA nephropathy: What we know and what we don’t know. J. Clin. Med. 2021, 10, 3467. [Google Scholar] [CrossRef]
  29. Knoppova, B.; Reily, C.; King, R.G.; Julian, B.A.; Novak, J.; Green, T.J. Pathogenesis of IgA nephropathy: Current understanding and implications for development of disease-specific treatment. J. Clin. Med. 2021, 10, 4501. [Google Scholar] [CrossRef] [PubMed]
  30. Fukao, Y.; Suzuki, H.; Kim, J.S.; Jeong, K.H.; Makita, Y.; Kano, T.; Nihei, Y.; Nakayama, M.; Lee, M.; Kato, R.; et al. Galactose-deficient IgA1 as a candidate urinary marker of IgA nephropathy. J. Clin. Med. 2022, 11, 3173. [Google Scholar] [CrossRef] [PubMed]
  31. Poppelaars, F.; Faria, B.; Schwaeble, W.; Daha, M.R. The Contribution of complement to the pathogenesis of IgA nephropathy: Are complement-targeted therapies moving from rare disorders to more common diseases? J. Clin. Med. 2021, 10, 4715. [Google Scholar] [CrossRef] [PubMed]
  32. Mizerska-Wasiak, M.; Such-Gruchot, A.; Cichoń-Kawa, K.; Turczyn, A.; Małdyk, J.; Miklaszewska, M.; Drożdż, D.; Firszt-Adamczyk, A.; Stankiewicz, R.; Rybi-Szumińska, A.; et al. The role of complement component C3 activation in the clinical presentation and prognosis of IgA nephropathy—A national study in children. J. Clin. Med. 2021, 10, 4405. [Google Scholar] [CrossRef] [PubMed]
  33. Hong, Y.A.; Min, J.W.; Ha, M.A.; Koh, E.S.; Kim, H.D.; Ban, T.H.; Kim, Y.S.; Kim, Y.K.; Kim, D.; Shin, S.J.; et al. The impact of obesity on the severity of clinicopathologic parameters in patients with IgA nephropathy. J. Clin. Med. 2020, 9, 2824. [Google Scholar] [CrossRef] [PubMed]
  34. Wehbi, B.; Pascal, V.; Zawil, L.; Cogné, M.; Aldigier, J.-C. History of IgA nephropathy mouse models. J. Clin. Med. 2021, 10, 3142. [Google Scholar] [CrossRef] [PubMed]
  35. Cheung, C.K.; Rajasekaran, A.; Barratt, J.; Rizk, D.V. An update on the current state of management and clinical trials for IgA nephropathy. J. Clin. Med. 2021, 10, 2493. [Google Scholar] [CrossRef] [PubMed]
  36. Maixnerova, D.; El Mehdi, D.; Rizk, D.V.; Zhang, H.; Tesar, V. New treatment strategies for IgA nephropathy: Targeting plasma cells as the main source of pathogenic antibodies. J. Clin. Med. 2022, 11, 2810. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Suzuki, H.; Novak, J. Special Issue: New Insights into the Pathogenesis and Therapies of IgA Nephropathy. J. Clin. Med. 2022, 11, 4378. https://doi.org/10.3390/jcm11154378

AMA Style

Suzuki H, Novak J. Special Issue: New Insights into the Pathogenesis and Therapies of IgA Nephropathy. Journal of Clinical Medicine. 2022; 11(15):4378. https://doi.org/10.3390/jcm11154378

Chicago/Turabian Style

Suzuki, Hitoshi, and Jan Novak. 2022. "Special Issue: New Insights into the Pathogenesis and Therapies of IgA Nephropathy" Journal of Clinical Medicine 11, no. 15: 4378. https://doi.org/10.3390/jcm11154378

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop