Rev Esp Endocrinol Pediatr

No Spanish version available for this Article
Rev Esp Endocrinol Pediatr 2022;13 Suppl(2):59-68 | Doi. 10.3266/RevEspEndocrinolPediatr.pre2022.Apr.742
Genetic disorders of primary adrenal insufficiency beyond CAH
Trastornos genéticos de la insuficiencia suprarrenal primaria más allá de la HSC

Sent for review: 25 Apr. 2022 | Accepted: 25 Apr. 2022  | Published: 5 May. 2022
Christa E. Flück
Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland. Department of Biomedical Research, University of Bern, Switzerland.
Correspondence:Christa E. Flück, Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland, Department of Biomedical Research, University of Bern, Switzerland
Table 1 - Table 1. Reported monogenetic causes of primary adrenal insufficiency and associated phenotype (2).
Figure 1
Abstract

Primary adrenal insufficiency (PAI) is caused by disorders of the adrenal cortex that lead to cortisol deficiency. Underlying genetic defects may manifest with an adrenal phenotype only or affect other organ systems. Genetic causes of PAI may be sub-grouped into disorders of steroidogenesis (mainly congenital adrenal hyperplasia), adrenal dysgenesis, familial glucocorticoid deficiency (FGD), and metabolic and autoimmune disorders. Current genetic work-up of PAI yields a specific diagnosis in up to 80% of cases. The latest gene defects discovered in syndromic cases of PAI include variants of CDKN1C and POLE1 in IMAGe syndrome, SAMD9 in MIRAGE syndrome and variants of SPGL1 in a new sphingolipidosis. The latest gene defects manifesting as FGD were found in the mitochondrial gene network regulating reactive oxygen species (NNT, TXNRD2). Knowing the exact genetic diagnosis in a patient with PAI is important to provide optimal care and for genetic counseling. It forms the basis for future gene therapeutic options and cell replacement strategies.

Introduction

Primary adrenal insufficiency (PAI) is defined by cortisol deficiency in humans due to disorders of the adrenal cortex affecting its production. Cortisol is produced in the zona fasciculata (zF, middle zone) of the adrenal cortex. This glucocorticosteroid hormone is responsible for the acute and chronic stress response and thus the regulation and maintenance of the energy homeostasis of the human body. Other steroid hormones produced by the adrenal cortex are mineralocorticoids in the zona glomerulosa (zG) for maintaining water and electrolyte balance, and adrenal androgens in the zona reticularis (zR), which contribute to the sex steroid pool (1). Cortisol production of the adrenal cortex is controlled by the hypothalamic-pituitary-adrenal (HPA) axis. Essential players comprised in this axis are the hormones corticotropin releasing hormone (CRH), adrenocorticotropic hormone ACTH, the ACTH receptor (MC2R), as well as the GPCR/cAMP/MAPK signaling pathway and cortisol. Cortisol then acts predominantly on the glucocorticoid (GC) receptor NR3C1 to exert its action on multiple biological processes and organs. The HPA axis also co-regulates adrenal androgen production, while mineralocorticoid production (mainly aldosterone) is controlled by the renin-angiotensin-aldosterone system.

Genetic variants causing PAI either disrupt cortisol production and steroidogenesis only leading to isolated PAI or they cause additional organ malfunctions as part of a syndrome in syndromic PAI. In isolated PAI, genetic disorders may affect the structure and function of the adrenal cortex or adrenal steroidogenesis specifically, but often also lead to overall disturbances of steroidogenesis affecting other steroid organs (mainly the gonads or the placenta). This can then result in different disorders of sex development (DSD), as steroid hormone biosynthesis, regulation and metabolism rely on a common gene network. Nevertheless, cell- and tissue-specific expression and regulation of steroidogenic genes leads to organ-specific steroid production. Genetic variants in core genes of steroid hormone biosynthesis may therefore be recognized by characteristic clinical phenotypes and changes in steroid profiles assessed in biosamples such as plasma and urine. However, considerable overlap exists so that a characteristic clinical and biochemical profile may be caused by variants in more than one gene, and different variants in one gene may manifest as phenotypically variable.

Although syndromic forms of PAI seem easier to recognize through their broader range of typically involved organ systems, this remains just a theory in many cases. Syndromes with PAI can manifest similar, oligosymptomatic or atypical mainly because the typical spectrum may only develop over time or simply because the phenotype is only recognized when searched for.

However, disorders leading to PAI may not only be grouped according to whether they affect just the adrenal structure and function or lead to defects in other organ systems. They may also be characterized by their suggested molecular disease mechanism. Table 1 gives an overview of all monogenetic disorders causing PAI reported in the literature to date.

PAI often manifests (very) early in life or even goes undiagnosed when embryonic or neonatal lethal, although late-onset manifestation in adulthood is also seen. Clinical signs of adrenal insufficiency are non-specific, but with severe stress such as major illness, trauma or surgery an acute adrenal crisis may be triggered. Signs and symptoms of an acute crisis include abdominal pain, fever, hypoglycemic seizures, weakness, apathy, nausea, vomiting, anorexia, hyponatremia, hypochloremic acidemia, hyperkalemia, hypotension, shock, cardiovascular collapse, and sudden death.

Milestones in medicine concerning PAI were the first clinical description of a patient with PAI by Thomas Addison in 1849, the first description of a patient with congenital adrenal hyperplasia (CAH) by Luigi De Crecchio in 1865, the isolation of cortisol from adrenal extracts by Edward Calvin Kendall and its synthesis for medical use by Lewis H. Sarett in 1946, and the identification of the first genes causing CAH (CYP21A2) by Perrin White in 1984 and familial glucocorticoid deficiency (MC2R) by Adrian J.L. Clark and Constantine Tsigos in 1993.

Several comprehensive reviews have recently been published on the topic of genetic disorders of the adrenal cortex and steroidogenesis (2-11). This update summarizes the genetic causes of PAI with an emphasis on the most recent findings and a perspective on current diagnostic yield and future therapeutic options.

Update on more recent findings on the topic of monogenetic disorders causing PAI

In the group of steroid biosynthetic defects (Table 1), we have learned from affected persons that autosomal recessive gene variants coding for enzymes and cofactors involved in adrenal steroidogenesis (and beyond) may manifest with variable phenotype. This has long been known for variants in CYP21A2, where loss of function mutations cause classic CAH and milder enzyme defects lead to non-classic, late-onset CAH (10). Likewise, less severe autosomal recessive genetic variants in STAR (12) and CYP11A1 (13) have been identified in patients with an adrenal phenotype only mimicking familial GC deficiency (FGD) (14) and not typical lipoid CAH, in which adrenal and gonadal steroidogenesis is affected leading to PAI and 46,XY DSD (15). Similarly, different variants in P450 oxidoreductase can manifest with a vast range of phenotypes ranging from the severest Antley Bixler skeletal and genital malformation syndrome to moderate forms with a CAH phenotype, and to mild forms overlapping with polycystic ovary syndrome in females or lyase deficiency syndrome in males (16-18).

In the group of structural developmental defects of the adrenal cortex, X-linked adrenal hypoplasia congenita due to pathogenic variants in the DAX1/NROB1 gene remains the most frequently identified genetic defect in males with PAI (8). By contrast, genetic variants in SF1/NR5A1 are rarely identified in PAI, but they typically manifest with a broad range of DSDs in both sexes (19). Conversely, very rare syndromic forms of PAI affecting adrenal development and function and affecting fetal growth have been identified more recently through the efforts of next-generation genetic work-up. These include the IMAGe and MIRAGE syndromes (Table 1).

In IMAGe syndrome specific heterozygous gain-of-function mutations in the CDKN1C (cyclin-dependent kinase inhibitor 1C) gene were found in patients manifesting with intrauterine growth retardation, metaphyseal dysplasia, adrenal hypoplasia congenita, and genital anomalies (20). In addition, biallelic POLE mutations causing DNA polymerase epsilon deficiency with a variable degree of immunodeficiency were discovered in another 15 patients of 12 families with IMAGe syndrome (21). POLe is one of the leading polymerases for DNA replication essential for the correct transmission of genetic information. To date, all CDKN1C mutations identified in IMAGe syndrome cluster in the proliferating cell nuclear antigen (PCNA) binding domain. At the initiation of replication, PCNA loads with POLe, and thus the phenotypic overlap of CDKN1C with POLE-associated IMAGe syndrome suggests a mechanistic link (21). Together with MCM4 mutations, CDKN1C and POLE mutations share a common profile of PAI including growth restriction and immunodeficiency due to impaired replisome function (21, 22).

MIRAGE syndrome is characterized by myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, enteropathy and early death. In patients with MIRAGE syndrome, SAMD9 (sterile alpha motif domain-containing protein 9) mutations have been discovered (23, 24). SAMD9 is involved in endosome fusion and is reported to play a role in growth factor signaling transduction. Thus, heterozygote SAMD9 mutations seem to enhance its intrinsic endosome-fusing activity and may thereby lead to abnormal tissue development including dysgenetic and hypoplastic adrenal glands, ovaries and thymus, and result in deleterious overall growth-restricting effects and short survival. In some MIRAGE patients, longer survival seems possible through genetic escape mechanisms such as somatic mutations and progressive monosomy resulting in loss of mutated SAMD9 effects in the bone marrow (23). However, this may come at the cost of increased risk for myelodysplastic syndromes in the long run. Interestingly, SAMD9 mutations were recently also found in a severely undervirilized 46,XY DSD child born SGA without PAI, but otherwise typical features of MIRAGE syndrome (25). And another recent report of two unrelated patients carrying novel and known SAMD9 mutations expanded the typical phenotypic spectrum by including PAI by CNS anomalies and global developmental delay, dysautonomy, hearing loss and chronic lung disease (26).

Overall, it is remarkable how few variants in genes that are critically involved in adrenal development, as revealed by basic research studies, have been identified in humans with dysgenetic, syndromic PAI (27). One of these genes is WNT4. But so far a homozygous mutation in WNT4 has been identified in only one family with the SERKAL syndrome (female sex reversal and dysgenesis of kidneys, adrenals, and lungs) (28). It is therefore very likely that pathogenic variants in genes essential for adrenal development and beyond are embryonic lethal.

In the early era of genetics, first two genes causing familial glucocorticoid deficiency (FGD) were identified, namely mutations in the ACTH receptor (MC2R) and its accessory protein (MRAP) (29, 30). In addition, the syndromic variant of FGD, triple A or Allgrove syndrome, was solved genetically by identifying mutations in the AAAS gene (31). AAAS is a nucleoporin component thought to be involved in cellular stress response (32). However, mutations in the aforementioned genes were identified in less than a third of patients with FGD. More recently, newer genetic methods discovered a novel group of genes located in the mitochondria responsible for oxidative stress homeostasis (33). Very quickly, many mutations in the nicotinamide nucleotide transhydrogenase (NNT) gene of the energy transfer system of the respiratory chain have been found in a fairly large number of unsolved patients with non-syndromic FGD (34, 35). Nnt deficient mice show disorganized adrenal gland zonation, high apoptosis, and diminished steroid production, while NNT knockdown in adrenal cells revealed impaired redox potential through elevated reactive oxygen species and decreased glutathione (GSH) to GSH-disulfide (GSSG) ratio (35). Meanwhile, mutations in other genes associated with the mitochondrial redox system have been found in FGD patients including variants in TXNRD2, GPX1 and PRDX3 (34, 36).

It seems important to be aware that NNT is widely expressed in adrenal, heart, kidney, thyroid, and adipose tissues. Accordingly, the latest reports of older patients with PAI due to NNT mutations describe a broader phenotype with mineralocorticoid deficiency and extra-adrenal anomalies predominantly affecting the gonads (e.g., cryptorchidism, testis adenoma, precocious puberty, and azoospermia) (37). In addition, hypothyroidism, hypertrophic cardiomyopathy and insulin-dependent diabetes mellitus have been reported (37, 38). Phenotypical variability was seen with the same NNT mutations and within families informing that careful follow-up of each individual is warranted.

Several metabolic disorders can lead to PAI (Table 1). Most of them manifest with other characteristic anomalies. However, the X-linked form of adrenoleukodystrophy (X-ALD) due to ABCD1 gene mutations can sometimes present first with an adrenal-only phenotype. The latest syndromic form of PAI due to a metabolic disorder is sphingosine-1-phosphate lyase (SGPL1) deficiency (39-41). It manifests typically as a syndrome comprising steroid-resistant nephrotic syndrome and PAI, optionally associated with ichthyosis, primary hypothyroidism, cryptorchidism, immunodeficiency, and neurological anomalies. This novel sphingolipidosis results from impaired breakdown of sphingosine 1-phosphate (S1P). S1P regulates cell migration, differentiation, survival as well as angiogenesis and development. Identified human SGPL1 mutations were shown to behave as recessive loss-of-function mutations affecting protein expression and localization, enzyme activity, and thus degradation of long-chain sphingoids (40). The pathomechanism of PAI in SGPL1 deficiency includes both compromised adrenal development as well as disrupted steroidogenesis (41). Recent reports of patients carrying SGPL1 mutations have broadened the clinical spectrum when finding individuals without PAI, without renal manifestations or without a central nervous phenotype (42, 43). Long-term follow-up might be necessary to understand the whole spectrum of disease. Of 31 patients with reported SGPL1 mutations, 27 and 26 had PAI and steroid-resistant nephrotic syndrome, respectively (80%), most of them had skin anomalies and immunodeficiency (80%), and 60% had a neurological phenotype (39-41, 44, 45).

Role of the underlying genetic diagnosis for treatment and outcome of PAI

Phenotype-genotype correlation in PAI can be difficult in both syndromic and non-syndromic forms. Generally, the clinical and biochemical phenotype of PAI and family history (including consanguinity and ancestral background) may be hinting at the underlying genetic defect. Comprehensive biochemical testing may be specifically informative for steroidogenesis disorders or metabolic diseases (e.g., X-ALD). Thus, the neonatal screening program includes testing for elevated 17-hydroxyprogesterone as a marker for 21-hydroxylase deficiency CAH (CYP21A2) in many countries (10). More recently, screening for X-ALD has been recommended in the USA to allow early diagnosis for considering hematopoietic stem cell transplantation as a treatment option (46). This screening in dried blood spots is based on the elevation of a lysophosphatidylcholine derivative C26:0-LPC of a very-long-chain fatty acid marker. However, even in these disorders a specific diagnosis at the molecular genetic level is advised to provide detailed information for personalized patient care and family counseling.

Next-generation sequencing (NGS) methods, including whole exome (WES) and genome sequencing (WGS) and hybridization techniques have revolutionized the diagnostic yield for genetic disorders over the past decade. While the gene candidate approach for defining CYP21A2 mutations is still valid in clinical routine, gene panels comprising a selected group of genes are nowadays used for the genetic work-up of PAI offering a molecular genetic diagnosis in 60-80% of cases (34, 47, 48). For the remaining cases, TRIO WES or WGS are used to search for novel genetic defects. NGS may also reveal thus far missed genetic hits in genes known to cause PAI. For instance, predicted benign synonymous variants in the CYP11A1 gene (Thr330= and Ser391=) were recently reported to cause PAI in 19 individuals of 13 families when occurring in a compound heterozygous state with a rare disruptive mutation (49); corresponding mutations were then identified in a large number of European people with PAI.

Current and future therapeutic aspects and perspectives

Lack of corticosteroids will lead to life-threatening conditions in the short or long run. Treatment of all forms of PAI consists in immediate replacement of steroid hormones including glucocorticoids and mineralocorticoids as needed (1). This can prevent affected individuals from suffering a deadly adrenal crisis. But hormonal replacement treatment is not trivial with current drugs bearing a considerable risk for adverse effects due to over- and undertreatment. Sex hormone replacement may be added, if gonadal steroidogenesis is affected. In addition, adrenal androgen excess as a consequence of negative feedback overstimulation through the HPA axis in CAH (mainly due to CYP21A2 deficiency) is often difficult to control with GC replacement only, and may therefore require additional anti-androgenic treatments, especially in females (10, 11).

Overall, there is a need for improved therapy of PAI including newer GC formulations with near-physiological, pharmacokinetic properties, and novel adrenal regenerative strategies (9-11, 50). While newer drugs and treatment modalities mimicking the circadian rhythm of physiologic cortisol production are well on their way into routine clinical care, regenerative approaches are still in the research phase. Treatment or even a cure for some monogenic forms of PAI might be feasible in the future through gene therapeutic options, but they will only become possible when the underlying genetic defect is solved. In the research setting, enzyme replacement therapy using adenoviral gene carrier vectors has been successful in transiently restoring 21-hydroxylase activity in CYP21A2 deficient mice, and may be used in humans in the near future (51). On the other hand, permanent correction of pathogenic variants would be desirable. Gene therapy directed at patients’ own stem cells could theoretically cure steroid disorders. Early studies aimed at achieving cell-based therapies have shown promising results (50), when patient-derived mesenchymal cells were reprogrammed to steroidogenic cells with functional activity (52). The additional use of gene-editing technology may be an option for a future cure for the disease.

Conclusion

Genetic studies conducted to understand adrenal cortex development and function in health and disease are fundamental to improve diagnostic and therapeutic options for individuals with PAI, and to offer informed counseling for affected families. To date, comprehensive genetic work-up of individuals with PAI yields a diagnosis in up to 80%. This may not only have short-term consequences, but also informs on possible long-term effects that might be prevented.

Conflicts of interest

Authors declare no potential Conflicts of Interest.

References

1. Miller WL, Fluck CE, Breault DT, Feldman BJ. Adrenal cortex and its disorders. In: Sperling MA, editor. Sperling - Pediatric Endocrinology: Elsevier; 2020.

2. Fluck CE. MECHANISMS IN ENDOCRINOLOGY: Update on pathogenesis of primary adrenal insufficiency: beyond steroid enzyme deficiency and autoimmune adrenal destruction. Eur J Endocrinol. [Review]. 2017 Sep;177(3):R99-R111.

3. Guran T. Latest Insights on the Etiology and Management of Primary Adrenal Insufficiency in Children. J Clin Res Pediatr Endocrinol. [Review]. 2017 Dec 30;9(Suppl 2):9-22.

4. Hannah-Shmouni F, Stratakis CA. An overview of inborn errors of metabolism manifesting with primary adrenal insufficiency. Rev Endocr Metab Disord. [Research Support, N.I.H., Intramural Review]. 2018 Mar;19(1):53-67.

5. Maharaj A, Maudhoo A, Chan LF, Novoselova T, Prasad R, Metherell LA, et al. Isolated glucocorticoid deficiency: Genetic causes and animal models. J Steroid Biochem Mol Biol. [Review]. 2019 May;189:73-80.

6. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol. [Review]. 2018 Sep;179(3):R125-R41.

7. Roucher-Boulez F, Mallet-Motak D, Tardy-Guidollet V, Menassa R, Goursaud C, Plotton I, et al. News about the genetics of congenital primary adrenal insufficiency. Ann Endocrinol (Paris). [Review]. 2018 Jun;79(3):174-81.

8. Buonocore F, Achermann JC. Primary adrenal insufficiency: New genetic causes and their long-term consequences. Clin Endocrinol (Oxf). [Research Support, Non-U.S. Gov't [Review]. 2020 Jan;92(1):11-20.

9. Husebye ES, Pearce SH, Krone NP, Kampe O. Adrenal insufficiency. Lancet. [Review]. 2021 Feb 13;397(10274):613-29.

10. Claahsen-van der Grinten HL, Speiser PW, Ahmed SF, Arlt W, Auchus RJ, Falhammar H, et al. Congenital Adrenal Hyperplasia-Current Insights in Pathophysiology, Diagnostics, and Management. Endocrine reviews. 2022 Jan 12;43(1):91-159.

11. Merke DP, Auchus RJ. Congenital Adrenal Hyperplasia Due to 21-Hydroxylase Deficiency. The New England journal of medicine. [Review]. 2020 Sep 24;383(13):1248-61.

12. Baker BY, Lin L, Kim CJ, Raza J, Smith CP, Miller WL, et al. Nonclassic congenital lipoid adrenal hyperplasia: a new disorder of the steroidogenic acute regulatory protein with very late presentation and normal male genitalia. J Clin Endocrinol Metab. 2006 Dec;91(12):4781-5.

13. Sahakitrungruang T, Tee MK, Blackett PR, Miller WL. Partial defect in the cholesterol side-chain cleavage enzyme P450scc (CYP11A1) resembling nonclassic congenital lipoid adrenal hyperplasia. J Clin Endocrinol Metab. 2011 Mar;96(3):792-8.

14. Metherell LA, Naville D, Halaby G, Begeot M, Huebner A, Nurnberg G, et al. Nonclassic lipoid congenital adrenal hyperplasia masquerading as familial glucocorticoid deficiency. J Clin Endocrinol Metab. 2009 Oct;94(10):3865-7381.

15. Bose HS, Sugawara T, Strauss JF, 3rd, Miller WL. The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. N Engl J Med. 1996 Dec 19;335(25):1870-8.

16. Fluck CE, Tajima T, Pandey AV, Arlt W, Okuhara K, Verge CF, et al. Mutant P450 oxidoreductase causes disordered steroidogenesis with and without Antley-Bixler syndrome. Nat Genet. 2004 Mar;36(3):228-30.

17. Miller WL. The syndrome of 17,20 lyase deficiency. J Clin Endocrinol Metab. 2012 Jan;97(1):59-67.

18. Pandey AV, Fluck CE. NADPH P450 oxidoreductase: structure, function, and pathology of diseases. Pharmacol Ther. [Research Support, Non-U.S. Gov't Review]. 2013 May;138(2):229-54.

19. Camats N, Fernandez-Cancio M, Audi L, Schaller A, Fluck CE. Broad phenotypes in heterozygous NR5A1 46,XY patients with a disorder of sex development: an oligogenic origin? Eur J Hum Genet. [Case Reports [Research Support, Non-U.S. Gov't]. 2018 Sep;26(9):1329-38.

20. Arboleda VA, Lee H, Parnaik R, Fleming A, Banerjee A, Ferraz-de-Souza B, et al. Mutations in the PCNA-binding domain of CDKN1C cause IMAGe syndrome. Nature genetics. [Research Support, N.I.H., Extramural [Research Support, Non-U.S. Gov't]. 2012 May 27;44(7):788-92.

21. Logan CV, Murray JE, Parry DA, Robertson A, Bellelli R, Tarnauskaite Z, et al. DNA Polymerase Epsilon Deficiency Causes IMAGe Syndrome with Variable Immunodeficiency. Am J Hum Genet. [Research Support, N.I.H., Extramural [Research Support, Non-U.S. Gov't]. 2018 Dec 6;103(6):1038-44.

22. Cabrera-Salcedo C, Kumar P, Hwa V, Dauber A. IMAGe and Related Undergrowth Syndromes: The Complex Spectrum of Gain-of-Function CDKN1C Mutations. Pediatr Endocrinol Rev. [Review]. 2017 Mar;14(3):289-97.

23. Buonocore F, Kuhnen P, Suntharalingham JP, Del Valle I, Digweed M, Stachelscheid H, et al. Somatic mutations and progressive monosomy modify SAMD9-related phenotypes in humans. J Clin Invest. 2017 May 1;127(5):1700-13.

24. Narumi S, Amano N, Ishii T, Katsumata N, Muroya K, Adachi M, et al. SAMD9 mutations cause a novel multisystem disorder, MIRAGE syndrome, and are associated with loss of chromosome 7. Nature genetics. 2016 Jul;48(7):792-7.

25. Shima H, Hayashi M, Tachibana T, Oshiro M, Amano N, Ishii T, et al. MIRAGE syndrome is a rare cause of 46,XY DSD born SGA without adrenal insufficiency. PLoS One. [Case Reports Research Support, Non-U.S. Gov't]. 2018;13(11):e0206184.[Pubmed]

26. Jeffries L, Shima H, Ji W, Panisello-Manterola D, McGrath J, Bird LM, et al. A novel SAMD9 mutation causing MIRAGE syndrome: An expansion and review of phenotype, dysmorphology, and natural history. Am J Med Genet A. [Case Reports Research Support, Non-U.S. Gov't]. 2018 Feb;176(2):415-20.

27. Pignatti E, Fluck CE. Adrenal cortex development and related disorders leading to adrenal insufficiency. Molecular and cellular endocrinology. [Research Support, Non-U.S. Gov't]. 2021 May 1;527:111206.

28. Mandel H, Shemer R, Borochowitz ZU, Okopnik M, Knopf C, Indelman M, et al. SERKAL syndrome: an autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. American journal of human genetics. 2008 Jan;82(1):39-47.

29. Clark AJ, Weber A. Adrenocorticotropin insensitivity syndromes. Endocr Rev. 1998 Dec;19(6):828-43.

30. Metherell LA, Chapple JP, Cooray S, David A, Becker C, Ruschendorf F, et al. Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2. Nat Genet. 2005 Feb;37(2):166-70.

31. Handschug K, Sperling S, Yoon SJ, Hennig S, Clark AJ, Huebner A. Triple A syndrome is caused by mutations in AAAS, a new WD-repeat protein gene. Hum Mol Genet. 2001 Feb 1;10(3):283-90.

32.Prasad R, Metherell LA, Clark AJ, Storr HL. Deficiency of ALADIN impairs redox homeostasis in human adrenal cells and inhibits steroidogenesis. Endocrinology. [Research Support, Non-U.S. Gov't]. 2013 Sep;154(9):3209-18.

33. Prasad R, Kowalczyk JC, Meimaridou E, Storr HL, Metherell LA. Oxidative stress and adrenocortical insufficiency. J Endocrinol. [Research Support, Non-U.S. Gov't [Review]. 2014 Jun;221(3):R63-73.

34. Chan LF, Campbell DC, Novoselova TV, Clark AJ, Metherell LA. Whole-Exome Sequencing in the Differential Diagnosis of Primary Adrenal Insufficiency in Children. Front Endocrinol (Lausanne). 2015;6:113.[Pubmed]

35. Meimaridou E, Kowalczyk J, Guasti L, Hughes CR, Wagner F, Frommolt P, et al. Mutations in NNT encoding nicotinamide nucleotide transhydrogenase cause familial glucocorticoid deficiency. Nat Genet. 2012 Jul;44(7):740-2.

36. Prasad R, Chan LF, Hughes CR, Kaski JP, Kowalczyk JC, Savage MO, et al. Thioredoxin Reductase 2 (TXNRD2) mutation associated with familial glucocorticoid deficiency (FGD). J Clin Endocrinol Metab. [Research Support, Non-U.S. Gov't]. 2014 Aug;99(8):E1556-63.

37. Roucher-Boulez F, Mallet-Motak D, Samara-Boustani D, Jilani H, Ladjouze A, Souchon PF, et al. NNT mutations: a cause of primary adrenal insufficiency, oxidative stress and extra-adrenal defects. Eur J Endocrinol. 2016 Jul;175(1):73-84.

38. Scott R, Van Vliet G, Deladoey J. Association of adrenal insufficiency with insulin-dependent diabetes mellitus in a patient with inactivating mutations in nicotinamide nucleotide transhydrogenase: a phenocopy of the animal model. Eur J Endocrinol. [Case Reports]. 2017 Mar;176(3):C1-C2.

39. Janecke AR, Xu R, Steichen-Gersdorf E, Waldegger S, Entenmann A, Giner T, et al. Deficiency of the sphingosine-1-phosphate lyase SGPL1 is associated with congenital nephrotic syndrome and congenital adrenal calcifications. Hum Mutat. [Research Support, Non-U.S. Gov't [Research Support, N.I.H., Extramural]. 2017 Apr;38(4):365-72.

40. Lovric S, Goncalves S, Gee HY, Oskouian B, Srinivas H, Choi WI, et al. Mutations in sphingosine-1-phosphase lyase cause nephrosis with ichthyosis and adrenal insufficiency. J Clin Invest. 2017 Feb 06.

41. Prasad R, Hadjidemetriou I, Maharaj A, Meimaridou E, Buonocore F, Saleem M, et al. Sphingosine-1-phosphate lyase mutations cause primary adrenal insufficiency and steroid-resistant nephrotic syndrome. J Clin Invest. 2017 Feb 06.

42. Atkinson D, Nikodinovic Glumac J, Asselbergh B, Ermanoska B, Blocquel D, Steiner R, et al. Sphingosine 1-phosphate lyase deficiency causes Charcot-Marie-Tooth neuropathy. Neurology. [Case Reports]. 2017 Feb 7;88(6):533-42.

43. Settas N, Persky R, Faucz FR, Sheanon N, Voutetakis A, Lodish M, et al. SGPL1 Deficiency: A Rare Cause of Primary Adrenal Insufficiency. J Clin Endocrinol Metab. 2019 May 1;104(5):1484-90.

44. Bamborschke D, Pergande M, Becker K, Koerber F, Dotsch J, Vierzig A, et al. A novel mutation in sphingosine-1-phosphate lyase causing congenital brain malformation. Brain Dev. [Case Reports]. 2018 Jun;40(6):480-3.

45. Linhares ND, Arantes RR, Araujo SA, Pena SDJ. Nephrotic syndrome and adrenal insufficiency caused by a variant in SGPL1. Clin Kidney J. 2018 Aug;11(4):462-7.

46. Kemper AR, Brosco J, Comeau AM, Green NS, Grosse SD, Jones E, et al. Newborn screening for X-linked adrenoleukodystrophy: evidence summary and advisory committee recommendation. Genet Med. 2017 Jan;19(1):121-6.

47. Amano N, Narumi S, Hayashi M, Takagi M, Imai K, Nakamura T, et al. Genetic defects in pediatric-onset adrenal insufficiency in Japan. Eur J Endocrinol. 2017 Aug;177(2):187-94.

48. Guran T, Buonocore F, Saka N, Ozbek MN, Aycan Z, Bereket A, et al. Rare Causes of Primary Adrenal Insufficiency: Genetic and Clinical Characterization of a Large Nationwide Cohort. J Clin Endocrinol Metab. [Research Support, Non-U.S. Gov't]. 2016 Jan;101(1):284-92.

49. Maharaj A, Buonocore F, Meimaridou E, Ruiz-Babot G, Guasti L, Peng HM, et al. Predicted Benign and Synonymous Variants in CYP11A1 Cause Primary Adrenal Insufficiency Through Missplicing. J Endocr Soc. 2019 Jan 1;3(1):201-21.

50. Bornstein SR, Malyukov M, Heller C, Ziegler CG, Ruiz-Babot G, Schedl A, et al. New Horizons: Novel Adrenal Regenerative Therapies. J Clin Endocrinol Metab. [Research Support, Non-U.S. Gov't]. 2020 Sep 1;105(9).

51. Perdomini M, Dos Santos C, Goumeaux C, Blouin V, Bougneres P. An AAVrh10-CAG-CYP21-HA vector allows persistent correction of 21-hydroxylase deficiency in a Cyp21(-/-) mouse model. Gene Ther. [Research Support, Non-U.S. Gov't]. 2017 May;24(5):275-81.

52. Ruiz-Babot G, Balyura M, Hadjidemetriou I, Ajodha SJ, Taylor DR, Ghataore L, et al. Modeling Congenital Adrenal Hyperplasia and Testing Interventions for Adrenal Insufficiency Using Donor-Specific Reprogrammed Cells. Cell Rep. [Research Support, Non-U.S. Gov't]. 2018 Jan 30;22(5):1236-49.

53. Eriksson D, Royrvik EC, Aranda-Guillen M, Berger AH, Landegren N, Artaza H, et al. GWAS for autoimmune Addison's disease identifies multiple risk loci and highlights AIRE in disease susceptibility. Nat Commun. [Research Support, Non-U.S. Gov't]. 2021 Feb 11;12(1):959.



Comments
Name*: Surname*:
E-mail*:
Hospital*:
Address:
C.P.: Country:
Comments*:
(450 Words)
Security code*:
* Required Fields
Send
Send Send Send
Send