3,055
Views
2
CrossRef citations to date
0
Altmetric
Collection

Prevalence and natural history of variants in the ANKRD26 gene: a short review and update of reported cases

, , , , ORCID Icon &
Pages 1107-1112 | Received 03 Jan 2022, Accepted 19 Apr 2022, Published online: 19 May 2022

Abstract

ANKRD26 is a highly conserved gene located on chromosome 10p12.1 which has shown to play a role in normal megakaryocyte differentiation. ANKRD26-related thrombocytopenia, or thrombocytopenia 2, is an inherited thrombocytopenia with mild bleeding diathesis resulting from point mutations the 5ʹUTR of the ANKRD26 gene. Point mutations in the 5ʹUTR region have been shown to prevent transcription factor-mediated downregulation of ANKRD26 in normal megakaryocyte differentiation. Patients with ANKRD26-related thrombocytopenia have a predisposition to developing hematological malignancies, with acute myeloid leukemia and myelodysplastic syndrome most commonly described in the literature. We review the clinical features and biological mechanisms of ANKRD26-related thrombocytopenia and summarize known cases in the literature.

Introduction

Inherited thrombocytopenias (IT) are a group of disorders which present with a reduced platelet count but varied functional and morphological platelet characteristics. Some ITs are associated with extra-hematological manifestations such as sensorineural deafness (MYH9-related disease, DIAPH1-related disease) or myopathy (Storkmorken syndrome) whilst others have a predisposition to hematological malignancies such as acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML), and myelodysplastic syndrome (MDS) [Citation1–4]. At least 40 genes and their mutations have been implicated in the development of inherited thrombocytopenia [Citation3,Citation5,Citation6]. Definitive identification of the genetic nature of thrombocytopenia can be important as some forms differ in disease history and prognosis. Molecular diagnosis may impact clinical management, including monitoring for associated hematological malignancies.

ANKRD26-related thrombocytopenia (ANKRD26-RT), also known as thrombocytopenia 2 (THC2) (OMIM #188000), is a non-syndromic autosomal dominant thrombocytopenic disorder [Citation7]. Though first described in a large Italian family, in which 17 individuals were affected and the gene locus on chromosome 10 identified using linkage analysis and candidate mutation screening, localization of pathological variants to the 5’ untranslated region (5ʹUTR) of ANKRD26 was not made until 2011 [Citation8,Citation9]. Since then, multiple causative variants have been shown to be the result of single nucleotide changes in the highly conserved 5’ UTR region of the gene [Citation10,Citation11]. Case reports of variants in the coding region segregating with thrombocytopenia have also been reported [Citation12–14].

Clinical features

Patients with ANKRD26-RT typically have lifelong mild (100–150 x109 cells/L) to moderate (50–99 x109 cells/L) thrombocytopenia, although counts may temporarily normalize in response to infection or inflammation [Citation5,Citation15]. The bleeding phenotype is variable. Most have a normal or mild bleeding phenotype without a history of spontaneous or prolonged surgical bleeding [Citation7,Citation16]. However, some individuals experiencing spontaneous epistaxis, bruising, or menorrhagia have also been reported. Morphologically, platelets appear normal in size and mean platelet volume is usually within normal range. Under light microscopy, platelets appear predominantly normogranular, with occasional hypogranular forms noted in some individuals. By electron microscopy, a reduction in alpha-granules has been described, as well as, increased particulate proteasome-rich cytoplasmic structures, the cause of which is yet to be clarified [Citation16,Citation17]. Platelet aggregation studies are often normal, however, reduced platelet responses to arachidonic acid and epinephrine have been reported. GPIa is commonly reduced when evaluated by flow cytometry and up to a sevenfold increase in serum thrombopoietin levels is seen in some cases [Citation10,Citation11,Citation16]. Dysmegakaryopoiesis with an increase in small and hypolobulated megakaryocytes with reduced cytoplasmic volume is commonly cited in those who have undergone bone marrow biopsies [Citation11,Citation18]. Hemoglobin and white cell counts are generally within the normal range, with inconsistent reports of leukocytosis and erythrocytosis described [Citation10,Citation19].

Predisposition to malignancy

A clinically important feature of ANKRD26-RT reported in the literature is the increased risk of developing hematological malignancy. AML and MDS are the most commonly described, though there are some reports of lymphoid malignancies and a single case report of a patient with a 5ʹUTR mutation developing multiple myeloma [Citation6,Citation10–12,Citation16,Citation20]. An extended case series of 118 subjects with confirmed or highly probable ANKRD26-RT revealed an 8% incidence of myeloid malignancy (AML, MDS, and chronic myeloid leukemia (CML)) [Citation10]. An estimated 24-fold increase in acute leukemia incidence alone is reported compared to the general population in this study, and there have been multiple other case reports describing affected patients or their relatives developing hematological malignancy ().

Table I. Malignancies associated with variants in the 5ʹUTR sequence of ANKRD26.

Differential diagnosis

Diagnosis of ANKRD26-RT may be difficult due to the lack of distinct clinical and laboratory characteristics [Citation16]. In many cases, patients may be misdiagnosed with immune thrombocytopenia (ITP), which should be a diagnosis of exclusion. This is especially the case if there is no obvious family history of thrombocytopenia or if the patient has had fluctuating platelet counts in the past [Citation28,Citation29]. There are also documented cases of patients being incorrectly diagnosed with MDS on the basis of persistently low platelet counts and bone marrow biopsy demonstrating dysmegakaryopoiesis, both of which may be features of ANKRD26-RT [Citation30].

Other ITs to consider which may also present with thrombocytopenia with normal platelet size include RUNX1-related thrombocytopenia (RUNX1-RT), ETV6-related thrombocytopenia (ETV6-RT) and CYCS-related thrombocytopenia (CYCS-RT) [Citation3]. Of these, RUNX1-RT demonstrates a 30–40% lifetime risk of developing MDS/AML and ETV6-RT conferring a 20% lifetime risk of B-ALL with a 30% overall lifetime risk of hematological malignancy [Citation1,Citation5,Citation31–33].

Pathophysiology

Regulation of ANKRD26 expression

ANKRD26 is located on chromosome 10p12.1 and contains 34 exons that result in a number of protein isoforms expressed at low levels in multiple human tissues, including platelets, leukocytes, adrenal glands, prostate, ovary, liver, spleen, and central nervous system [Citation34,Citation35]. ANKRD26 shares regions of homology with the POTE family of genes that are characterized by ankyrin repeats (involved in protein–protein interactions) close to the N-terminal region and a helical region that forms coiled-coil domains similar to that of spectrins, suggesting involvement in signal transmission across the plasma membrane () [Citation36–38]. ANKRD26 is highly conserved between the different species, suggesting an important function [Citation38,Citation39]. Mouse Ankrd26 protein localizes to the cell membrane in cell lines and human ANKRD26 protein is identified in centriolar distal appendages and cilial basal bodies in human cell lines [Citation39–41].

Figure 1. (a) Schematic structure of 5ʹUTR of ANKRD26 mapping single point variants currently identified in the literature. (b) General structure of ANKRD26 protein [Citation36].

Figure 1. (a) Schematic structure of 5ʹUTR of ANKRD26 mapping single point variants currently identified in the literature. (b) General structure of ANKRD26 protein [Citation36].

In healthy subjects, ANKRD26 mRNA expression is high in CD34+ progenitor cells and immature megakaryocytes, and decreases over time, becoming almost undetectable in mature platelets [Citation42]. Predictive software identified two transcription factors that bind the ANKRD26 5’ UTR: runt-related transcription factor 1 (RUNX1) and friend leukemia integration 1 (FLI1). Knockdown of either gene in human megakaryocytes led to increased ANKRD26 mRNA expression, while overexpression of these genes in K562 cell lines led to decreased ANKRD26 mRNA. The effect was synergistic when both genes were overexpressed. Other studies suggest metabolic and/or inflammatory factors may also influence mRNA expression in mouse adipose tissue and human leukocytes through epigenetic alterations [Citation43,Citation44]. Taken together, these lines of evidence suggest expression of ANKRD26 is regulated by interaction of transcription factors with the 5ʹUTR and epigenetic alterations.

Mechanisms contributing to thrombocytopenia

The 5ʹUTR regulatory sequence of ANKRD26 is the most common site of single nucleotide variants identified in ANKRD26-RT. A total of 318 patients reported in the literature have a single point variant in this region (). The most common variants include c.-128 G > A, c.-134 G > A and c.-127A>T, and lie close to RUNX1 and FLI1 binding sites (). These single nucleotide variants in the 5’ UTR alter ANKRD26 transcription by preventing RUNX1/FLI1-mediated repression and result in persistently high levels of ANKRD26 mRNA at all stages of platelet development [Citation42,Citation52]. Electron microscopy of megakaryocyte cell lines from these patients showed slightly lower ploidy, decreased granule concentration, and abnormal proplatelet formation compared to controls.

Table II. A summary of point mutations in the 5’ untranslated region of the ANKRD26 gene as reported in the literature. Mean averages reported where data available and ranges and number of individuals accounted for are in brackets.

Components of the mitogen-activated protein kinase (MAPK) pathways appear to play an important role in this process, because the megakaryocytic changes were associated with increased ERK phosphorylation, and were abrogated by ANKRD26 knockdown or ERK pathway inhibition using a MEK inhibitor [Citation42]. MEK (also known as MKK or MAP2K) inhibition has previously been shown to increase ploidy and proplatelet formation in thrombopoietin-stimulated human megakaryocytes [Citation53]. Therefore, the evidence suggests that persistence of ANKRD26 expression in ANKRD26-RT leads to persistent ERK activation, which may in turn be responsible for, or contribute to, reduced megakaryocyte ploidy, impaired proplatelet formation, and subsequent thrombocytopenia.

At least one other mechanism for increased ANKRD26 expression has been described in a thrombocytopenic family using long-read genomic sequencing. In this pedigree, a complex structural variant resulting in a paired duplication-inversion of part of the ANKRD26 gene was identified that caused a juxtaposition of the promotor of WAC and exons 10–34 of ANKRD26. This gene duplication resulted in high WAC-ANKRD26 mRNA levels and increased ERK phosphorylation similar to the phenotype caused by 5ʹUTR variants [Citation54].

To date, no animal model of ANKRD26-RT has been described. A mouse Ankrd26 knockdown model did not report blood abnormalities but showed mice with hyperphagia, organomegaly, obesity, and reduced expression of ciliary proteins in the brain [Citation40,Citation43,Citation55].

ANKRD26 function in other cellular processes - the centrosome

As suggested by its localization, ANKRD26 appears to play a role in centriole biology [Citation41]. Centrioles are important in ciliogenesis and motility. They are components of centrosomes that have been implicated in cancer pathogenesis [Citation56,Citation57]. Centrosome amplification triggers p53-dependent apoptosis through activation of a multiprotein complex known as the PIDDosome [Citation56]. In centrosome amplification (e.g. cytokinesis failure), ANKRD26 recruits the p53-induced death domain protein 1 (PIDD1) to the centriole distal appendages to form part of the PIDDosome [Citation57–59]. When ANKRD26 is inactivated, cells cannot sustain PIDDosome assembly and show enhanced growth following centrosome amplification [Citation57,Citation59]. Whether these actions of ANKRD26 protein play any role in the pathogenesis of ANKRD26-RT is unknown.

Concluding remarks

ANKRD26-RT is characterized by a relatively nonspecific phenotype of mild to moderate thrombocytopenia with normal platelet size and function. Most individuals lack significant mucocutaneous bleeding symptoms. A concerning association with hematological malignancy has been observed in cohorts with variants in the 5ʹUTR region. However, precise prevalence estimates and strategies to guide clinical monitoring, counseling, and treatment will only be possible through further analysis of large patient cohorts and exploration of the pathophysiological mechanisms underpinning this disorder.

Acknowledgements

The work in the author’s laboratories is supported by the British Heart Foundation (PG/13/36/30275; FS/15/18/31317; PG/16/103/32650; FS/18/11/33443; NVM).

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by the British Heart Foundation [FS/15/18/31317,FS/18/11/33443,PG/13/36/30275,PG/16/103/32650].

References

  • Paola JD, Porter CC. ETV6-related thrombocytopenia and leukemia predisposition. Blood 2019;134(8):663–667. doi:10.1182/blood.2019852418.
  • Morgan NV, Daly ME. Gene of the issue: RUNX1 mutations and inherited bleeding. Platelets 2017;28(2):1–3. doi:10.1080/09537104.2017.1280151.
  • Almazni I, Stapley R, Morgan NV. Inherited thrombocytopenia: update on genes and genetic variants which may be associated with bleeding. Front Cardiovasc Med 2019;6:80. doi:10.3389/fcvm.2019.00080.
  • Palma-Barqueros V, Revilla N, Sánchez A, Cánovas AZ, Rodriguez-Alén A, Marín-Quílez A, González-Porras JR, Vicente V, Lozano ML, Bastida JM, et al., Inherited platelet disorders: an updated overview. Int J Mol Sci 2021;22(9):4521. doi:10.3390/ijms22094521.
  • Galera P, Dulau‐Florea A, Calvo KR. Inherited thrombocytopenia and platelet disorders with germline predisposition to myeloid neoplasia. Int J Lab Hematol 2019;41(S1):131–141. doi:10.1111/ijlh.12999.
  • Martin ES, Ferrer A, Mangaonkar AA, Khan SP, Kohorst MA, Joshi AY, Hogan WJ, Olteanu H, Moyer AM, Al‐Kali A, et al., Spectrum of hematological malignancies, clonal evolution and outcomes in 144 Mayo Clinic patients with germline predisposition syndromes. Am J Hematol 2021;96(11):1450–1460. doi:10.1002/ajh.26321.
  • Botero JP, Dugan SN, Anderson MW. ANKRD26-related thrombocytopenia. In: Adam MP, Ardinger HH, Pagon RA, editors. GeneReviews®. Seattle:University of Washington, Seattle; 2018 Jun 21. p. 1993–2022
  • Savoia A, Vecchio MD, Totaro A, Perrotta S, Amendola G, Moretti A, Zelante L, Iolascon A. An autosomal dominant thrombocytopenia gene maps to chromosomal region 10p. Am J Hum Genetics 1999;65(5):1401–1405. doi:10.1086/302637.
  • Pippucci T, Savoia A, Perrotta S, Pujol-Moix N, Noris P, Castegnaro G, Pecci A, Gnan C, Punzo F, Marconi C, et al., Mutations in the 5′ UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2. Am J Hum Genetics 2011;88(1):115–120. doi:10.1016/j.ajhg.2010.12.006.
  • Noris P, Favier R, Alessi M-C, Geddis A, Kunishima S, Heller P, Giordano P, Niederhoffer KY, Bussel JB, Podda GM, et al., ANKRD26-related thrombocytopenia and myeloid malignancies. Blood 2013;122(11):1987–1989. doi:10.1182/blood-2013-04-499319.
  • Noris P, Perrotta S, Seri M, Pecci A, Gnan C, Giuseppe L, Pujol-Moix N, Zecca M, Scognamiglio F, De Rocco D, et al., Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood 2011;117(24):6673–6680. doi:10.1182/blood-2011-02-336537.
  • Marconi C, Canobbio I, Bozzi V, Pippucci T, Simonetti G, Melazzini F, Angori S, Martinelli G, Saglio G, Torti M, et al., 5’UTR point substitutions and N-terminal truncating mutations of ANKRD26 in acute myeloid leukemia. J Hematol Oncol 2017;10(1):18. doi:10.1186/s13045-016-0382-y.
  • Zhang L, Yu J, Xian Y, Wen X, Guan X, Guo Y, Luo M, Dou Y. Application of high‐throughput sequencing for hereditary thrombocytopenia in southwestern China. J Clin Lab Anal 2021;35(8):e23896. doi:10.1002/jcla.23896.
  • Daama SAA, Housawi YH, Dridi W, Sager M, Otieno FG, Hou C, Vasquez L, Kim C, Tian L, Sleiman P, et al., A missense mutation in ANKRD26 segregates with thrombocytopenia. Blood 2013;122(3):461–462. doi:10.1182/blood-2013-03-489344.
  • Rahman ZA, Miller KC, Jabbour H, Alkhatib Y, Donthireddy V. Outcomes of patients with thrombocytopenia evaluated at hematology subspecialty clinics. Hematol Oncol Stem Cell Ther. 2021;11:2-9. doi: 10.1016/j.hemonc.2021.01.002.
  • Botero JP, Chen D, He R, Viswanatha DS, Majerus JA, Coon LM, Nguyen PL, Reichard KK, Oliveira JL, Tefferi A, Gangat N. Clinical and laboratory characteristics in congenital ANKRD26 mutation-associated thrombocytopenia: a detailed phenotypic study of a family. Platelets 2016;27(7):1–4. doi:10.3109/09537104.2015.1129225.
  • Necchi V, Balduini A, Noris P, Barozzi S, Sommi P, Di Buduo C, Balduini C, Solcia E, Pecci A. Ubiquitin/proteasome-rich particulate cytoplasmic structures (PaCSs) in the platelets and megakaryocytes of ANKRD26-related thrombocytopenia. Thromb Haemostasis 2013;109(2):263–271. doi:10.1160/TH12-07-0497.
  • Tsang HC, Bussel JB, Mathew S, Liu Y-C, Imahiyerobo AA, Orazi A, Geyer JT. Bone marrow morphology and disease progression in congenital thrombocytopenia: a detailed clinicopathologic and genetic study of eight cases. Modern Pathol 2017;30(4):486–498. doi:10.1038/modpathol.2016.218.
  • Zidan NI, AbdElmonem DM, Elsheikh HM, Metwally EA, Mokhtar WA, Osman GM. Relation between mutations in the 5′ UTR of ANKRD26 gene and inherited thrombocytopenia with predisposition to myeloid malignancies. An Egyptian study. Platelets 2020;32(5):1–9. doi:10.1080/09537104.2020.1854544.
  • Husnain M, Wang T, Valdes M, Hoffman J, Lekakis L. Multiple myeloma in a patient with ANKRD26-related thrombocytopenia successfully treated with combination therapy and autologous stem cell transplant. Case Rep Hematol 2019;2019:9357572. doi:10.1155/2019/9357572.
  • Zaninetti C, Melazzini F, Croci GA, Boveri E, Balduini CL. Extramedullary hematopoiesis: a new feature of inherited thrombocytopenias? J Thromb Haemost 2017;15(11):2226–2229. doi:10.1111/jth.13850.
  • Boutroux H, Petit A, Auvrignon A, Lapillonne H, Ballerini P, Favier R, Leverger G. Childhood diagnosis of genetic thrombocytopenia with mutation in the ankyrine repeat domain 26 gene. Eur J Pediatr 2015;174(10):1399–1403. doi:10.1007/s00431-015-2549-x.
  • Botero JP, Oliveira JL, Chen D, Reichard KK, Viswanatha DS, Nguyen PL, Pruthi RK, Majerus J, Gada P, Gangat N, et al., ASXL1 mutated chronic myelomonocytic leukemia in a patient with familial thrombocytopenia secondary to germline mutation in ANKRD26. Blood Cancer J 2015;5(5):e315. doi:10.1038/bcj.2015.41.
  • Ferrari S, Lombardi AM, Putti MC, Bertomoro A, Cortella I, Barzon I, Girolami A, Fabris F. Spectrum of 5’UTR mutations in ANKRD26 gene in patients with inherited thrombocytopenia: c.-140C>G mutation is more frequent than expected. Platelets 2017;28(6):1–4. doi:10.1080/09537104.2016.1267337.
  • Marquez R, Hantel A, Lorenz R, Neistadt B, Wong J, Churpek JE, Mardini NA, Shaukat I, Gurbuxani S, Miller JL, et al., A new family with a germline ANKRD26 mutation and predisposition to myeloid malignancies. Leukemia Lymphoma 2014;55(12):2945–2946. doi:10.3109/10428194.2014.903476.
  • Diep RT, Corey K, Arcasoy MO. A novel nucleotide substitution in the 5′ untranslated region of ANKRD26 gene is associated with inherited thrombocytopenia: a report of two new families. Ann Hematol 2019;98(7):1789–1791. doi:10.1007/s00277-019-03632-y.
  • Lazaro E, Houssin C, Sentilhes L, Blouin L, Fiore M. Successful management of a pregnant woman with severe ANKRD26-related thrombocytopenia and anti-HPA-5b alloimmunization. Platelets 2019;31(6):1–3
  • Pecci A. Diagnosis and treatment of inherited thrombocytopenias. Clin Genet 2016;89(2):141–153. doi:10.1111/cge.12603.
  • Pecci A, Balduini CL. Inherited thrombocytopenias: an updated guide for clinicians. Blood Rev 2020;48:100784. doi:10.1016/j.blre.2020.100784.
  • Zaninetti C, Santini V, Tiniakou M, Barozzi S, Savoia A, Pecci A. Inherited thrombocytopenia caused by ANKRD26 mutations misdiagnosed and treated as myelodysplastic syndrome: report on two cases. J Thromb Haemost 2017;15(12):2388–2392. doi:10.1111/jth.13855.
  • Bellissimo DC, Speck NA. RUNX1 mutations in inherited and sporadic leukemia. Front Cell Dev Biol 2017;5:111. doi:10.3389/fcell.2017.00111.
  • Godley LA, Shimamura A. Genetic predisposition to hematologic malignancies: management and surveillance. Blood 2017;130(4):424–432. doi:10.1182/blood-2017-02-735290.
  • Feurstein S, Godley LA. Germline ETV6 mutations and predisposition to hematological malignancies. Int J Hematol 2017;106(2):189–195. doi:10.1007/s12185-017-2259-4.
  • Kikuno R, Nagase T, Ishikawa K, Hirosawa M, Miyajima N, Tanaka A, Kotani H, Nomura N, Ohara O. Prediction of the coding sequences of unidentified human genes. XIV. The complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro. DNA Res 1999;6(3):197–205. doi:10.1093/dnares/6.3.197.
  • Kim M-S, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R, Jain S, et al., A draft map of the human proteome. Nature 2014;509(7502):575–581. doi:10.1038/nature13302.
  • The Uniprot Consortium UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res 2021;49(D1):D480–9. doi: 10.1093/nar/gkaa1100.
  • Lee Y, Ise T, Ha D, Fleur AS, Hahn Y, Liu X-F, Nagata S, Lee B, Bera TK, Pastan I, et al., Evolution and expression of chimeric POTE–actin genes in the human genome. Proc National Acad Sci 2006;103(47):17885–17890. doi:10.1073/pnas.0608344103.
  • Yoonsoo H, Tapan KB, Ira HP, Byungkook L. Duplication and extensive remodeling shaped POTE family genes encoding proteins containing ankyrin repeat and coiled coil domains. Gene 2006 Feb 1;366(2):238–245. doi:10.1016/j.gene.2005.07.045.
  • Yan H, Chen C, Chen H, Hong H, Huang Y, Ling K, Hu J, Wei Q. TALPID3 and ANKRD26 selectively orchestrate FBF1 localization and cilia gating. Nat Commun 2020;11(1):2196. doi:10.1038/s41467-020-16042-w.
  • Bera TK, Liu X-F, Yamada M, Gavrilova O, Mezey E, Tessarollo L, Anver M, Hahn Y, Lee B, Pastan I, et al., A model for obesity and gigantism due to disruption of the Ankrd26 gene. Proc National Acad Sci 2008;105(1):270–275. doi:10.1073/pnas.0710978105.
  • Bowler M, Kong D, Sun S, Nanjundappa R, Evans L, Farmer V, Holland A, Mahjoub MR, Sui H, Loncarek J, et al., High-resolution characterization of centriole distal appendage morphology and dynamics by correlative STORM and electron microscopy. Nat Commun 2019;10(1):993. doi:10.1038/s41467-018-08216-4.
  • Bluteau D, Balduini A, Balayn N, Currao M, Nurden P, Deswarte C, Leverger G, Noris P, Perrotta S, Solary E, et al., Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation. J Clin Invest 2014;124(2):580–591. doi:10.1172/JCI71861.
  • Raciti GA, Spinelli R, Desiderio A, Longo M, Parrillo L, Nigro C, D’Esposito V, Mirra P, Fiory F, Pilone V, et al., Specific CpG hyper-methylation leads to Ankrd26 gene down-regulation in white adipose tissue of a mouse model of diet-induced obesity. Sci Rep Uk 2017;7(1):43526. doi:10.1038/srep43526.
  • Desiderio A, Longo M, Parrillo L, Campitelli M, Cacace G, de Simone S, Spinelli R, Zatterale F, Cabaro S, Dolce P, et al., Epigenetic silencing of the ANKRD26 gene correlates to the pro-inflammatory profile and increased cardio-metabolic risk factors in human obesity. Clin Epigenetics 2019;11(1):181. doi:10.1186/s13148-019-0768-0.
  • Fournier E, Debord C, Soenen V, Trillot N, Gonzales F, Tintiller V, Terriou L, Derrieux C, Abou Chahla W, Paris C, et al., Baseline dysmegakaryopoiesis in inherited thrombocytopenia/platelet disorder with predisposition to haematological malignancies. Brit J Haematol 2020;189(4):e119–22. doi:10.1111/bjh.16543.
  • Ouchi‐Uchiyama M, Sasahara Y, Kikuchi A, Goi K, Nakane T, Ikeno M, Noguchi Y, Uike N, Miyajima Y, Matsubara K, et al., Analyses of genetic and clinical parameters for screening patients with inherited thrombocytopenia with small or normal‐sized platelets. Pediatr Blood Cancer 2015;62(12):2082–2088. doi:10.1002/pbc.25668.
  • Ventz R, Hundemer M, Witzens-Harig M, Lehmann B, Felbor U, Najm J. Milde Blutungsneigung bei einer 62-Jährigen mit hereditärer Thrombozytopenie. Internist (Berl) 2013;54(6):765–768. doi:10.1007/s00108-013-3284-x.
  • Kewan T, Noss R, Godley LA, Rogers HJ, Carraway HE. Inherited thrombocytopenia caused by germline ANKRD26 mutation should be considered in young patients with suspected myelodysplastic syndrome. J Invest Med High Impact Case Rep 2020;8:2324709620938941. doi:10.1177/2324709620938941.
  • Averina M, Jensvoll H, Strand H, Sovershaev M. A novel ANKRD26 gene variant causing inherited thrombocytopenia in a family of Finnish origin: another brick in the wall? Thromb Res 2017;151:41–43. doi:10.1016/j.thromres.2017.01.001.
  • Tan C, Dai L, Chen Z, Yang W, Wang Y, Zeng C, Xiang Z, Wang X, Zhang X, Ran Q, Guo H. A rare big Chinese family with thrombocytopenia 2: a case report and literature review. Frontiers Genetics. 2020;11:340.
  • Yoshimi A, Toya T, Nannya Y, Takaoka K, Kirito K, Ito E, Nakajima H, Hayashi Y, Takahashi T, Moriya-Saito A, et al., Spectrum of clinical and genetic features of patients with inherited platelet disorder with suspected predisposition to hematological malignancies: a nationwide survey in Japan. Ann Oncol 2016;27(5):887–895. doi:10.1093/annonc/mdw066.
  • Tan C, Dai L, Yang W, Li F, Wang L, Xiao Y, Wang X, Zhang Y, Wang Y, Zeng C, et al. Generation of the human induced pluripotent stem cell line (SHAMUi001-A) carrying the heterozygous c.-128G>T mutation in the 5′-UTR of the ANKRD26 gene. Stem Cell Res 2020;48:102002. doi:10.1016/j.scr.2020.102002.
  • Minamiguchi H, Kimura T, Urata Y, Miyazaki H, Bamba T, Abe T, Sonoda Y. Simultaneous signalling through c‐mpl, c‐kit and CXCR4 enhances the proliferation and differentiation of human megakaryocyte progenitors: possible roles of the PI3‐K, PKC and MAPK pathways. Brit J Haematol 2001;115(1):175–185. doi:10.1046/j.1365-2141.2001.03068.x.
  • Wahlster L, Verboon JM, Ludwig LS, Black SC, Luo W, Garg K, Voit RA, Collins RL, Garimella K, Costello M, et al., Familial thrombocytopenia due to a complex structural variant resulting in a WAC-ANKRD26 fusion transcript. J Exp Med 2021;218(6):e20210444. doi:10.1084/jem.20210444.
  • Acs P, Bauer PO, Mayer B, Bera T, Macallister R, Mezey E, Pastan I. A novel form of ciliopathy underlies hyperphagia and obesity in Ankrd26 knockout mice. Brain Struct Funct 2015;220(3):1511–1528. doi:10.1007/s00429-014-0741-9.
  • Conduit PT, Wainman A, Raff JW. Centrosome function and assembly in animal cells. Nat Rev Mol Cell Bio 2015;16(10):611–624. doi:10.1038/nrm4062.
  • Evans LT, Anglen T, Scott P, Lukasik K, Loncarek J, Holland AJ. ANKRD26 recruits PIDD1 to centriolar distal appendages to activate the PIDDosome following centrosome amplification. Embo J 2021;40(4):e105106. doi:10.15252/embj.2020105106.
  • Burigotto M, Mattivi A, Migliorati D, Magnani G, Valentini C, Roccuzzo M, Offterdinger M, Pizzato M, Schmidt A, Villunger A, et al., Centriolar distal appendages activate the centrosome-PIDDosome-p53 signalling axis via ANKRD26. Embo J 2021;40(4):e104844. doi:10.15252/embj.2020104844.
  • Matteo B, Luca LF. The PIDDosome: centrosome guardian and backup on the DNA damage response. Mol Cell Oncol 2021 May 4;8(3):1893625. doi:10.1080/23723556.2021.1893625.