Next Article in Journal
Abemaciclib, Palbociclib, and Ribociclib in Real-World Data: A Direct Comparison of First-Line Treatment for Endocrine-Receptor-Positive Metastatic Breast Cancer
Previous Article in Journal
Kinetic Intricacies of the Light Emission and Antiradical Influence of Exogenous Bioantioxidants Transformation Products in the Chemiluminescence Bioantioxidant Assay
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase

1
Department of Molecular Biology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
2
Department of Human Genetics, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
3
Central Clinical School, University of Sydney, Sydney 2006, Australia
4
Ph.D. Program in Biology-Neuroscience, Graduate Center, City University of New York, New York, NY 10016, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(10), 8487; https://doi.org/10.3390/ijms24108487
Submission received: 28 March 2023 / Revised: 27 April 2023 / Accepted: 2 May 2023 / Published: 9 May 2023
(This article belongs to the Section Molecular Neurobiology)

Abstract

:
Type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10), a homo-tetrameric multifunctional protein with 1044 residues encoded by the HSD17B10 gene, is necessary for brain cognitive function. Missense mutations result in infantile neurodegeneration, an inborn error in isoleucine metabolism. A 5-methylcytosine hotspot underlying a 388-T transition leads to the HSD10 (p.R130C) mutant to be responsible for approximately half of all cases suffering with this mitochondrial disease. Fewer females suffer with this disease due to X-inactivation. The binding capability of this dehydrogenase to Aβ-peptide may play a role in Alzheimer’s disease, but it appears unrelated to infantile neurodegeneration. Research on this enzyme was complicated by reports of a purported Aβ-peptide-binding alcohol dehydrogenase (ABAD), formerly referred to as endoplasmic-reticulum-associated Aβ-binding protein (ERAB). Reports concerning both ABAD and ERAB in the literature reflect features inconsistent with the known functions of 17β-HSD10. It is clarified here that ERAB is reportedly a longer subunit of 17β-HSD10 (262 residues). 17β-HSD10 exhibits L-3-hydroxyacyl-CoA dehydrogenase activity and is thus also referred to in the literature as short-chain 3-hydorxyacyl-CoA dehydrogenase or type II 3-hydorxyacyl-CoA dehydrogenase. However, 17β-HSD10 is not involved in ketone body metabolism, as reported in the literature for ABAD. Reports in the literature referring to ABAD (i.e., 17β-HSD10) as a generalized alcohol dehydrogenase, relying on data underlying ABAD’s activities, were found to be unreproducible. Furthermore, the rediscovery of ABAD/ERAB’s mitochondrial localization did not cite any published research on 17β-HSD10. Clarification of the purported ABAD/ERAB function derived from these reports on ABAD/ERAB may invigorate this research field and encourage new approaches to the understanding and treatment of HSD17B10-gene-related disorders. We establish here that infantile neurodegeneration is caused by mutants of 17β-HSD10 but not ABAD, and so we conclude that ABAD represents a misnomer employed in high-impact journals.

1. Introduction

The HSD17B10 gene was first cloned from the human brain in 1997 (GenBankTM accession number: AF037438) and mapped to Xp11.2 [1]. It encodes a mitochondrial multifunctional protein with a molecular weight of 108 kDa, composed of four identical subunit molecules. It plays an essential role in isoleucine as well as neurosteroid metabolism [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15]. The rat mitochondrial homolog [16] and the rat HSD17B10 gene [17] have also been characterized. Because the first enzymatic function identified for this member of the short-chain dehydrogenase/reductase (SDR) family was an L-3-hydroxyacyl-CoA dehydrogenase activity, it was formerly referred to as short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD) or L-3-hydroxyacyl-CoA dehydrogenase type 2 (HADII), having 261 amino acid residues per subunit [1].
Nonetheless, a concurrent report described a 27 kDa protein consisting of 262 residues as being associated with the endoplasmic reticulum (ER) and was thus referred to as endoplasmic-reticulum-associated Aβ-binding protein (ERAB) [18,19]. However, this report of an ERAB was found to be a subunit of SCHAD, which contained errors in its reported sequence as well as an erroneous intracellular localization [3,4,5,6,7,8]. Although it cannot be reasonably disputed that the ERAB researchers were aware of reports in the literature of the sequence and activities of SCHAD (i.e., 17β-HSD10), a SCHAD report [1] was cited as a reference in an ERAB JBC article [19] regarding the necessity of a corrigendum was readily apparent. Yet, no corrigendum was ever published, despite the journal Nature having a policy that “Classes uncorrected errors as misconduct” [20].
Data in the literature establish that SCHAD (i.e., 17β-HSD10) is a new type of 17β-hydroxysteroid dehydrogenase [3,4,5,6]. It is localized in the mitochondria [1,2,3,4,5,8,9,10,11,12,13], although most 17β-hydroxysteroid dehydrogenases are associated with the endoplasmic reticulum [21,22,23,24,25,26,27]. The N-terminal eleven residues of 17β-HSD10/SCHAD form an alpha-helix that serves as a mitochondrial-targeting signal (see Figure 5 of Ref. [5]). This explains why this multifunctional protein is localized in the mitochondria—it is not associated with the endoplasmic reticulum, as the literature concerning ERAB suggests (see Figure 4 of Ref. [18]). Efforts to identify additional mitochondrial-targeting signals in this protein are ongoing. This new 17β-HSD was designated type 10 17β-hydroxysteroid dehydrogenase (17β-HSD10) in an international 17β-HSDs conference [23]. It plays a critical role not only in steroid metabolism (see Figure 1 of Ref. [3]), but also in the metabolism of acyl thioesters [26] (Figure 1). Further, the SCHAD gene encoding this multifunctional protein was definitively designated HSD17B10 by the human genome organization (HUGO) [28,29]. The expression levels of 17β-HSD10 are dependent upon the disease state [30,31].
Because the HSD17B10 gene product was reported to be a mitochondrial protein [1,2,3,4,5,6,7,8,9,10,11,21,22], moniker ERAB was abandoned by ERAB researchers and thereafter referred to as amyloid beta-peptide-binding alcohol dehydrogenase (ABAD) [32,33] based upon its purported generalized alcohol dehydrogenase activity (Table 1), thus removing reference to the localization of ERAB in the endoplasmic reticulum [18,19]. Nonetheless, reports in the literature persisted in asserting that ABAD is associated with the endoplasmic reticulum (see Figure 2C of Ref. [32]). Unsurprisingly, there were reports in the literature concerning relocalized ABAD in the mitochondria [33], but the ABAD reports omitted any reference to the 17β-HSD10/SCHAD literature that had shown its N-terminal-targeting signal resulting in the mitochondrial localization of this protein [1,5,8,9,10,11,21,22]. This series of ABAD/ERAB reports [18,19,32] not only conflicted with the 17β-HSD10 research but also encouraged a trend in the literature to designate the HSD17B10 gene with various confounding names allegedly related to 17β-HSD10′s individual function. Fifteen alternative names were listed in the Gene ID: 3028, including ABAD, CAMR, ERAB, HCD2, MHBD, HADH2, MRPP2, MRX17, MRX31, SCHAD, MRXS10, SDR5C1, HSD10MD, 17b-HSD10 and DUPXp11.22. This ongoing nomenclature problem [32,33,34,35] directly confounded studies on 17β-HSD10 as well as HSD17B10-gene-related disorders. Since 17β-HSD10 is involved in the etiology of human diseases [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65], this is not a trivial issue. This issue deserves to be resolved by a clear and unambiguous correction in the scientific literature [66]. Further studies are desperately needed to firmly eliminate the ongoing confusion in the literature concerning this enzyme to thoroughly resolve potentially harmful inaccuracies. The published corrigenda and/or withdrawal of erroneous reports in the literature may go far in aiding the scientific community to arrive at an accurate consensus concerning this enzyme and, quite likely, benefit the affected patients and their treating physicians.
Although 17β-HSD10 and ABAD were found to have an identical structure [67], they have different reported functions. 17β-HSD10 can bind not only to Aβ [68], but also to other mitochondrial proteins to form RNase P [69], which is not inhibited by Aβ binding [70]. A missense mutation at 17β-HSD10 results in infantile neurodegeneration 17β-HSD10 deficiency [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57], for which Aβ peptide levels were undetectable or barely detectable in CSF [71]. This indicated that 17β-HSD10 plays a pathogenetic role in neurodegeneration beyond “the mediation of Aβ neurotoxicity” [18,19,32,33,34,35,72]. Apparently, it is necessary to establish conclusively whether previous reports in the ABAD/ERAB literature concerning its enzymatic function are accurate and reliable or not. Clarification of the alleged differences between 17β-HSD10 and ABAD will go far in helping to restore a proper understanding in the field as well as addressing research integrity issues in this field, which is essential for blazing an accurate and reliable path forward in the search for effective therapeutic targets for HSD17B10-gene-related disorders, including HSD10 deficiency or infantile neurodegeneration [36] and Alzheimer’s disease [7].

2. Could ABAD Catalyze the Oxidation of D- and L-Hydroxybutyrate at a Similar Rate?

With respect to the catalytic properties of human brain 17β-HSD10/SCHAD, it has been reported [1] that “the substrate stereospecificity of this novel enzyme is opposite to that of acetoacetyl-CoA reductase. The human brain dehydrogenase was found to metabolize only L-3-hydroxyacyl-CoA, but not the D-isomer”. Obviously, the substrate stereospecificity must originate from the orientation of the hydroxy group of β-hydroxybutyrate but not the coenzyme A moiety. Enzymologists would be duly surprised that the oxidation rate of D-3-hydroxybutyrate catalyzed by ABAD/ERAB could be measured spectrophotometrically (see Figure 1 of Ref. [32]). Since the primary structure of ABAD/ERAB is identical to 17β-HSD10/SCHAD, it is highly questioned how ABAD/ERAB could catalyze the oxidation of L-hydroxybutyrate and the D-isomer with almost the same rate (Vmax = ∼4 mU/mg). Furthermore, it was concluded by ABAD/ERAB researchers [19] that “we asked whether it might also catalyze the oxidation of the ketone body D-3-hydroxybutyrate. This was indeed the case, and oxidation proceeded with Km of ∼4.5 mM and Vmax of ∼4 mU/mg”. Assuming for the sake of argument that those reported kinetic constants were not erroneous, it is still questionable whether such an “ER-associated NAD+-dependent dehydrogenase” (see Figure 2C of Ref. [32]) could play a physiological role in ketone body metabolism; it is well known that most of the cell’s NAD+ is sequestered in the mitochondria. Further, life scientists are aware that β-hydroxybutyrate dehydrogenase, β-HBD [73] rather than 17β-HSD10, is involved in ketone body metabolism (see Figure 1). Although data about D-3-hydroxybutyrate supporting the growth of COS cells were provided by Yan et al. [32], such data could not serve as actual evidence for ABAD’s involvement in ketone body metabolism because β-HBD had not yet been knocked out by those investigators. These reports [18,19,32,33,34,35,72] have never been corrected and continue to confound efforts to advance in the physiological role of 17β-HSD10, and thus must be addressed.

3. Are Generalized Alcohol Dehydrogenase Activities Reported for ABAD Accurate?

The oxidation of aliphatic alcohols (C2–C10) catalyzed by ABAD/ERAB was reported by Yan et al. [19] (see Table 1), which is responsible for its misnomer as an Aβ-peptide-binding alcohol dehydrogenase [32,33,34,35,72]. The published experimental procedure in that report is quoted here: ”Alcohol dehydrogenase assays employed ERAB/HADH II (20 µg/mL), a range of alcohol substrate (methanol, ethanol, n-propanol, n-butanol, isobutanol, n-pentanol, (+)-2-octanol, (+)-2-octanol, (−)-2-octanol, and n-decanol; Sigma), and NAD+ (7.5 mM) in 22 mM sodium pyrophosphate, 0.3 mM sodium phosphate (pH 8.8). The reaction was run for 2 h at 25 °C, and the absorbance at 340 nm was monitored every 5 min as described above.” When assay mixtures containing (−)-2-octanol or its enantiomers as a substrate with or without 1% DMSO for spectrophotometrically measuring any generalized alcohol dehydrogenase activities were prepared according to the reported experimental procedure (see Table 2), an interface was found between two layers in all of the prepared 2-octanol assay mixtures with or without 1% DMSO (see Figure 2). Thus, it was physically impossible to determine the absorbance changes at 340 nm by using a spectrophotometer, as reported in Ref. [19]. Most of (−)-2-octanol or its enantiomers (the upper layer) remained undissolved in phosphate buffer (the bottom layer) following the addition of 1% DMSO, vortexing for 3 min and then being incubated at 25 °C for 72 h [19], which is due to extremely low solubilities of long-chain alcohols in aqueous media. For example, the solubility of 2-octanol in water is only 8.6 mM at 25 °C [74]. Obviously, the bottom layer contains a rather low concentration of any long-chain alcohol, and so it is impossible to accurately determine the Km value of (−)-2-octanol, previously reported to be as high as 43 ± 9.0 mM (see Table 1), which was derived from the published Figure 2B of Ref. [19]. This establishes, beyond physical doubt, that it would be (and indeed was) impossible for anyone to use 210 mM (−)-2-octanol as a substrate (see the legend of Figure 5B in Ref. [19]) to spectrophotometrically measure any purported generalized alcohol dehydrogenase activity that supports the misnomer Aβ-peptide-binding alcohol dehydrogenase [19,32,33,34,35,72]. Thus, it is not appropriate to consider ABAD/ERAB as an equivalent of 17β-HSD10, e.g., “ABAD/17β-HSD10”, as had been employed in some reports [75,76,77,78,79,80,81].

4. The Impact of Aβ-Peptide on the Stereo Structure of ABAD

The three-dimensional structure of 17β-HSD10 [67] is shown in Figure 3C. According to a report in Science [33], this structure would be dramatically altered as Aβ binds to this protein (see Figure 3A,B). Since no electron density of Aβ was reported, it would be highly questionable how the binding of Aβ with ABAD could result in so many different stereo structures (see Figure 3A,B). Although the dissociation constant for Aβ binding to ABAD/ERAB was reported to be approximately 65 nM [19], hundreds-of-fold higher concentrations of Aβ are reportedly required to inhibit the HAD activity of this multifunctional protein [68].
Compared with 17β-HSD10 (see Figure 3C), Aβ-bound ABAD seems to be a very unstable protein (Figure 3B), although it was found [68] that neither RNase P nor methyltransferase activities were specifically affected by the Aβ binding, even though 17β-HSD10 serves as the core of RNase P [69]. Therefore, the accuracy of the three-dimensional structures of Aβ-bound ABAD, previously published in a highly respected journal [33], needs to be verified independently.

5. HSD10 Deficiency Is an X-Linked Inherited Metabolic Disease

The HSD17B10 gene is highly conserved across a large evolutionary distance [75], implying that its gene product 17β-HSD10 plays a vital role [77,78]. It was also found [6] that the post-translational modification of 17β-HSD10 regulates its mitochondrial abundance and its enzymatic activity as well as the formation of RNase P. Missense mutations at the HSD17B10 gene results in 17β-HSD10 loss-of-function mutants causing HSD10 deficiency [36,37,38,39,40,41,42,43,44,45], formerly referred to as 2-methyl-3-hydroxybutyryl-CoA dehydrogenase (MHBD) deficiency [46,47,48,49,50,51,52,53,54,55,56,57]—an inborn error in isoleucine metabolism. In contrast, a silent mutation R192R alters the quantities but not the quality of 17β-HSD10 caused X-linked mental disability, choreoathetosis and abnormal behavior (MRXS10) [62,63], of which patients showed no blockage in the isoleucine metabolic pathway. HSD10 mitochondrial disease and MRXS10 resulted in different kinds of damage to the mitochondria [64] (see Figure 4). This suggested that a restoration of the OMIM#300220 would be appropriate. Gene duplication of HSD17B10 also results in mental disability [58]. Since 17β-HSD10 is an essential component of mtRNase P, it was proposed [59,60] that the abnormal processing of mtRNAs is related to the pathogenesis of HSD10 mitochondrial disease.

5.1. HSD10 Deficiency Is a Mitochondrial Metabolic Disease

The rediscovery of ERAB/ABAD in mitochondria was published as a Science report [33], which cited none of the previously reported literature on 17β-HSD10/SCHAD [1,5,8,9,10,11]. Most patients with this mitochondrial disease suffer from infantile neurodegeneration [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60]. There are at least sixteen reported and clinically related missense mutations in the HSD17B10 gene, but approximately half of such patients bear a 388C-T transition, resulting in a HSD10 (pR130C) mutant (Figure 5).

5.2. Diagnosis of HSD10 Deficiency

Patients suffering with this disease may have microcephaly but no dysmorphism or organomegaly. Clinical manifestations include epilepsy, choreoathetoid movements, ophthalmologic disorders and progressive neurodegeneration, psychomotor retardation or regression, hearing impairment and even cardiomyopathy, which causes some patients to experience a very short lifespan. This disease is an inborn error in isoleucine metabolism so that high levels of isoleucine metabolites—e.g., tiglylglycine, 2-methyl-3-hydroxybutyrate and 2-ethylhydracrylic acid (EHA), an intermediate in the R-pathway of isoleucine oxidation—would be detected in patients’ urine (see Figure 6). For the differential diagnosis of other isoleucine metabolism disorders, e.g., β-ketothiolase deficiency [57], a missense mutation must be identified in the HSD17B10 gene by deoxynucleotide sequencing as hard evidence for this disease [36].

5.3. A Hot Spot in the HSD17B10 Gene

Approximately half of the cases of HSD10 deficiency are due to a C > T mutation in exon 4 of the HSD17B10 gene; male patients with this mutation have a much more severe clinical phenotype than those bearing other mutations [3]. In order to answer the question as to why the p.R130C mutation is more frequently found in such patients, Rauschenberg et al. [56] suggested that “most other mutations in the HSD17B10 gene are not observed because they are incompatible with life”. Such an explanation may be plausible, but it does not address the underlying molecular mechanism.
A methylation analysis of the HSD17B10 gene [82,83] has later found a mutational hotspot, mCpG, at +2259 nucleotides from the initiation codon ATG of the HSD17B10 gene. The cytosine at that nucleotide position is >90% methylated in both active and inactive X chromosomes (Figure 7). Since the 5mC > T transition occurs approximately ten times more frequently than other transitions, this not only explains the high incidence of the p.R130C mutation in patients with HSD10 deficiency, but also suggests the possibility of a de novo mutation due to some environmental factor.

5.4. Sexuality Preference of This Disease Due to X-Inactivation

HSD10 deficiency is an X-linked, inherited metabolic disease, and its onset occurs at 6–18 months after birth. Because the lifespan of most male patients is short and they do not reach their sexual maturity, a missense mutation at the HSD17B10 gene in a child is usually inherited from his/her mother, a female carrier. In contrast to the hemizygosity of the X chromosome in males, females have two X chromosomes and one of them is inactive [84] due to X-inactivation (Figure 8). If the mutation happens in the inactive X chromosome, the mutated HSD17B10 gene may not be able to generate HSD10 mutants. This mechanism explains why female carriers have relatively milder, or even no, symptoms.

5.5. Pathogenesis of HSD10 Deficiency

Although the accumulation of isoleucine and branched-chain fatty acid metabolites may not be completely benign [85,86], it does not cause neurological symptoms [36]. The excretion of isoleucine metabolite, e.g., tiglylglycine, 2-methyl-3-hydroxybutyrate and 2-ethylhydracrylic acid, is a clinical indicator [36]. However, the pathogenesis of HSD10 deficiency may lie in the disruption of other functions of 17β-HSD10, e.g., an imbalance of neurosteroid metabolism [2,3,13,36,82] and the impairing of mtRNA metabolism [49,52,56]. The interference of allopregnanolone metabolism may alter neuroexcitability [3] and lead to neurological disorders in patients (Figure 9).

6. Concluding Remarks

The reported stereo structures, intracellular localization and activities of ABAD/ERAB are indeed extremely distinct from those of 17β-HSD10. This is almost certainly due to the inability to reproduce any valid experiments concerning the localization and functions of ABAD/ERAB, notwithstanding their publication in highly regarded and widely read journals. It is no longer appropriate to confound the literature by employing the misnomer ABAD because it frustrates the progress of critical work in the studies of HSD17B10-gene-related disorders, including Alzheimer’s disease and intellectual disabilities. More importantly, clarification of the differences between ABAD and 17β-HSD10 will address the research integrity issues in this area. The removal of ABAD/ERAB contents from the entry OMIM300256, which is undoubtedly specific for the type 10 17β-hydroxysteroid dehydrogenase, appears to be essential for restoring a proper starting point in the treatment of HSD17B10-gene-related disorders including HSD10 deficiency, an inborn error in isoleucine and neurosteroid metabolism. Furthermore, the identification of more compounds to adjust 17beta-HSD10 or to regulate the HSD17B10 gene expression may promote a new approach to infantile or senile neurodegeneration in the future.

Funding

This work was supported in part by the New York State Office for People with Developmental Disabilities.

Institutional Review Board Statement

This article has been approved by institutional reviewers.

Conflicts of Interest

Authors declare no conflict of interest.

Abbreviations

ABAD: Aβ-peptide-binding alcohol dehydrogenase; AcAcCoA, acetoacetyl coenzyme A; AD, Alzheimer’s disease; CSF, cerebrospinal fluid; DMSO (Me2SO), dimethyl sulfoxide; ER, endoplasmic reticulum; ERAB, endoplasmic-reticulum-associated Aβ-binding protein; HAD, L-3-hydroxyacyl-CoA dehydrogenase; HAD2 or HADH II, 3-hydroxyacyl-CoA dehydrogenase type 2; HSD, hydroxysteroid dehydrogenase; 17β-HSD10, 17β-hydroxysteroid dehydrogenase type 10; HUGO, human genome organization; MHBD, 2-methyl-3-hydroxybutyryl-CoA dehydrogenase; MRXS10, mental disability, X-linked, syndromic 10; mtRNA, mitochondrial tRNA; PDB, protein data base; OMIM, Online Mendelian Inheritance in Man; SCHAD, short-chain 3-hydroxyacyl-CoA dehydrogenase.

References

  1. He, X.Y.; Schulz, H.; Yang, S.Y. A human brain L-3-hydroxyacyl coenzyme A dehydrogenase is identical with an amyloid β-peptide binding protein involved in Alzheimer’s disease. J. Biol. Chem. 1998, 273, 10741–10746. [Google Scholar] [CrossRef] [PubMed]
  2. Porcu, P.; Barron, A.M.; Frye, C.A.; Walf, A.A.; Yang, S.Y.; He, X.Y.; Morrow, A.L.; Panzica, G.C.; Melcangi, R.C. Neurosteroidogenesis Today: Novel Targets for Neuroactive Steroid Synthesis and Action and Their Relevance for Translational Research. J. Neuroendocr. 2016, 28, 12351. [Google Scholar] [CrossRef] [PubMed]
  3. He, X.Y.; Dobkin, C.; Yang, S.Y. 17beta-Hydroxysteroid dehydrogenases and neurosteroid metabolism in the central nervous system. Mol. Cell. Endocrinol. 2019, 489, 92–97. [Google Scholar] [CrossRef] [PubMed]
  4. He, X.Y.; Isaacs, C.; Yang, S.Y. Roles of mitochondrial 17beta-hydroxysteroid dehydrogenase type 10 in Alzheimer’s disease. J. Alzheimer’s Dis. 2018, 62, 665–673. [Google Scholar] [CrossRef] [PubMed]
  5. Yang, S.Y.; He, X.Y. Role of type 10 17beta-hydroxysteroid dehydrogenase in the pathogenesis of Alzheimer’s disease. In Neuropathology and Genetics of Dementia; Tolnay, M., Probst, A., Eds.; Kluwer Academic/Plenum Publishers: New York, NY, USA, 2001; pp. 101–110. [Google Scholar]
  6. Liu, L.; Chen, S.; Yu, M.; Ge, C.; Ren, M.; Liu, B.; Yang, X.; Christian, T.W., Jr.; Hou, Y.M.; Zou, J.; et al. Deacetylation of HSD17B10 by SIRT3 regulates cell growth and cell resistance under oxidative and starvation stresses. Cell Death Dis. 2020, 11, 563. [Google Scholar] [CrossRef]
  7. He, X.Y.; Dobkin, C.; Brown, W.T.; Yang, S.Y. 3-Hydroxyacyl-CoA and alcohol dehydrogenase activities of mitochondrial type 10 17β-hydroxysteroid dehydrogenase in neurodegeneration study. J. Alzheimer’s Dis. 2022, 88, 1487–1497. [Google Scholar] [CrossRef]
  8. He, X.Y.; Merz, G.; Mehta, P.; Schulz, H.; Yang, S.Y. Human brain short chain L-3-hydroxyacyl coenzyme A dehydrogenase is a single-domain multifunctional enzyme. Characterization of a novel 17β-hydroxysteroid dehydrogenase. J. Biol. Chem. 1999, 274, 15014–15019. [Google Scholar] [CrossRef]
  9. He, X.Y.; Merz, G.; Yang, Y.Z.; Pullakart, R.; Mehta, P.; Schulz, H.; Yang, S.Y. Function of human short chain L-3-hydroxyacyl-CoA dehydrogenase in androgen metabolism. Biochim. Biophys. Acta 2000, 1484, 267–277. [Google Scholar] [CrossRef]
  10. He, X.Y.; Merz, G.; Yang, Y.Z.; Schulz, H.; Yang, S.Y. Characterization and localization of human type 10 17β-hydroxysteroid dehydrogenase. Eur. J. Biochem. 2001, 268, 4899–4907. [Google Scholar] [CrossRef]
  11. He, X.Y.; Yang, Y.Z.; Peehl, D.M.; Lauderdale, A.; Schulz, H.; Yang, S.Y. Oxidative 3α-hydroxysteroid dehydrogenase activity of human type 10 17β-hydroxysteroid dehydrogenase. J. Steroid Biochem. Mol. Biol. 2003, 87, 191–198. [Google Scholar] [CrossRef]
  12. Shafqat, N.; Elleby, B.; Svensson, S.; Shafqat, J.; Jörnvall, H.; Abrahmsen, L.; Oppermann, U. Expanded substrate screenings of human and Drosophila type 10 17beta-hydroxysteroid dehydrogenases (HSDs) reveal multiple specificities in bile acid and steroid hormone metabolism: Characterization of multifunctional 3alpha/7alpha/7beta/17beta/20beta/21-HSD. Biochem. J. 2003, 376 Pt 1, 49–60. [Google Scholar]
  13. Yang, S.Y.; He, X.Y.; Schulz, H. Multiple functions of type 10 17beta-hydroxysteroid dehydrogenase. Trends Endocrinol. Metab. 2005, 16, 167–175. [Google Scholar] [CrossRef]
  14. He, X.Y.; Yang, S.Y. Roles of type 10 17beta-hydroxysteroid dehydrogenase in intracrinology and metabolism of isoleucine and fatty acids. Endocr. Metab. Immune Disord.-Drug Targets 2006, 6, 95–102. [Google Scholar] [CrossRef]
  15. Yang, S.Y.; He, X.Y.; Miller, D. Hydroxysteroid (17beta) dehydrogenase X in human health and disease. Mol. Cell. Endocrinol. 2011, 343, 1–6. [Google Scholar] [CrossRef]
  16. Luo, M.J.; Mao, L.-F.; Schulz, H. Short chain 3-hydroxy-2-methylacyl-CoA dehydrogenase from rat liver: Purification and characterization of a novel enzyme of isoleucine metabolism. Arch. Biochem. Biophys. 1995, 321, 214–220. [Google Scholar] [CrossRef]
  17. He, X.Y.; Merz, G.; Chu, C.H.; Lin, D.; Yang, Y.Z.; Mehta, P.; Schulz, H.; Yang, S.Y. Molecular cloning, modeling, and localization of rat type 10 17β-hydroxysteroid dehydrogenase. Mol. Cell. Endocrinol. 2001, 171, 89–98. [Google Scholar] [CrossRef]
  18. Yan, S.D.; Fu, J.; Soto, C.; Chen, X.; Zhu, H.; Al-Mohanna, F.; Collison, K.; Zhu, A.; Stern, E.; Saido, T.; et al. An intracellular protein that binds amyloid-beta peptide and mediates neurotoxicity in Alzheimer’s disease. Nature 1997, 389, 689–695. [Google Scholar] [CrossRef]
  19. Yan, S.D.; Shi, Y.; Zhu, A.; Fu, J.; Zhu, H.; Zhu, Y.; Gibson, L.; Stern, E.; Collison, K.; Al-Mohanna, F.; et al. Role of ERAB/L-3-hydroxyacyl-coenzyme A dehydrogenase type II activity in Abeta-induced cytotoxicity. J. Biol. Chem. 1999, 274, 2145–2156. [Google Scholar] [CrossRef]
  20. Kamoun, S.; Zipfal, C. Class uncorrected errors as misconduct. Nature 2016, 531, 173. [Google Scholar] [CrossRef]
  21. He, X.Y.; Yang, Y.Z.; Schulz, H.; Yang, S.Y. Intrinsic alcohol dehydrogenase and hydroxysteroid dehydrogenase activities of human mitochondrial short chain L-3-hydroxyacyl-CoA dehydrogenase. Biochem. J. 2000, 345 Pt 1, 139–143. [Google Scholar] [CrossRef]
  22. Wen, G.Y.; Yang, S.Y.; Kaczmarski, W.; He, X.Y.; Pappas, K.S. Presence of hydroxysteroid dehydrogenase type 10 in amyloid plaques (Aps) of Hsiao’s APP-Sw transgenic mouse brains, but absence in Aps of Alzheimer’s disease brains. Brain Res. 2002, 954, 115–122. [Google Scholar] [CrossRef] [PubMed]
  23. Adamski, J.; Jakob, F.J. A guide to 17beta-hydroxysteroid dehydrogenases. Mol. Cell. Endocrinol. 2001, 171, 1–4. [Google Scholar] [CrossRef] [PubMed]
  24. Biswas, M.G.; Rusell, D.W. Expression, cloning and characterization of oxidative 17β- and 3α-hydroxysteroid dehydrogenases from rat and human prostate. J. Biol. Chem. 1997, 272, 15959–15966. [Google Scholar] [CrossRef] [PubMed]
  25. Poutanen, M.; Penning, T.M. Biology and clinical relevance of hydroxysteroid (17beta) dehydrogenase enzymes. Mol. Cell. Endocrinol. 2019, 489, 1–2. [Google Scholar] [CrossRef]
  26. Hiltunen, J.K.; Kastaniotis, A.J.; Autio, K.J.; Jiang, G.; Chen, Z.; Glumoff, T. 17B-hydroxysteroid dehydrogenases as acyl thioester metabolizing enzymes. Mol. Cell. Endocrinol. 2019, 489, 107–118. [Google Scholar] [CrossRef]
  27. Yang, S.Y.; He, X.Y.; Dobkin, C.; Isaacs, C.; Brown, W.T. Mental retardation and isoleucine metabolism. In Branched Chain Amino Acids in Clinical Nutrition: Volume I, Nutrition and Health; Rajendram, R., Preedy, V.R., Patel, V.B., Eds.; Springer: New York, NY, USA, 2015; pp. 157–170. [Google Scholar]
  28. Korman, S.H.; Yang, S.Y. HSD17B10 replaces HADH2 as the approved designation for the gene mutated in 2-methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency. Mol. Genet. Metab. 2007, 91, 115. [Google Scholar] [CrossRef]
  29. Yang, S.Y.; He, X.Y.; Miller, D. HSD17B10: A gene involved in cognitive function through metabolism of isoleucine and neuroactive steroids. Mol. Genet. Metab. 2007, 92, 36–42. [Google Scholar] [CrossRef]
  30. Kristofiková, Z.; Bocková, M.; Hegnerová, K.; Bartos, A.; Klaschka, J.; Rícný, J.; Rípová, D.; Homola, J. Enhanced levels of mitochondrial enzyme 17beta-hydroxysteroid dehydrogenase type 10 in patients with Alzheimer disease and multiple sclerosis. Mol. Biosyst. 2009, 5, 1174–1179. [Google Scholar] [CrossRef]
  31. Frackowiak, J.; Mazur-Kolecka, B.; Kaczmarski, W.; Dickson, D. Deposition of Alzheimer’s vascular amyloid-beta is associated with decreased expression of brain L-3-hydroxyacyl-coenzyme A dehydrogenase (ERAB). Brain Res. 2001, 907, 44–53. [Google Scholar] [CrossRef]
  32. Yan, S.D.; Zhu, Y.; Stern, E.D.; Hwang, Y.C.; Hori, O.; Ogawa, S.; Frosch, M.P.; Connolly, E.S., Jr.; McTaggert, R.; Pinsky, D.J.; et al. Amyloid β-peptide-binding alcohol dehydrogenase is a component of the cellular response to nutritional stress. J. Biol. Chem. 2000, 275, 27100–27109. [Google Scholar] [CrossRef]
  33. Lustbader, J.W.; Cirilli, M.; Lin, C.; Xu, H.W.; Takuma, K.; Wang, N.; Caspersen, C.; Chen, X.; Pollak, S.; Chaney, M.; et al. ABAD directly links Abeta to mitochondrial toxicity in Alzheimer’s disease. Science 2004, 304, 448–452. [Google Scholar] [CrossRef]
  34. Morsy, A.; Trippier, P.C. Amyloid-Binding Alcohol Dehydrogenase (ABAD) Inhibitors for the Treatment of Alzheimer’s Disease. J. Med. Chem. 2019, 62, 4252–4264. [Google Scholar] [CrossRef]
  35. Morsy, A.; Maddeboina, K.; Gao, J.; Wang, H.; Valdez, J.; Dow, L.F.; Wang, X.; Trippier, P.C. Functionalized Allopurinols Targeting Amyloid-Binding Alcohol Dehydrogenase Rescue Aβ-Induced Mitochondrial Dysfunction. ACS Chem. Neurosci. 2022, 13, 2176–2190. [Google Scholar] [CrossRef]
  36. Yang, S.Y.; He, X.Y.; Olpin, S.E.; Sutton, V.R.; McMenamin, J.; Philipp, M.; Denman, R.B.; Malik, M. Mental retardation linked to mutations in the HSD17B10 gene interfering with neurosteroid and isoleucine metabolism. Proc. Natl. Acad. Sci. USA 2009, 106, 14820–14824. [Google Scholar] [CrossRef]
  37. Seaver, L.H.; He, X.Y.; Abe, K.; Cowan, T.; Enns, G.M.; Sweetman, L.; Philipp, M.; Lee, S.; Malik, M.; Yang, S.Y. A novel mutation in the HSD17B10 gene of a 10-year-old boy with refractory epilepsy, choreoathetosis and learning disability. PLoS ONE 2011, 6, e27348. [Google Scholar] [CrossRef]
  38. Zschocke, J. HSD10 disease: Clinical consequences of mutations in the HSD17B10 gene. J. Inherit. Metab. Dis. 2012, 35, 81–89. [Google Scholar] [CrossRef]
  39. Vilardo, E.; Rossmanith, W.; Vilardo, E.; Rossmanith, W. Molecular insights into HSD10 disease: Impact of SDR5C1 mutations on the human mitochondrial RNase P complex. Nucleic Acids Res. 2015, 43, 5112–5119. [Google Scholar] [CrossRef]
  40. Falk, M.J.; Gai, X.; Shigematsu, M.; Vilardo, E.; Takase, R.; McCormick, E.; Christian, T.; Lace, E.; Pierce, E.A.; Consugar, M.; et al. A novel HSD17B10 mutation impairing the activities of the mitochondrial RNase P complex causes X-linked intractable epilepsy and neurodevelopmental regression. RNA Biol. 2016, 13, 477–485. [Google Scholar] [CrossRef]
  41. Oerum, S.; Roovers, M.; Leichsenring, M.; Acquaviva-Bourdain, C.; Beermann, F.; Gemperle-Britschgi, C.; Fouilhoux, A.; Korwitz-Reichelt, A.; Bailey, H.J.; Droogmans, L.; et al. Novel patient missense mutations in the HSD17B10 gene affect dehydrogenase and mitochondrial tRNA modification functions of the encoded protein. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 3294–3302. [Google Scholar] [CrossRef]
  42. Richardson, A.; Berry, G.T.; Garganta, C.; Abbott, M.A. Hydroxysteroid 17-Beta Dehydrogenase Type 10 Disease in Siblings. JIMD Rep. 2017, 32, 25–32. [Google Scholar]
  43. Waters, P.J.; Lace, B.; Buhas, D.; Gravel, S.; Cyr, D.; Boucher, R.M.; Bernard, G.; Lévesque, S.; Maranda, B. HSD10 mitochondrial disease: P.Leu122Val variant, mild clinical phenotype, and founder effect in French-Canadian patients from Quebec. Mol. Genet. Genom. Med. 2019, 7, e1000. [Google Scholar] [CrossRef] [PubMed]
  44. Akagawa, S.; Fukao, T.; Akagawa, Y.; Sasai, H.; Kohdera, U.; Kino, M.; Shigematsu, Y.; Aoyama, Y.; Kaneko, K. Japanese Male Siblings with 2-Methyl-3-Hydroxybutyryl-CoA Dehydrogenase Deficiency (HSD10 Disease) Without Neurological Regression. JIMD Rep. 2017, 32, 81–85. [Google Scholar] [PubMed]
  45. Su, L.; Li, X.; Lin, R.; Sheng, H.; Feng, Z.; Liu, I. Clinical and molecular analysis of 6 Chinese patients with isoleucine metabolism defects: Identification of 3 novel mutations in the HSD17B10 and ACAT1 genes. Metab. Brain Dis. 2017, 32, 2063–2071. [Google Scholar] [CrossRef] [PubMed]
  46. Zschocke, J.; Ruiter, J.P.; Brand, J.; Lindner, M.; Hoffmann, G.F.; Wanders, R.J.; Mayatepek, E. Progressive infantile neurodegeneration caused by 2-methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency: A novel inborn error of branched-chain fatty acid and isoleucine metabolism. Pediatr. Res. 2000, 48, 852–855. [Google Scholar] [CrossRef]
  47. Ensenauer, R.; Niederhoff, H.; Ruiter, J.P.; Wanders, R.J.; Schwab, K.O.; Brandis, M.; Lehnert, W. Clinical variability in 3-hydroxy-2-methylbutyryl-CoA dehydrogenase deficiency. Ann. Neurol. 2002, 51, 656–659. [Google Scholar] [CrossRef]
  48. Olpin, S.E.; Pollitt, R.J.; McMenamin, J.; Manning, N.J.; Besley, G.; Ruiter, J.P.; Wanders, R.J. 2-methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency in a 23-year-old man. J. Inherit. Metab. Dis. 2002, 25, 477–482. [Google Scholar] [CrossRef]
  49. Ofman, R.; Ruiter, J.P.; Feenstra, M.; Duran, M.; Poll-The, B.T.; Zschocke, J.; Ensenauer, R.; Lehnert, W.; Sass, J.O.; Sperl, W.; et al. 2-Methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency is caused by mutations in the HADH2 gene. Am. J. Hum. Genet. 2003, 72, 1300–1307. [Google Scholar] [CrossRef]
  50. Poll-The, B.T.; Wanders, R.J.; Ruiter, J.P.; Ofman, R.; Majoie, C.B.; Barth, P.G.; Duran, M. Spastic diplegia and periventricular white matter abnormalities in 2-methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency, a defect of isoleucine metabolism: Differential diagnosis with hypoxic-ischemic brain diseases. Mol. Genet. Metab. 2004, 81, 295–299. [Google Scholar] [CrossRef]
  51. Sass, J.O.; Forstner, R.; Sperl, W. 2-Methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency: Impaired catabolism of isoleucine presenting as neurodegenerative disease. Brain Dev. 2004, 26, 12–14. [Google Scholar] [CrossRef]
  52. Perez-Cerda, C.; García-Villoria, J.; Ofman, R.; Sala, P.R.; Merinero, B.; Ramos, J.; García-Silva, M.T.; Beseler, B.; Dalmau, J.; Wanders, R.J.; et al. 2-Methyl-3-hydroxybutyryl-CoA dehydrogenase (MHBD) deficiency: An X-linked inborn error of isoleucine metabolism that may mimic a mitochondrial disease. Pediatr. Res. 2005, 58, 488–491. [Google Scholar] [CrossRef]
  53. Sass, J.O.; Ensenauer, R.; Röschinger, W.; Reich, H.; Steuerwald, U.; Schirrmacher, O.; Engel, K.; Häberle, J.; Andresen, B.S.; Mégarbané, A.; et al. 2-Methylbutyryl-coenzyme A dehydrogenase deficiency: Functional and molecular studies on a defect in isoleucine catabolism. Mol. Genet. Metab. 2008, 93, 30–35. [Google Scholar] [CrossRef]
  54. García-Villoria, J.; Navarro-Sastre, A.; Fons, C.; Pérez-Cerdá, C.; Baldellou, A.; Fuentes-Castelló, M.A.; González, I.; Hernández-Gonzalez, A.; Fernández, C.; Campistol, J.; et al. Study of patients and carriers with 2-methyl-3-hydroxybutyryl-CoA dehydrogenase (MHBD) deficiency: Difficulties in the diagnosis. Clin. Biochem. 2009, 42, 27–33. [Google Scholar] [CrossRef]
  55. Marques, A.T.; Fernandes, P.A.; Ramos, M.J. Structural analysis of ABAD point mutations causing 2-methyl-3-hydroxybytyryl-CoA deficiency. Int. J. Quantum Chem. 2009, 110, 148–160. [Google Scholar] [CrossRef]
  56. Rauschenberger, K.; Schöler, K.; Sass, J.O.; Sauer, S.; Djuric, Z.; Rumig, C.; Wolf, N.I.; Okun, J.G.; Kölker, S.; Schwarz, H.; et al. A non-enzymatic function of 17beta-hydroxysteroid dehydrogenase type 10 is required for mitochondrial integrity and cell survival. EMBO Mol. Med. 2010, 2, 51–62. [Google Scholar] [CrossRef]
  57. Shu, J.B.; Zhang, Y.Q.; Jiang, S.Z.; Zhang, C.H.; Meng, Y.T.; Wang, H.; Song, L. Mutation analysis of a family with 2-Methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency. J. Chin. Pediatr. 2013, 51, 783–786. [Google Scholar]
  58. Wang, Q.; Chen, P.; Lou, J.; Liu, Y.; Yuan, H. Xp11.22 duplications in four unrelated Chinese families: Delineating the genotype-phenotype relationship for HSD17B10 and FGD1. BMC Med. Genom. 2020, 13, 66. [Google Scholar] [CrossRef]
  59. Deutschmann, A.J.; Amberger, A.; Zavadil, C.; Steinbeisser, H.; Mayr, J.A.; Feichtinger, R.G.; Oerum, S.; Yue, W.W.; Zschocke, J. Mutation or knock-down of 17β-hydroxysteroid dehydrogenase type 10 cause loss of MRPP1 and impaired processing of mitochondrial heavy strand transcripts. Hum. Mol. Genet. 2014, 23, 3618–3628. [Google Scholar] [CrossRef]
  60. Chatfield, K.C.; Coughlin, C.R., 2nd; Friederich, M.W.; Gallagher, R.C.; Hesselberth, J.R.; Lovell, M.A.; Ofman, R.; Swanson, M.A.; Thomas, J.A.; Wanders, R.J.; et al. Mitochondrial energy failure in HSD10 disease is due to defective mtDNA transcript processing. Mitochondrion 2015, 21, 1–10. [Google Scholar] [CrossRef]
  61. Bedoyan, J.K.; Hage, R.; Shin, H.K.; Linard, S.; Ferren, E.; Ducich, N.; Wilson, K.; Lehman, A.; Schillaci, L.A.; Manickam, K.; et al. Utility of specific amino acid ratios in screening for pyruvate dehydrogenase complex deficiencies and other mitochondrial disorders associated with congenital lactic acidosis and newborn screening prospects. JIMD Rep. 2020, 56, 70–81. [Google Scholar] [CrossRef]
  62. Reyniers, E.; Van Bogaert, N.P.; Vits, L.; Pauly, F.; Fransen, E.; Van Regemorter, V.; Kooy, F.R. A new neurological syndrome with mental retardation, choreoathetosis, and abnormal behavior maps to chromosome Xp11. Am. J. Hum. Genet. 1999, 65, 1406–1412. [Google Scholar] [CrossRef]
  63. Lenski, C.; Kooy, R.F.; Reyniers, E.; Loessner, D.; Wanders, R.J.; Winnepenninckx, B.; Hellerand, H.; Engert, S.; Schwartz, C.E.; Meindl, A.; et al. The reduced expression of the HADH2 protein causes X-linked mental retardation, choreoathetosis, and abnormal behavior. Am. J. Hum. Genet. 2007, 80, 372–377. [Google Scholar] [CrossRef]
  64. He, X.Y.; Wegiel, J.; Kooy, R.F.; Brown, W.T.; Yang, S.Y. HSD17B10 gene-related disorders are associated with abnormalities of mitochondrial function, morphology, dynamics and clearance. Ann. Genet. Genom. 2021, 1, 1005. [Google Scholar]
  65. Vinklarova, L.; Schmidt, M.; Benek, O.; Kuca, K.; Gunn-Moore, F.; Musilek, K. Friend or enemy? Review of 17β-HSD10 and its role in human health or disease. J. Neurochem. 2020, 155, 231–249. [Google Scholar] [CrossRef] [PubMed]
  66. Pulverer, B. When things go wrong: Correcting the scientific record. EMBO J. 2015, 34, 2483–2485. [Google Scholar] [CrossRef]
  67. Kissinger, C.R.; Rejto, P.A.; Pelletier, L.A.; Thomson, J.A.; Russell, A.; Villafranca, J.E. Crystal structure of human ABAD/HSD10 with a bound inhibitor: Implications for design of Alzheimer’s disease therapeutics. J. Mol. Biol. 2004, 342, 943–952. [Google Scholar] [CrossRef] [PubMed]
  68. Oppermann, U.C.; Salim, S.; Tjernberg, L.O.; Terenius, L.; Jörnvall, H. Binding of amyloid beta-peptide to mitochondrial hydroxyacyl-CoA dehydrogenase (ERAB): Regulation of an SDR enzyme activity with implications for apoptosis in Alzheimer’s disease. FEBS Lett. 1999, 451, 238–242. [Google Scholar] [CrossRef] [PubMed]
  69. Holzmann, J.; Frank, P.; Löffler, E.; Bennett, K.L.; Gerner, C.; Rossmanith, W. RNase P without RNA: Identification and functional reconstitution of the human mitochondrial tRNA processing enzyme. Cell 2008, 135, 462–474. [Google Scholar] [CrossRef] [PubMed]
  70. Vilardo, E.; Rossmanith, W. The amyloid-β-SDR5C1 (ABAD) interaction does not mediate a specific inhibition of mitochondrial RNase P. PLoS ONE 2013, 8, e65609. [Google Scholar] [CrossRef]
  71. Ortez, C.; Villar, C.; Fons, C.; Duarte, S.T.; Pérez, A.; García-Villoria, J.; Ribes, A.; Ormazábal, A.; Casado, M.; Campistol, J.; et al. Undetectable levels of CSF amyloid-beta peptide in a patient with 17beta-hydroxysteroid dehydrogenase deficiency. J. Alzheimer’s Dis. 2011, 27, 253–257. [Google Scholar] [CrossRef]
  72. Borger, E.; Aitken, L.; Du, H.; Zhang, W.; Gunn-Moor, F.J.; Yan, S.S. Is amyloid binding alcohol dehydrogenase a drug target for treating Alzheimer’s disease? Curr. Alzheimer Res. 2013, 10, 21–29. [Google Scholar]
  73. Churchill, P.; Hempel, J.; Romovacek, H.; Zhang, W.W.; Churchill, S.; Brennan, M. Primary structure of rat liver D-beta-hydroxybutyrate dehydrogenase from cDNA and protein analyses: A short-chain alcohol dehydrogenase. Biochemistry 1992, 31, 3793–3799. [Google Scholar] [CrossRef]
  74. Amidon, G.L.; Yalkowsky, S.H.; Leung, S. Solubility of nonelectrolytes in polar solvents II: Solubility of aliphatic alcohols in water. J. Pharm. Sci. 1974, 63, 1858–1866. [Google Scholar] [CrossRef]
  75. Marques, A.T.; Antunes, A.; Fernandes, P.A.; Ramos, M.J. Comparative evolutionary genomics of the HADH2 gene encoding Abeta-binding alcohol dehydrogenase/17beta-hydroxysteroid dehydrogenase type 10 (ABAD/HSD10). BMC Genom. 2006, 7, 202. [Google Scholar] [CrossRef]
  76. Benek, O.; Hroch, L.; Aitken, L.; Dolezal, R.; Guest, P.; Benkova, M.; Soukup, O.; Musil, K.; Kuca, K.; Smith, T.K.; et al. 6-benzothiazolyl ureas, thioureas and guanidines are potent inhibitors of ABAD/17beta-HSD10 and potential drugs for Alzheimer’s disease treatment: Design, synthesis and in vitro evaluation. Med. Chem. 2017, 13, 345–358. [Google Scholar] [CrossRef]
  77. Yang, S.Y.; He, X.Y.; Schulz, H. 3-Hydroxyacyl-CoA dehydrogenase and short-chain 3-hydroxyacyl-CoA dehydrogenase in human health and disease. FEBS J. 2005, 272, 4874–4883. [Google Scholar] [CrossRef]
  78. Yang, S.Y.; He, X.Y.; Isaacs, C.; Dobkin, C.; Miller, D.; Philipp, M. Roles of 17beta-hydroxysteroid dehydrogenase type 10 in neurodegenerative disorders. J. Steroid Biochem. Mol. Biol. 2014, 143, 460–472. [Google Scholar] [CrossRef]
  79. Hroch, L.; Guest, P.; Benek, O.; Soukup, O.; Janockova, J.; Dolezal, R.; Kuca, K.; Aitken, L.; Smith, T.K.; Gunn-Moore, F.; et al. Synthesis and evaluation of frentizole-based indolyl thiourea analogues as MAO/ABAD inhibitors for Alzheimer’s disease treatment. Bioorganic Med. Chem. 2017, 25, 1143–1152. [Google Scholar] [CrossRef]
  80. Xiao, X.; Chen, Q.; Zhu, X.; Wang, Y. ABAD/17β-HSD10 reduction contributes to the protective mechanism of huperzine a on the cerebral mitochondrial function in APP/PS1 mice. Neurobiol. Aging 2019, 81, 77–87. [Google Scholar] [CrossRef]
  81. Marques, A.T.; Fernandes, P.A.; Ramos, M.J. ABAD: A potential therapeutic target for Abeta-induced mitochondrial dysfunction in Alzheimer’s disease. Mini Rev. Med. Chem. 2009, 9, 1002–1008. [Google Scholar] [CrossRef]
  82. Yang, S.Y.; Dobkin, C.; He, X.Y.; Philipp, M.; Brown, W.T. A 5-methyl-cytosine hotspot responsible for the prevalent HSD17B10 mutation. Gene 2013, 515, 280–284. [Google Scholar] [CrossRef]
  83. Yang, S.Y.; Dobkin, C.; He, X.Y.; Brown, W.T. Transcription start sites and epigenetic analysis of the HSD17B10 proximal promoter. BMC Biochem. 2013, 14, 17. [Google Scholar] [CrossRef] [PubMed]
  84. He, X.Y.; Dobkin, C.; Yang, S.Y. Does the HSD17B10 gene escape from X-Inactivation? Eur. J. Hum. Genet. 2011, 19, 123–124. [Google Scholar] [CrossRef] [PubMed]
  85. Fukao, T.; Nakamura, H.; Nakamura, K.; Perez-Cerda, C.; Baldellou, A.; Barrionuevom, C.R.; Castello, F.G.; Kohno, Y.; Ugarte, M.; Kondo, N. Characterization of six mutations in five Spanish patients with mitochondrial acetoacetyl-CoA thiolase deficiency: Effects of amino acid substitutions on tertiary structure. Mol. Genet. Metab. 2002, 75, 235–243. [Google Scholar] [CrossRef] [PubMed]
  86. Rossa, R.B.; Schuck, P.F.; de Assis, D.R.; Latini, A.; Dalcin, K.B.; Ribeiro, C.A.J.; da C Ferreira, G.; Maria, R.C.; Leipnitz, G.; Perry, M.L.S.; et al. Inhibition of energy metabolism by 2-methylacetoacetate and 2-methyl-3-hydroxybutyrate in cerebral cortex of developing rats. J. Inherit. Metab. Dis. 2005, 28, 501–515. [Google Scholar] [CrossRef]
Figure 1. A summary of isoleucine, ketone bodies and glucose metabolism as well as bioenergetics in the brain mitochondria. Abbreviations: AST, acetoacetyl succinyl-CoA transferase; BCATm, branched-chain amino acid transaminase; BCAT-PLP, branched-chain amino acid transaminase pyridoxal 5′-phosphate; BCAT-PMP, branched-chain amino acid transaminase pyridoxamine phosphate; BCKDC, branched-chain α-keto acid dehydrogenase complex; β-HBD, β-hydroxybutyrate dehydrogenase; HAD, 3-hydroxyacyl-CoA dehydrogenase; 17β-HSD10, 17β-hydroxysteroid dehydrogenase type 10; MM, methylmalonyl-CoA mutase; PCC, propionyl-CoA carboxylase; MR, methylmalonyl-CoA racemase; PDH, pyruvate dehydrogenase (reproduced from Figure 1 of Ref. [7]).
Figure 1. A summary of isoleucine, ketone bodies and glucose metabolism as well as bioenergetics in the brain mitochondria. Abbreviations: AST, acetoacetyl succinyl-CoA transferase; BCATm, branched-chain amino acid transaminase; BCAT-PLP, branched-chain amino acid transaminase pyridoxal 5′-phosphate; BCAT-PMP, branched-chain amino acid transaminase pyridoxamine phosphate; BCKDC, branched-chain α-keto acid dehydrogenase complex; β-HBD, β-hydroxybutyrate dehydrogenase; HAD, 3-hydroxyacyl-CoA dehydrogenase; 17β-HSD10, 17β-hydroxysteroid dehydrogenase type 10; MM, methylmalonyl-CoA mutase; PCC, propionyl-CoA carboxylase; MR, methylmalonyl-CoA racemase; PDH, pyruvate dehydrogenase (reproduced from Figure 1 of Ref. [7]).
Ijms 24 08487 g001
Figure 2. Interface between alcohol and phosphate buffer in generalized alcohol dehydrogenase assay mixtures reported in the literature. Assay mixtures were prepared according to the experimental procedure [19] with 160 mM (-)-2-octanol (tube 1), 210 mM (-)-2-octanol (tube 2), 210 mM (-)-2-octanol with 1% DMSO (tube 3), 210 mM (-)-2-octanol with 1% DMSO mixed with alcohol first (tube 3′), 84 mM (+)-2-octanol (tube 4) and 85 mM (+)-2-octanol (tube 5), respectively. To highlight the interface, the top 1 mL of the assay mixture was transferred to a narrower tube (reproduced from Figure 5 of Ref. [7]).
Figure 2. Interface between alcohol and phosphate buffer in generalized alcohol dehydrogenase assay mixtures reported in the literature. Assay mixtures were prepared according to the experimental procedure [19] with 160 mM (-)-2-octanol (tube 1), 210 mM (-)-2-octanol (tube 2), 210 mM (-)-2-octanol with 1% DMSO (tube 3), 210 mM (-)-2-octanol with 1% DMSO mixed with alcohol first (tube 3′), 84 mM (+)-2-octanol (tube 4) and 85 mM (+)-2-octanol (tube 5), respectively. To highlight the interface, the top 1 mL of the assay mixture was transferred to a narrower tube (reproduced from Figure 5 of Ref. [7]).
Ijms 24 08487 g002
Figure 3. Comparison of the crystal structure images of human Aβ-bound ABAD (A,B) to that of human 17β-HSD10 (C). (A) A section of the crystal packing interactions, showing the large solvent channels. Each ABAD molecule is shown in a different color. The ordered ends of the LD loop, residues 94 and 114, are marked as red and blue balls, respectively, and the hypothetical loops are shown in pink as dotted lines [33]; (B) PDB: 1SO8 [33] shows only one subunit associated with another three subunits which have no contact with each other; (C) PDB: 1U7T [67] shows a close association of four subunits in human HSD10.
Figure 3. Comparison of the crystal structure images of human Aβ-bound ABAD (A,B) to that of human 17β-HSD10 (C). (A) A section of the crystal packing interactions, showing the large solvent channels. Each ABAD molecule is shown in a different color. The ordered ends of the LD loop, residues 94 and 114, are marked as red and blue balls, respectively, and the hypothetical loops are shown in pink as dotted lines [33]; (B) PDB: 1SO8 [33] shows only one subunit associated with another three subunits which have no contact with each other; (C) PDB: 1U7T [67] shows a close association of four subunits in human HSD10.
Ijms 24 08487 g003
Figure 4. Comparison of the electron-microscopic images of lymphoblastoid cells from patients with an intellectual disability suffering from MRXS10 (A) and HSD10 mitochondrial disease (B), and normal male control (C). The arrows indicate the mitochondria (reproduced from Figure 3 of Ref. [64]).
Figure 4. Comparison of the electron-microscopic images of lymphoblastoid cells from patients with an intellectual disability suffering from MRXS10 (A) and HSD10 mitochondrial disease (B), and normal male control (C). The arrows indicate the mitochondria (reproduced from Figure 3 of Ref. [64]).
Ijms 24 08487 g004
Figure 5. The missense mutations of 17β-HSD10, encoded by the HSD17B10 gene, result in HSD10 deficiency, whereas a silent mutation causes mental disability, X-linked syndromic 10 (MRXS10). Female cases are indicated by a red vertical bar, whereas male cases are in blue. The bar height is approximately proportional to the number of cases. The contents shown in a dotted square were expanded by approximately 6x. The N and C represent the N- and C-terminals of 17β-HSD10, respectively. (Updated from Figure 5 of Ref. [3]).
Figure 5. The missense mutations of 17β-HSD10, encoded by the HSD17B10 gene, result in HSD10 deficiency, whereas a silent mutation causes mental disability, X-linked syndromic 10 (MRXS10). Female cases are indicated by a red vertical bar, whereas male cases are in blue. The bar height is approximately proportional to the number of cases. The contents shown in a dotted square were expanded by approximately 6x. The N and C represent the N- and C-terminals of 17β-HSD10, respectively. (Updated from Figure 5 of Ref. [3]).
Ijms 24 08487 g005
Figure 6. Urine organic acids were analyzed as trimethylsilyl (TMS) derivatives by gas chromatography–mass spectrometry. Urine organic acid profiles: (A) normal control; (B) patient with HSD10 deficiency; (C) patient with HSD10 deficiency under a low protein dietary regimen. 2MHBA, 2-methyl-3-hydroxybutyrc acid; EHA, 2-ethylhydracrylic acid; TG, tiglylglycine; IS, internal standard (reproduced from Figure 13.2 of Ref. [27]).
Figure 6. Urine organic acids were analyzed as trimethylsilyl (TMS) derivatives by gas chromatography–mass spectrometry. Urine organic acid profiles: (A) normal control; (B) patient with HSD10 deficiency; (C) patient with HSD10 deficiency under a low protein dietary regimen. 2MHBA, 2-methyl-3-hydroxybutyrc acid; EHA, 2-ethylhydracrylic acid; TG, tiglylglycine; IS, internal standard (reproduced from Figure 13.2 of Ref. [27]).
Ijms 24 08487 g006
Figure 7. Methylation analysis of exon 4 of the HSD17B10 gene. The bisulfite sequencing of chromosomal DNA from a normal male is displayed in (A), and that from a normal female in (B). The ordinate shows the relative light unit detected after dispensation of each nucleotide substrate. The abscissa shows the dispensation order (“e” and “s” are the controls). The “bisulfite” line shows the predicted sequence after modification. “Y” represents C or T. Lines connecting the dispensed nucleotide to the bisulfite sequence indicate how the light signal represents the modified sequence. The height of the signal is proportional to the number of nucleotides at that position [82] (reproduced from Figure 1 of Ref. [82]).
Figure 7. Methylation analysis of exon 4 of the HSD17B10 gene. The bisulfite sequencing of chromosomal DNA from a normal male is displayed in (A), and that from a normal female in (B). The ordinate shows the relative light unit detected after dispensation of each nucleotide substrate. The abscissa shows the dispensation order (“e” and “s” are the controls). The “bisulfite” line shows the predicted sequence after modification. “Y” represents C or T. Lines connecting the dispensed nucleotide to the bisulfite sequence indicate how the light signal represents the modified sequence. The height of the signal is proportional to the number of nucleotides at that position [82] (reproduced from Figure 1 of Ref. [82]).
Ijms 24 08487 g007
Figure 8. Escape of the HSD17B10 gene from X-chromosome inactivation. The expression of HSD17B10 transcripts from inactive X (Xi) hybrids. Samples scored as positive are expressed as at least >10% of the active X (Xa) levels; their number is shown as the numerator, whereas the total number of hybrids tested is shown as the denominator. An X-linked intellectual disability by gene mutation(s) bears an asterisk. Reproduced from Ref. [84].
Figure 8. Escape of the HSD17B10 gene from X-chromosome inactivation. The expression of HSD17B10 transcripts from inactive X (Xi) hybrids. Samples scored as positive are expressed as at least >10% of the active X (Xa) levels; their number is shown as the numerator, whereas the total number of hybrids tested is shown as the denominator. An X-linked intellectual disability by gene mutation(s) bears an asterisk. Reproduced from Ref. [84].
Ijms 24 08487 g008
Figure 9. Roles of 17β-HSD10 in neurosteroid metabolism. 17β-HSD10 (red) is localized in the mitochondria and catalyzes the oxidation of allopregnanolone (ALLOP, blue), a positive modulator of GABAA receptors that potentiates GABA to increase the opening of Cl- channels. The postsynaptic GABAA receptor was magnified and is shown at the right side. ―• indicates the binding sites of individual modulators on the GABAA receptor [3]. Reproduced from Figure 2 of Ref. [3].
Figure 9. Roles of 17β-HSD10 in neurosteroid metabolism. 17β-HSD10 (red) is localized in the mitochondria and catalyzes the oxidation of allopregnanolone (ALLOP, blue), a positive modulator of GABAA receptors that potentiates GABA to increase the opening of Cl- channels. The postsynaptic GABAA receptor was magnified and is shown at the right side. ―• indicates the binding sites of individual modulators on the GABAA receptor [3]. Reproduced from Figure 2 of Ref. [3].
Ijms 24 08487 g009
Table 1. Generalized alcohol dehydrogenase and HAD activities of ABAD/ERAB *.
Table 1. Generalized alcohol dehydrogenase and HAD activities of ABAD/ERAB *.
Substrate or AlcoholKm (mM)Vmax (Units/mg)Kcat a (s−1)Catalytic Efficiency, Kcat/Km (M−1s−1)
Reduction of S-acetoacetyl-CoA
S-acetoacetyl-CoA0.068 ± 0.020430 ± 451902.8 × 106
Oxidation of alcohol substrate b
17β-Estradiol0.014 ± 0.00623 ± 3107.4 × 105
MethanolNo activityNo activityNo activityNo activity
Ethanol1210 ± 2602.2 ± 0.41.00.82
Isopropanol150 ± 1736 ± 216110
n-Propanol272 ± 624.2 ± 0.51.96.9
n-butanol53 ± 69.0 ± 0.34.076
Isobutanol56 ± 168.0 ± 0.73.664
n-Pentanol18 ± 56.9 ± 0.43.1170
(±)-2-Octanol85 ± 17245 ± 201101300
(+)-2-Octanol84 ± 16102 ± 846540
(−)-2-Octanol43 ± 9.0133 ± 23601400
n-Decanol14 ± 6.32.8 ± 0.51.390
* Reproduced from Table 13.1 of Ref. [27]. Data reported for a number of alcohols such as 2-octanol enantiomers and 17β-estradiol [19] were non-reproducible [4,7,21]. a Calculation based on 1 unit representing 1 μmol of product formed per min, and a molecular mass of the enzyme used for the calculation of Kcat was 26,926 Da because there was 1 active site per subunit. b Experiments were performed by incubating ERAB/HAD II with a range of concentrations of the indicated substrates in the presence of NAD+/NADH. Details of the experimental procedures are described in the text of Ref. [19].
Table 2. The preparation of assay mixtures to purportedly measure the generalized alcohol dehydrogenase activities of ABAD .
Table 2. The preparation of assay mixtures to purportedly measure the generalized alcohol dehydrogenase activities of ABAD .
Tube1233′45
(±)-2-Octanol/////135 μL
(+)-2-Octanol////133 μL/
(−)-2-Octanol255 μL333 μL333 μL333 μL//
0.2 M Sodium phosphate (pH 8.8)1100 μL1100 μL1100 μL1100 μL1100 μL1100 μL
10 mM Sodium phosphate (pH 8.8)300 μL300 μL300 μL300 μL300 μL300 μL
75 mM NAD+1000 μL1000 μL1000 μL1000 μL1000 μL1000 μL
DMSO//100 μL100 μL *//
H2O7345 μL7267 μL7167 μL7167 μL7467 μL7465 μL
Reproduced from Table 1 of Ref. [7] * mixed with alcohol first and vortexed for 3 min; /, means “do not apply”.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

He, X.-Y.; Dobkin, C.; Brown, W.T.; Yang, S.-Y. Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase. Int. J. Mol. Sci. 2023, 24, 8487. https://doi.org/10.3390/ijms24108487

AMA Style

He X-Y, Dobkin C, Brown WT, Yang S-Y. Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase. International Journal of Molecular Sciences. 2023; 24(10):8487. https://doi.org/10.3390/ijms24108487

Chicago/Turabian Style

He, Xue-Ying, Carl Dobkin, William Ted Brown, and Song-Yu Yang. 2023. "Infantile Neurodegeneration Results from Mutants of 17β-Hydroxysteroid Dehydrogenase Type 10 Rather Than Aβ-Binding Alcohol Dehydrogenase" International Journal of Molecular Sciences 24, no. 10: 8487. https://doi.org/10.3390/ijms24108487

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