Editorial Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 7, 2017; 23(29): 5257-5265
Published online Aug 7, 2017. doi: 10.3748/wjg.v23.i29.5257
Liver and the defects of cholesterol and bile acids biosynthesis: Rare disorders many diagnostic pitfalls
Gaetano Corso, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
Antonio Dello Russo, Monica Gelzo, Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
Author contributions: Corso G and Gelzo M have substantially contributed to conception and design of the study and writing the manuscript; Dello Russo A has contributed to the critical discussion of the manuscript.
Conflict-of-interest statement: The authors declare no conflict of interest related to this publication.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Monica Gelzo, PhD, Researcher, Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy. monicagelzo@gmail.com
Telephone: +39-81-7463653 Fax: +39-81-7463653
Received: March 18, 2017
Peer-review started: March 21, 2017
First decision: April 26, 2017
Revised: May 1, 2017
Accepted: July 4, 2017
Article in press: July 4, 2017
Published online: August 7, 2017
Processing time: 142 Days and 7.9 Hours

Abstract

In recent decades, biotechnology produced a growth of knowledge on the causes and mechanisms of metabolic diseases that have formed the basis for their study, diagnosis and treatment. Unfortunately, it is well known that the clinical features of metabolic diseases can manifest themselves with very different characteristics and escape early detection. Also, it is well known that the prognosis of many metabolic diseases is excellent if diagnosed and treated early. In this editorial we briefly summarized two groups of inherited metabolic diseases, the defects of cholesterol biosynthesis and those of bile acids. Both groups show variable clinical manifestations but some clinical signs and symptoms are common in both the defects of cholesterol and bile acids. The differential diagnosis can be made analyzing sterol profiles in blood and/or bile acids in blood and urine by chromatographic techniques (GC-MS and LC-MS/MS). Several defects of both biosynthetic pathways are treatable so early diagnosis is crucial. Unfortunately their diagnosis is made too late, due either to the clinical heterogeneity of the syndromes (severe, mild and very mild) that to the scarcity of scientific dissemination of these rare diseases. Therefore, the delay in diagnosis leads the patient to the medical observation when the disease has produced irreversible damages to the body. Here, we highlighted simple clinical and laboratory descriptions that can potentially make you to suspect a defect in cholesterol biosynthesis and/or bile acids, as well, we suggest appropriate request of the laboratory tests that along with common clinical features can help to diagnose these defects.

Key Words: Cholesterol, Bile acids, Liver metabolism, Gas chromatography coupled to mass spectrometry, Liquid chromatography coupled to tandem mass spectrometry

Core tip: The genetic defects of cholesterol and bile acid biosynthesis are characterized by a diversity of clinical findings affecting the liver, the intestine, and the nervous system. Many of these defects are efficaciously treatable but owing to mild phenotypes many cases can escape to an earlier diagnosis and are identified after few years or adulthood with irreversible injuries or more difficult to treat. Here, we highlighted simple clinical and laboratory descriptions that can potentially make you to suspect a defect in cholesterol biosynthesis and/or bile acids, in order to reduce the diagnostic delay and to improve the prognosis of these defects.



INTRODUCTION

Metabolic diseases are genetic based conditions in which the defective gene results in deficiency of an enzyme that leads to metabolic disorders. Today, several hundred of genetic metabolic disorders are known, and their symptoms, treatments, and prognosis vary widely.

The genetic defects in the biosynthesis of cholesterol and bile acids have been well studied in recent decades and several of these defects are treatable with results that greatly improve the prognosis and the patient’s health.

Cholesterol is essential for the maintenance of life in vertebrates[1]. It is an essential metabolite of cellular membrane structure, and is also the precursor of bile acids, steroid hormones, and oxysterols, which play multiple and important biological functions throughout the body[2,3]. Especially, the synthesis of bile acids and the bile formation plays a pivotal role to survival of organism.

Body cholesterol levels are finely regulated by the balance between mechanisms of diet cholesterol absorption, liver synthesis, biliary excretion, and peripheral tissue uptake. All this requires close molecular collaboration between the liver, the intestine and all the tissues of the whole organism[4]. Plasma levels of cholesterol are controlled by the liver through specific lipoprotein receptors, sterol transporters and intracellular nuclear receptors, which translate the signals derived from variations of cholesterol levels in selective variations of gene expression[5-8]. The liver is the main organ of cholesterol synthesis in most mammals[9,10], through an intricate pathway involving more than 30 enzymatic steps[11,12].

The intestine plays a pivotal role in the homeostasis of cholesterol. In fact, it is the main site for cholesterol absorption and excretion. Cholesterol that is absorbed into the lumen of the small intestine derives from food digestion, partial reabsorption of biliary cholesterol, and from that one released by the intestinal epithelial cells. The presence of bile acids as detergents is obligatory for the solubilization and uptake of intestinal cholesterol. They are excreted from liver and reabsorbed from the intestinal lumen back to the liver through the entero-hepatic circulation[13].

Beside cholesterol, also plant sterols (e.g., campesterol and β-sitosterol) and stanols are absorbed by Niemann-Pick C1 Like 1 (NPC1L1), a transporter localized in the microvilli cells of small intestine in the apical plasma membrane, which facilitates their uptake and transport. NPC1L1 is also localized near to other transporters such as ATP-binding cassette G5/G8 (ABCG5/G8) that act as inverted transporters pumping cholesterol and other sterols from enterocytes back to the intestinal lumen, as well as from hepatocytes to bile ducts. Instead, the bile acids are excreted into bile via specific pump proteins (BSEP or ABCB11)[14,15].

Mutations in ABCG5/G8 genes cause Sitosterolemia (STSL, OMIM #210250), a rare autosomal recessive lipid storage disease where it is found elevated plasma levels of cholesterol and non-cholesterol sterols due to impaired intestinal absorption and biliary sterol excretion[16].

ABCG5/G8 are controlled by cholesterol through liver X receptors (LXRs), which in turn are activated by oxysterols and other plant sterol derivatives[17]. Another target of LXR is ABCA1, which is the reverse transporter of cholesterol. By this mechanism the surplus of free cholesterol is eliminated from peripheral tissues through biliary excretion or non-biliary trans-intestinal cholesterol efflux (TICE) that, to our knowledge, has been demonstrated only in animal models[18,19].

The fecal excretion of neutral and acidic sterols, coming from cholesterol and non-cholesterol sterols, also contributes to cholesterol balance in whole body. The production of bile acids from cholesterol is the main mechanism for removing sterols from our body. At the same time, it has been showed that bile acids play various functions in signal transduction pathways for energy metabolism regulation. Biosynthesis of bile acids occurs through two main pathways involving 17 different enzymes, many of which are predominantly expressed in the liver[20].

Since excess and cholesterol deficiency can cause serious damage to our body’s structures, cholesterol homeostasis is required to be tightly controlled. In fact, hypercholesterolemia is the main risk factor for the development of atherosclerosis and consequently of cardiovascular disease[21,22]. The different types of hypercholesterolemia, such as familial hypercholesterolemia, secondary hypercholesterolemia, polygenic hypercholesterolemia, have been well characterized[23] and will not be reported here.

Instead, inherited defects of cholesterol biosynthesis lead to severe clinical phenotypes; for example, the absence of 3-hydroxy-3-methylglutaryl CoA (HMG-CoA) reductase, which is the main regulatory enzyme of cholesterol biosynthesis, is not compatible with life[24]. In addition, the liver cholesterol catabolism disorders, such as the synthesis of bile acids, can be the cause or can be secondary to biliary stasis, which acts significantly on the effectiveness of regulatory mechanisms in the liver and intestine[4].

This work aims to draw the attention of physicians on clinical, diagnostic, and therapeutic knowledge published on some defects of biosynthesis of cholesterol and of bile acids. These defects have a widely variable clinical phenotype, but some signs and symptoms are common to the different genetic forms known so far. Unfortunately, since these defects also have mild phenotypes or even very mild, they can escape to an earlier diagnosis, and many cases are identified after a few years or in adulthood with irreversible injuries or more difficult to treat[25,26].

The diagnostic laboratory path of inborn errors of biosynthesis of cholesterol[27-29] and of bile acids[26,30] is of fundamental importance for the differential diagnosis and the diagnostic accuracy of these diseases. Therefore, we need of knowledge about the laboratory tests for the metabolic study of patients that putatively suffer from these defects.

Today, the applications in the clinical field of mass spectrometry coupled with chromatography methods (gaseous or liquid) are already widely used in many laboratories, and the determination of metabolic profiles, such as sterols[31-34] or bile acids[35], in biological fluids (blood, plasma, urine) facilitates the study of many inborn errors of metabolism including cholesterol and bile acids.

Here, we will focus on some treatable disorders for which the early diagnosis is fundamental, and this article also provides a simple diagnostic flow chart together to clinical characteristics that could be useful for the clinicians in the investigation of cholesterol and bile acid disorders.

CHOLESTEROL BIOSYNTHESIS DEFECTS

After the discovery that Smith-Lemli-Opitz syndrome (SLOS, OMIM # 270400), a congenital multi-malformative syndrome, is due by a disorder of post-squalene cholesterol biosynthesis caused by defects in 7-dehydrocholesterol reductase[36], other malformative syndromes have been discovered in humans caused by other cholesterol biosynthesis defects. In Table 1 are reported all defects to date discovered in the pathway of cholesterol synthesis in humans[25,28,36-52]. Generally, the onset of these diseases occurs at birth, even though some phenotypes may present during the first months of life (CDPX2)[25] or in the early infancy (SC4MOL deficiency)[49,50]. Since the phenotypes of these defects can be highly variable, in particular the patients with a mild phenotype, the diagnosis can be missed at birth and can be made very belatedly. The phenotypic spectrum of SLOS is extremely wide, and the cases more severely affected by malformations die in utero or early after birth. Instead, the patients mildly affected suffer from less severe malformations, intellectual disability and behavioral problems[28].

Table 1 Inherited defects of cholesterol biosynthesis in humans.
DisorderOMIM#FrequencyEnzyme (blood biomarkers)Primary site of expressionTreatableRef.
SLOS2704001:20000/1:500007-dehydrocholesterol reductase (7-dehydrocholesterol, 8-dehydrocholesterol)MultisystemicYes[28,36]
Mild cases
Desmosterolosis6023989 cases3β-hydroxysterol-∆24-reductase (desmosterol)MultisystemicNo[25,37]
CDPX21302960≤ 1:4000003β-hydroxysteroid-∆8,∆7-sterol isomerase [8-dehydrocholesterol, cholesta-8(9)-en-3β-ol]Skin and skeletal systemsNo[25,38]
CHILD syndrome308050< 1:10000003β-hydroxysteroid dehydrogenase [4α-carboxymethylcholest-8(9)-en-3β-ol, 4α-monomethyl-and 4,4’-dimethylsterols]2Skin and skeletal systemsNo[25,39]
Lathosterolosis6073304 cases3β-hydroxysteroid-∆5-desaturase (lathosterol)MultisystemicYes[40-43]
Two cases
Antley-Bixler syndrome3201750> 100 casesLanosterol 14α-demethylase (lanosterol, dihydrolanosterol)2Skin and genital systemsNo[44,45]
Greenberg dysplasia21514011 casesSterol-∆14-reductase (cholesta-8,14-dien-3β-ol, cholesta-8,14,24-trien-3β-ol)2Skeletal systemNo[25,46]
CK syndrome30083113 cases3β-hydroxysteroid dehydrogenase (4α-monomethyl- and 4,4’-dimethylsterols)2Nervous systemNo[25,47,48]
SC4MOL deficiency6168345 casesSterol-C4-methyl oxidase (4α-monomethyl- and 4,4’-dimethylsterols)Skin and eyeYes[49,50,88]
SystemsOne case

Plasma sterol analysis by gas chromatography is a useful screening test for most of these disorders, even though a negative result in plasma does not exclude disorders such as CHILD, CK, and mild CDPX2 syndromes (Table 1).

The cholesterol plasma levels in severe cases of SLOS are very low (< 20 mg/dL), while in the patients with mild phenotype may have total cholesterol levels quite variables ranging from low (< 100 mg/dL) to normal levels[27]. In fact, the mild cases of SLOS can be treated using cholesterol supplementation and simvastatin[28]. Instead, all other defects, with the exception of lathosterolosis and SC4MOL, there are no treatments that correct or attenuate the metabolic condition. Therefore, they are cured with surgical or medical treatments needed to alleviate symptoms[25,45].

Lathosterolosis is a very rare disease with only four cases reported in literature[40-43,53], of which two patients survived[40,43]. In particular, the first described patient[40] showed a progressive intrahepatic cholestasis that had caused liver failure at seven years when she was subjected to liver transplantation (LT), which removed liver disease, corrected the defect in cholesterol metabolism, and also improved some neurologic symptoms[54,55].

In some conditions that influence cholesterol metabolism, LT acts as a gene therapy or as a healthy gene producer[18]. Furthermore, the normal gene expression of cholesterol biosynthesis and its physiological levels in the liver graft may influence intrahepatic availability of cholesterol, which is essential for liver-regenerative capacity[56].

Instead, the last case of lathosterolosis has been diagnosed in a 22 mo-old male with a mild clinical and biochemical phenotype. One month after diagnosis, the patient was treated with simvastatin and it resulted in normalization of lathosterol level and neurodevelopmental improvement[43].

SC4MOL deficiency is a recently discovered defect into the C-4 demethylase complex of cholesterol biosynthesis well described by He et al[49]. Until now, only five SC4MOL patients have been reported with a high variable phenotype[50]. The first patient presented the more severe phenotype characterized by a psoriasiform dermatitis, she was treated by oral statin and supplementation with cholesterol plus bile acids. After two years of treatment, the symptoms dramatically ameliorated and methylsterol levels normalized.

BILE ACIDS SYNTHESIS DEFECTS

Hereditary biosynthesis defects of biliary acids cause fatal liver disease during childhood and/or progressive neurological diseases that occur later in infancy or in the adult. Some of these diseases can be effectively treated by the administration of bile acids, so early diagnosis of these disorders is very important and life-saving. Recently, Clayton[26] and Heubi et al[35] reviewed the defects to date discovered in the pathway of bile acid synthesis, which are reported in Table 2[26,35,57-77].

Table 2 Inherited defects of bile acids biosynthesis in humans.
DisorderOMIM#FrequencyEnzyme (urine biomarkers)1Primary site of expressionTreatableRef.
CTX2137001:50000Sterol 27-hydroxylase (tetrahydroxy-, pentahydroxy- and hexahydroxy-bile alcohols)2Eye, central and peripheral nervous systemsYes[26,30,35]
CBAS160776573 cases3β-hydroxy-∆5-C27-steroid oxidoreductase (3β-hydroxy-∆5 bile acids)LiverYes[26,35,57,58]
CBAS223555541 cases∆4-3-oxosteroid 5β-reductase (∆4-3-oxo bile acids)LiverYes[35,59-65]
SPG5A27080031 casesOxysterol 7α-hydroxylase (27-hydroxycholesterol)3Central and peripheral nervous systemsNo[66-69]
FHCA60774815 casesBAAT4 (unconjugated cholic acid)3Liver and intestineYes[35,70,71]
CBAS42149506 casesα-methylacyl-CoA racemase (THCA)5Liver, intestine and peripheral nervous systemsYes[35,72,73]
CBAS36138123 casesoxysterol 7α-hydroxylase (3β-hydroxy-5-cholenoic and 3β-hydroxy-5-cholestenoic acids)LiverNo[35,74,75]
BACL deficiencyNR8 casesBile acid-CoA ligase (unconjugated cholic acid)3Liver and intestineNo[76]
CYP7A1 deficiencyNR< 1:1000000CYP7A1 (3β-hydroxy-5-cholenoic and 3β-hydroxy-5-cholestenoic acids, 27-hydroxycholesterol)6Cardiovascular systemNo[35,77]

The onset of these defects usually occurs in neonatal period or childhood with cholestasis, vitamin deficiency (fat-soluble), and with rickets, or hypoprothrombinemia, chronic liver disease or growth failure. Differently, the mean age of onset of symptoms for CTX is 19 years[26,30,35] and CYP7A1 deficiency usually present in adult life with hyperlipidemia[35,77], whereas SPG5A is characterized by a high variable age of onset (from 8 to 40 years of life) with a wide phenotypic spectrum (spastic paraplegia with or without additional manifestations, including optic atrophy or cerebellar ataxia)[66-69]. The defect in bile acid synthesis causes a reduction of hepatic conversion of cholesterol to cholic acid and chenodeoxycholic acid (CDCA)[78]. Canalicular bile flow is stimulated by the increase of bile acids synthesis, even though a bile salts-independent bile flow is always present as basal state. Therefore, the lack or the reduced synthesis of bile acids causes a more or less serious reduction of bile flow (cholestasis) and regurgitation in the blood stream of normally excreted compounds (i.e., conjugate bilirubin). In addition, the presence of bile acids promotes the release of gamma-glutamyl transpeptidase (γ-GT) from the canalicular membrane, while the absence of bile acids in the bile, except for some cases, does not result in increased plasma γ-GT. In addition, since bile acids act as detergent for lipid absorption in the intestine, congenital bile acid synthesis errors can lead to steatorrhea, growth delay, and deficiencies in fat-soluble vitamins.

Among these disorders, we focused on Cerebrotendinous Xanthomatosis (CTX, OMIM # 213700), the most common inborn error of bile acids synthesis. The clinical findings of CTX are tendon xanthomas, diarrhea, cataracts, neurological manifestations, such as polyneuropathy, pyramidal and/or cerebellar signs, intellectual disability, psychiatric disturbance, and seizures[30,79,80]. In spite of the xanthomas and premature atherosclerosis, CTX patients are usually normocholesterolemic[81]. In these patients the 25-hydroxylated C27-bile alcohols are abundantly excreted in the urine and other biological materials, the accumulation of these metabolites is due to the incomplete oxidation of the cholesterol side-chain[82]. Instead, the production of CDCA is markedly reduced and in face of almost normal production of cholic acid[83]. In addition, the reduced levels of bile acids decreased the negative feedback on two regulatory enzymes, the cholesterol 7-hydroxylase, the first rate-limiting enzyme in bile acid pathway, and the HMG-CoA reductase, which regulates cholesterol production. As a result, the increased activities of both enzymes lead to an accumulation of bile acid precursors, such as cholestanol and various bile alcohols, and of intermediates of cholesterol biosynthesis[84]. In fact plasma sterol analyses in CTX patients reveal elevated cholestanol and cholesterol precursors levels[30,81,85].

Treatment of CTX patients with CDCA by a negative feedback inhibits the 7α-hydroxylase and the production of toxic bile acid intermediates, and reduces the production of cholesterol by the liver[82]. Clinical and laboratory parameters are improved by long-term oral administration of CDCA and without toxic effects[79]. In particular, when initiated in childhood, CDCA therapy may arrest the progression of disease and prevent neurological deterioration. Instead, once neurological impairment is manifest, a poor response to CDCA treatment may be recorded[79]. CDCA and statins therapy inhibits cholesterol synthesis reducing cholesterol precursors and cholestanol levels in plasma, and can improve lipoprotein metabolism[84]. The efficacy of treatment with HMG-CoA reductase inhibitors alone is controversial, and some adverse effects such as hepatic dysfunction and rhabdomyolysis may be observed[79,80,84].

DIFFERENTIAL DIAGNOSIS OF CHOLESTEROL AND BILE ACID BIOSYNTHESIS DEFECTS

Some treatable defects of cholesterol and bile acid biosynthesis that share different clinical findings are reported in Table 3. Nevertheless, these disorders are characterized by the accumulation of specific biomarkers (Tables 1 and 2) in blood and/or urine of affected patients[25,26,86,87]. Hence, accurate identification and quantification of these metabolites are essential to address the diagnostic-therapeutic process of these defects.

Table 3 Clinical and laboratory findings shared by some treatable defects of cholesterol and bile acid biosynthesis.
Clinical featuresSLOS1LATHOSC4MOLCTXCBAS1CBAS2CBAS4Shared findings
MicrocephalyYesYesYesNoNoNoNo3/7
Congenital cataractsYesYesYesYesNoNoNo4/7
Intellectual disabilityYesYesYesYesNoNoNo4/7
Neurological diseaseNoNoNoYesNoNoYes2/7
Developmental delayYesYesYesNoNoNoNo3/7
CholestasisNoYesNoYesYesYesYes5/7
SteatosisNoYesNoNoNoYesNo2/7
ASTNormalHighNormalHighHighHighNormal4/7
ALTNormalHighNormalHighHighHighNormal4/7
γGTNormalHighNormalNormalNormalHighNormal2/7
Conjugated bilirubinNormalHighNormalHighHighHighNormal4/7
Total cholesterolVery low to normalLow to normalLow to normalNormal to highNormalNormalNormaln.a.
Fat-soluble vitaminNormalLowNormalNormalLowLowLow4/7
Diagnostic method
GC-MSSterols (p)Sterols (p)Sterols (p)Sterols (p)
LC-MS/MSHBA (u)BA (u)BA (u, p)BA (u, p)
TreatmentCholesterol plus statinsStatins or LTCholesterol plus statins and bile acidsCDCA plus statinsCholic acidUDCA or cholic acidCholic acid2

The sterol profile can be easily analyzed by gas chromatography coupled to mass spectrometry (GC-MS; Table 3) in plasma, dried blood spot, red blood cell membrane and tissue homogenates[25,27,31,33].

Furthermore, an useful screening test for many of bile acid synthesis defects is the analysis of bile acids and bile alcohols in urine, which is easily performed by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS; Table 3)[26,30,35].

CONCLUSION

In this editorial, we have briefly focused on the main clinical signs and laboratory findings of the inherited defects of cholesterol and bile acids synthesis. In summary, the liver and the intestine are the strategic organs in the control of cholesterol and bile acids in biological fluids. Moreover, the physiologic biosynthesis of cholesterol and bile acids is essential for the most important functions of our body, including embryonic development and neuronal functions. To date, as summarized here, in humans have been discovered nine inborn defects of cholesterol synthesis and nine of the bile acids. Although these diseases are rare and some ones very rare, it is commonly believed that their frequency has been underestimated, particularly in those patients affected by mild forms. This may be due to the heterogeneity of clinical and laboratory features of these diseases, which represent the pitfalls that mask their early diagnosis but, more important, it is the lost of all cases that are never diagnosed. Moreover, many of the defects reported here are treatable and with good results in most of them, therefore this fact should prompt us to pay more attention to the patient history, particularly that of childhood, with symptoms compatible with these defects.

Therefore, we hope that this editorial will stimulate the reader’s thinking to extend their knowledge and to take into account that today we have many clinical and laboratory tools to suspect and diagnose a rare disease including the defects of cholesterol and bile acids biosynthesis.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: Italy

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Schmidt HHJ S- Editor: Gong ZM L- Editor: A E- Editor: Li D

References
1.  Maxfield FR, van Meer G. Cholesterol, the central lipid of mammalian cells. Curr Opin Cell Biol. 2010;22:422-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 251]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
2.  Režen T. The impact of cholesterol and its metabolites on drug metabolism. Expert Opin Drug Metab Toxicol. 2011;7:387-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
3.  Rezen T, Rozman D, Pascussi JM, Monostory K. Interplay between cholesterol and drug metabolism. Biochim Biophys Acta. 2011;1814:146-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 53]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
4.  Groen AK, Bloks VW, Verkade H, Kuipers F. Cross-talk between liver and intestine in control of cholesterol and energy homeostasis. Mol Aspects Med. 2014;37:77-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
5.  Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343:425-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3933]  [Cited by in F6Publishing: 3912]  [Article Influence: 115.1]  [Reference Citation Analysis (0)]
6.  Sato R. Sterol metabolism and SREBP activation. Arch Biochem Biophys. 2010;501:177-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
7.  Zhao C, Dahlman-Wright K. Liver X receptor in cholesterol metabolism. J Endocrinol. 2010;204:233-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 307]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
8.  Go GW, Mani A. Low-density lipoprotein receptor (LDLR) family orchestrates cholesterol homeostasis. Yale J Biol Med. 2012;85:19-28.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Dietschy JM, Turley SD, Spady DK. Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans. J Lipid Res. 1993;34:1637-1659.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Turley SD, Spady DK, Dietschy JM. Role of liver in the synthesis of cholesterol and the clearance of low density lipoproteins in the cynomolgus monkey. J Lipid Res. 1995;36:67-79.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Gaylor JL. Membrane-bound enzymes of cholesterol synthesis from lanosterol. Biochem Biophys Res Commun. 2002;292:1139-1146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
12.  Lange BM, Rujan T, Martin W, Croteau R. Isoprenoid biosynthesis: the evolution of two ancient and distinct pathways across genomes. Proc Natl Acad Sci USA. 2000;97:13172-13177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 628]  [Cited by in F6Publishing: 496]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
13.  Wang DQ. Regulation of intestinal cholesterol absorption. Annu Rev Physiol. 2007;69:221-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 196]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
14.  Dikkers A, Tietge UJ. Biliary cholesterol secretion: more than a simple ABC. World J Gastroenterol. 2010;16:5936-5945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 38]  [Reference Citation Analysis (0)]
15.  van der Wulp MY, Verkade HJ, Groen AK. Regulation of cholesterol homeostasis. Mol Cell Endocrinol. 2013;368:1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 129]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
16.  Yoo EG. Sitosterolemia: a review and update of pathophysiology, clinical spectrum, diagnosis, and management. Ann Pediatr Endocrinol Metab. 2016;21:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
17.  Sudhop T, Lütjohann D, von Bergmann K. Sterol transporters: targets of natural sterols and new lipid lowering drugs. Pharmacol Ther. 2005;105:333-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 61]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
18.  Nemes K, Åberg F, Gylling H, Isoniemi H. Cholesterol metabolism in cholestatic liver disease and liver transplantation: From molecular mechanisms to clinical implications. World J Hepatol. 2016;8:924-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
19.  Degirolamo C, Sabbà C, Moschetta A. Intestinal nuclear receptors in HDL cholesterol metabolism. J Lipid Res. 2015;56:1262-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
20.  Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem. 2003;72:137-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1372]  [Cited by in F6Publishing: 1356]  [Article Influence: 64.6]  [Reference Citation Analysis (0)]
21.  Kannel WB, Castelli WP, Gordon T. Cholesterol in the prediction of atherosclerotic disease. New perspectives based on the Framingham study. Ann Intern Med. 1979;90:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Liu J, Sempos CT, Donahue RP, Dorn J, Trevisan M, Grundy SM. Non-high-density lipoprotein and very-low-density lipoprotein cholesterol and their risk predictive values in coronary heart disease. Am J Cardiol. 2006;98:1363-1368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 212]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
23.  Utermann G, Menzel HJ. Genetic disorders of lipoprotein metabolism. Principles and treatment of lipoprotein disorders. Berlin: Springer Science Business Media 2012; 89-130.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Ohashi K, Osuga J, Tozawa R, Kitamine T, Yagyu H, Sekiya M, Tomita S, Okazaki H, Tamura Y, Yahagi N. Early embryonic lethality caused by targeted disruption of the 3-hydroxy-3-methylglutaryl-CoA reductase gene. J Biol Chem. 2003;278:42936-42941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 82]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
25.  Herman GE, Kratz L. Disorders of sterol synthesis: beyond Smith-Lemli-Opitz syndrome. Am J Med Genet C Semin Med Genet. 2012;160C:301-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
26.  Clayton PT. Disorders of bile acid synthesis. J Inherit Metab Dis. 2011;34:593-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
27.  Corso G, Gelzo M, Barone R, Clericuzio S, Pianese P, Nappi A, Dello Russo A. Sterol profiles in plasma and erythrocyte membranes in patients with Smith-Lemli-Opitz syndrome: a six-year experience. Clin Chem Lab Med. 2011;49:2039-2046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
28.  DeBarber AE, Eroglu Y, Merkens LS, Pappu AS, Steiner RD. Smith-Lemli-Opitz syndrome. Expert Rev Mol Med. 2011;13:e24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
29.  Kanungo S, Soares N, He M, Steiner RD. Sterol metabolism disorders and neurodevelopment-an update. Dev Disabil Res Rev. 2013;17:197-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 57]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
30.  Di Taranto MD, Gelzo M, Giacobbe C, Gentile M, Marotta G, Savastano S, Dello Russo A, Fortunato G, Corso G. Cerebrotendinous xanthomatosis, a metabolic disease with different neurological signs: two case reports. Metab Brain Dis. 2016;31:1185-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
31.  Corso G, D’Apolito O, Gelzo M, Paglia G, Dello Russo A. A powerful couple in the future of clinical biochemistry: in situ analysis of dried blood spots by ambient mass spectrometry. Bioanalysis. 2010;2:1883-1891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
32.  Paglia G, D’Apolito O, Gelzo M, Dello Russo A, Corso G. Direct analysis of sterols from dried plasma/blood spots by an atmospheric pressure thermal desorption chemical ionization mass spectrometry (APTDCI-MS) method for a rapid screening of Smith-Lemli-Opitz syndrome. Analyst. 2010;135:789-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
33.  Gelzo M, Clericuzio S, Barone R, D’Apolito O, Dello Russo A, Corso G. A routine method for cholesterol and 7-dehydrocholesterol analysis in dried blood spot by GC-FID to diagnose the Smith-Lemli-Opitz syndrome. J Chromatogr B Analyt Technol Biomed Life Sci. 2012;907:154-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
34.  Gelzo M, Dello Russo A, Corso G. Stability study of dehydrocholesterols in dried spot of blood from patients with Smith-Lemli-Opitz syndrome, using filter-paper treated with butylated hydroxytoluene. Clin Chim Acta. 2012;413:525-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
35.  Heubi JE, Setchell KD, Bove KE. Inborn errors of bile acid metabolism. Semin Liver Dis. 2007;27:282-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 110]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
36.  Tint GS, Irons M, Elias ER, Batta AK, Frieden R, Chen TS, Salen G. Defective cholesterol biosynthesis associated with the Smith-Lemli-Opitz syndrome. N Engl J Med. 1994;330:107-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 624]  [Cited by in F6Publishing: 575]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
37.  FitzPatrick DR, Keeling JW, Evans MJ, Kan AE, Bell JE, Porteous ME, Mills K, Winter RM, Clayton PT. Clinical phenotype of desmosterolosis. Am J Med Genet. 1998;75:145-152.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Kelley RI, Wilcox WG, Smith M, Kratz LE, Moser A, Rimoin DS. Abnormal sterol metabolism in patients with Conradi-Hünermann-Happle syndrome and sporadic lethal chondrodysplasia punctata. Am J Med Genet. 1999;83:213-219.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Grange DK, Kratz LE, Braverman NE, Kelley RI. CHILD syndrome caused by deficiency of 3beta-hydroxysteroid-delta8, delta7-isomerase. Am J Med Genet. 2000;90:328-335.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Brunetti-Pierri N, Corso G, Rossi M, Ferrari P, Balli F, Rivasi F, Annunziata I, Ballabio A, Russo AD, Andria G. Lathosterolosis, a novel multiple-malformation/mental retardation syndrome due to deficiency of 3beta-hydroxysteroid-delta5-desaturase. Am J Hum Genet. 2002;71:952-958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 82]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
41.  Krakowiak PA, Wassif CA, Kratz L, Cozma D, Kovárová M, Harris G, Grinberg A, Yang Y, Hunter AG, Tsokos M. Lathosterolosis: an inborn error of human and murine cholesterol synthesis due to lathosterol 5-desaturase deficiency. Hum Mol Genet. 2003;12:1631-1641.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Rossi M, D’Armiento M, Parisi I, Ferrari P, Hall CM, Cervasio M, Rivasi F, Balli F, Vecchione R, Corso G. Clinical phenotype of lathosterolosis. Am J Med Genet A. 2007;143A:2371-2381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
43.  Ho AC, Fung CW, Siu TS, Ma OC, Lam CW, Tam S, Wong VC. Lathosterolosis: a disorder of cholesterol biosynthesis resembling smith-lemli-opitz syndrome. JIMD Rep. 2014;12:129-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
44.  Kelley RI, Kratz LE, Glaser RL, Netzloff ML, Wolf LM, Jabs EW. Abnormal sterol metabolism in a patient with Antley-Bixler syndrome and ambiguous genitalia. Am J Med Genet. 2002;110:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 73]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
45.  Porter FD, Herman GE. Malformation syndromes caused by disorders of cholesterol synthesis. J Lipid Res. 2011;52:6-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 322]  [Cited by in F6Publishing: 311]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
46.  Waterham HR, Koster J, Mooyer P, Noort Gv Gv, Kelley RI, Wilcox WR, Wanders RJ, Hennekam RC, Oosterwijk JC. Autosomal recessive HEM/Greenberg skeletal dysplasia is caused by 3 beta-hydroxysterol delta 14-reductase deficiency due to mutations in the lamin B receptor gene. Am J Hum Genet. 2003;72:1013-1017.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  du Souich C, Chou A, Yin J, Oh T, Nelson TN, Hurlburt J, Arbour L, Friedlander R, McGillivray BC, Tyshchenko N. Characterization of a new X-linked mental retardation syndrome with microcephaly, cortical malformation, and thin habitus. Am J Med Genet A. 2009;149A:2469-2478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
48.  McLarren KW, Severson TM, du Souich C, Stockton DW, Kratz LE, Cunningham D, Hendson G, Morin RD, Wu D, Paul JE. Hypomorphic temperature-sensitive alleles of NSDHL cause CK syndrome. Am J Hum Genet. 2010;87:905-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
49.  He M, Kratz LE, Michel JJ, Vallejo AN, Ferris L, Kelley RI, Hoover JJ, Jukic D, Gibson KM, Wolfe LA. Mutations in the human SC4MOL gene encoding a methyl sterol oxidase cause psoriasiform dermatitis, microcephaly, and developmental delay. J Clin Invest. 2011;121:976-984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
50.  He M, Smith LD, Chang R, Li X, Vockley J. The role of sterol-C4-methyl oxidase in epidermal biology. Biochim Biophys Acta. 2014;1841:331-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
51.  Arnold AW, Bruckner-Tuderman L, Has C, Happle R. Conradi-Hünermann-Happle syndrome in males vs MEND syndrome (male EBP disorder with neurological defects). Br J Dermatol. 2012;166:1309-1313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
52.  Flück CE, Tajima T, Pandey AV, Arlt W, Okuhara K, Verge CF, Jabs EW, Mendonça BB, Fujieda K, Miller WL. Mutant P450 oxidoreductase causes disordered steroidogenesis with and without Antley-Bixler syndrome. Nat Genet. 2004;36:228-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 310]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
53.  Parnes S, Hunter AG, Jimenez C, Carpenter BF, MacDonald I. Apparent Smith-Lemli-Opitz syndrome in a child with a previously undescribed form of mucolipidosis not involving the neurons. Am J Med Genet. 1990;35:397-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
54.  Rossi M, Vajro P, Iorio R, Battagliese A, Brunetti-Pierri N, Corso G, Di Rocco M, Ferrari P, Rivasi F, Vecchione R. Characterization of liver involvement in defects of cholesterol biosynthesis: long-term follow-up and review. Am J Med Genet A. 2005;132A:144-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
55.  Calvo PL, Brunati A, Spada M, Romagnoli R, Corso G, Parenti G, Rossi M, Baldi M, Carbonaro G, David E. Liver transplantation in defects of cholesterol biosynthesis: the case of lathosterolosis. Am J Transplant. 2014;14:960-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
56.  Ginanni Corradini S, Siciliano M, Parlati L, Molinaro A, Cantafora A, Poli E, Mennini G, Melandro F, Vestri AR, Merli M. Recipient perioperative cholesterolaemia and graft cholesterol metabolism gene expression predict liver transplant outcome. Liver Int. 2014;34:e290-e301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
57.  Schwarz M, Wright AC, Davis DL, Nazer H, Björkhem I, Russell DW. The bile acid synthetic gene 3beta-hydroxy-Delta(5)-C(27)-steroid oxidoreductase is mutated in progressive intrahepatic cholestasis. J Clin Invest. 2000;106:1175-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 86]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Cheng JB, Jacquemin E, Gerhardt M, Nazer H, Cresteil D, Heubi JE, Setchell KD, Russell DW. Molecular genetics of 3beta-hydroxy-Delta5-C27-steroid oxidoreductase deficiency in 16 patients with loss of bile acid synthesis and liver disease. J Clin Endocrinol Metab. 2003;88:1833-1841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 58]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
59.  Setchell KD, Suchy FJ, Welsh MB, Zimmer-Nechemias L, Heubi J, Balistreri WF. Delta 4-3-oxosteroid 5 beta-reductase deficiency described in identical twins with neonatal hepatitis. A new inborn error in bile acid synthesis. J Clin Invest. 1988;82:2148-2157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 167]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
60.  Daugherty CC, Setchell KD, Heubi JE, Balistreri WF. Resolution of liver biopsy alterations in three siblings with bile acid treatment of an inborn error of bile acid metabolism (delta 4-3-oxosteroid 5 beta-reductase deficiency). Hepatology. 1993;18:1096-1101.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Shneider BL, Setchell KD, Whitington PF, Neilson KA, Suchy FJ. Delta 4-3-oxosteroid 5 beta-reductase deficiency causing neonatal liver failure and hemochromatosis. J Pediatr. 1994;124:234-238.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Siafakas CG, Jonas MM, Perez-Atayde AR. Abnormal bile acid metabolism and neonatal hemochromatosis: a subset with poor prognosis. J Pediatr Gastroenterol Nutr. 1997;25:321-326.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Kimura A, Kondo KH, Okuda KI, Higashi S, Suzuki M, Kurosawa T, Tohma M, Inoue T, Nishiyori A, Yoshino M. Diagnosis of the first Japanese patient with 3-oxo-delta4-steroid 5beta-reductase deficiency by use of immunoblot analysis. Eur J Pediatr. 1998;157:386-390.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Clayton PT, Mills KA, Johnson AW, Barabino A, Marazzi MG. Delta 4-3-oxosteroid 5 beta-reductase deficiency: failure of ursodeoxycholic acid treatment and response to chenodeoxycholic acid plus cholic acid. Gut. 1996;38:623-628.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Lemonde HA, Custard EJ, Bouquet J, Duran M, Overmars H, Scambler PJ, Clayton PT. Mutations in SRD5B1 (AKR1D1), the gene encoding delta(4)-3-oxosteroid 5beta-reductase, in hepatitis and liver failure in infancy. Gut. 2003;52:1494-1499.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Tsaousidou MK, Ouahchi K, Warner TT, Yang Y, Simpson MA, Laing NG, Wilkinson PA, Madrid RE, Patel H, Hentati F. Sequence alterations within CYP7B1 implicate defective cholesterol homeostasis in motor-neuron degeneration. Am J Hum Genet. 2008;82:510-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 127]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
67.  Schüle R, Brandt E, Karle KN, Tsaousidou M, Klebe S, Klimpe S, Auer-Grumbach M, Crosby AH, Hübner CA, Schöls L. Analysis of CYP7B1 in non-consanguineous cases of hereditary spastic paraplegia. Neurogenetics. 2009;10:97-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
68.  Biancheri R, Ciccolella M, Rossi A, Tessa A, Cassandrini D, Minetti C, Santorelli FM. White matter lesions in spastic paraplegia with mutations in SPG5/CYP7B1. Neuromuscul Disord. 2009;19:62-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
69.  Criscuolo C, Filla A, Coppola G, Rinaldi C, Carbone R, Pinto S, Wang Q, de Leva MF, Salvatore E, Banfi S. Two novel CYP7B1 mutations in Italian families with SPG5: a clinical and genetic study. J Neurol. 2009;256:1252-1257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
70.  Carlton VE, Harris BZ, Puffenberger EG, Batta AK, Knisely AS, Robinson DL, Strauss KA, Shneider BL, Lim WA, Salen G. Complex inheritance of familial hypercholanemia with associated mutations in TJP2 and BAAT. Nat Genet. 2003;34:91-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 195]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
71.  Setchell KD, Heubi JE, Shah S, Lavine JE, Suskind D, Al-Edreesi M, Potter C, Russell DW, O’Connell NC, Wolfe B. Genetic defects in bile acid conjugation cause fat-soluble vitamin deficiency. Gastroenterology. 2013;144:945-955.e6; quiz e14-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
72.  Ferdinandusse S, Denis S, Clayton PT, Graham A, Rees JE, Allen JT, McLean BN, Brown AY, Vreken P, Waterham HR. Mutations in the gene encoding peroxisomal alpha-methylacyl-CoA racemase cause adult-onset sensory motor neuropathy. Nat Genet. 2000;24:188-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 152]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
73.  Setchell KD, Heubi JE, Bove KE, O’Connell NC, Brewsaugh T, Steinberg SJ, Moser A, Squires RH Jr. Liver disease caused by failure to racemize trihydroxycholestanoic acid: gene mutation and effect of bile acid therapy. Gastroenterology. 2003;124:217-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 122]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
74.  Setchell KD, Schwarz M, O’Connell NC, Lund EG, Davis DL, Lathe R, Thompson HR, Weslie Tyson R, Sokol RJ, Russell DW. Identification of a new inborn error in bile acid synthesis: mutation of the oxysterol 7alpha-hydroxylase gene causes severe neonatal liver disease. J Clin Invest. 1998;102:1690-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 285]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
75.  Ueki I, Kimura A, Nishiyori A, Chen HL, Takei H, Nittono H, Kurosawa T. Neonatal cholestatic liver disease in an Asian patient with a homozygous mutation in the oxysterol 7alpha-hydroxylase gene. J Pediatr Gastroenterol Nutr. 2008;46:465-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 50]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
76.  Chong CP, Mills PB, McClean P, Gissen P, Bruce C, Stahlschmidt J, Knisely AS, Clayton PT. Bile acid-CoA ligase deficiency--a new inborn error of bile acid metabolism. J Inherit Metab Dis. 2012;35:521-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
77.  Pullinger CR, Eng C, Salen G, Shefer S, Batta AK, Erickson SK, Verhagen A, Rivera CR, Mulvihill SJ, Malloy MJ. Human cholesterol 7alpha-hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. J Clin Invest. 2002;110:109-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 142]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
78.  Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem. 1991;266:7779-7783.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Mignarri A, Gallus GN, Dotti MT, Federico A. A suspicion index for early diagnosis and treatment of cerebrotendinous xanthomatosis. J Inherit Metab Dis. 2014;37:421-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
80.  Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis: a comprehensive review of pathogenesis, clinical manifestations, diagnosis, and management. Orphanet J Rare Dis. 2014;9:179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 183]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
81.  Björkhem I. Cerebrotendinous xanthomatosis. Curr Opin Lipidol. 2013;24:283-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
82.  Batta AK, Shefer S, Batta M, Salen G. Effect of chenodeoxycholic acid on biliary and urinary bile acids and bile alcohols in cerebrotendinous xanthomatosis; monitoring by high performance liquid chromatography. J Lipid Res. 1985;26:690-698.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Salen G, Shefer S, Tint GS, Nicolau G, Dayal B, Batta AK. Biosynthesis of bile acids in cerebrotendinous xanthomatosis. Relationship of bile acid pool sizes and synthesis rates to hydroxylations at C-12, C-25, and C-26. J Clin Invest. 1985;76:744-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
84.  Kuriyama M, Tokimura Y, Fujiyama J, Utatsu Y, Osame M. Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. J Neurol Sci. 1994;125:22-28.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  de Sain-van der Velden MG, Verrips A, Prinsen BH, de Barse M, Berger R, Visser G. Elevated cholesterol precursors other than cholestanol can also be a hallmark for CTX. J Inherit Metab Dis. 2008;31 Suppl 2:S387-S393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
86.  Kuksis A. Plasma non-cholesterol sterols. J Chromatogr A. 2001;935:203-236.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Waterham HR. Defects of cholesterol biosynthesis. FEBS Lett. 2006;580:5442-5449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 79]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
88.  Frisso G, Gelzo M, Procopio E, Sica C, Lenza MP, Dello Russo A, Donati MA, Salvatore F, Corso G. A rare case of sterol-C4-methyl oxidase deficiency in a young Italian male: Biochemical and molecular characterization. Mol Genet Metab. 2017;121:329-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]