Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Exp Med. Mar 20, 2024; 14(1): 87202
Published online Mar 20, 2024. doi: 10.5493/wjem.v14.i1.87202
Emerging significance of butyrylcholinesterase
Gumpeny R Sridhar, Department of Endocrinology and Diabetes, Endocrine and Diabetes Centre, Visakhapatnam 530002, Andhra Pradesh, India
Lakshmi Gumpeny, Department of Internal Medicine, Gayatri Vidya Parishad Institute of Healthcare and Medical Technology, Visakhapatnam 530048, Andhra Pradesh, India
ORCID number: Gumpeny R Sridhar (0000-0002-7446-1251); Lakshmi Gumpeny (0000-0002-1368-745X).
Author contributions: The two authors contributed equally to the writing of the manuscript.
Conflict-of-interest statement: Both authors declare that they have no conflict of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (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: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Gumpeny R Sridhar, FRCP, Adjunct Professor, Department of Endocrinology and Diabetes, Endocrine and Diabetes Centre, 15-12-15 Krishnanagar, Visakhapatnam 530002, Andhra Pradesh, India. sridharvizag@gmail.com
Received: July 28, 2023
Peer-review started: July 28, 2023
First decision: September 5, 2023
Revised: October 4, 2023
Accepted: January 5, 2024
Article in press: January 5, 2024
Published online: March 20, 2024
Processing time: 234 Days and 19.9 Hours

Abstract

Butyrylcholinesterase (BChE; EC 3.1.1.8), an enzyme structurally related to acetylcholinesterase, is widely distributed in the human body. It plays a role in the detoxification of chemicals such as succinylcholine, a muscle relaxant used in anesthetic practice. BChE is well-known due to variant forms of the enzyme with little or no hydrolytic activity which exist in some endogamous communities and result in prolonged apnea following the administration of succinylcholine. Its other functions include the ability to hydrolyze acetylcholine, the cholinergic neurotransmitter in the brain, when its primary hydrolytic enzyme, acetylcholinesterase, is absent. To assess its potential roles, BChE was studied in relation to insulin resistance, type 2 diabetes mellitus, cognition, hepatic disorders, cardiovascular and cerebrovascular diseases, and inflammatory conditions. Individuals who lack the enzyme activity of BChE are otherwise healthy, until they are given drugs hydrolyzed by this enzyme. Therefore, BChE is a candidate for the study of loss-of-function mutations in humans. Studying individuals with variant forms of BChE can provide insights into whether they are protected against metabolic diseases. The potential utility of the enzyme as a biomarker for Alzheimer’s disease and the response to its drug treatment can also be assessed.

Key Words: Esterase, Acetylcholinesterase, Variant, Cholinergic, Metabolic syndrome, Cognition, Knockout model

Core Tip: Butyrylcholinesterase (BChE), a hepatic enzyme, hydrolyzes the muscle relaxant succinylcholine. Individuals with variant forms of the enzyme are healthy until they are administered succinylcholine during anesthesia. The enzyme may have regulatory roles in lipid metabolism, cholinergic response, and Alzheimer’s disease. People with variant forms of the enzyme are natural human knockout models and can be followed up to study the metabolic impact of harboring variant forms of BChE.



INTRODUCTION

Butyrylcholinesterase (BChE), belonging to the esterase group of enzymes, is a part of the serine hydrolase superfamily[1,2]. Esterases hydrolyze compounds that contain ester, amide, and thioester bonds[1]. BChE (EC 3.1.1.8) and acetylcholinesterase (AChE, EC 3.1.1.7) share a similar three-dimensional structure[3]. BChE is believed to have resulted from a duplication of an ancestral AChE gene[4]. AChE is responsible for the hydrolysis of acetylcholine at the neuromuscular junction. The roles of BChE are less well-defined: It hydrolyzes succinylcholine and bambuterol, which are used as muscle relaxants in anesthesiology[1].

EVOLUTIONARY ASPECTS AND CHEMISTRY

The BChE gene (HGNC: 983; MIM: 177100) exists across life forms[5] including invertebrates[6]. The concentrations of BChE exceed those of AChE in most tissues except the brain and muscle[3]. In the AChE knockout mouse model, BChE can compensate for the lack of AChE[7,8]. A convergent evolutionary mechanism is believed to have occurred between AChE and BChE[6].

BChE, which is mapped on chromosome 3 (3q26), exists in four molecular forms in plasma. The tetrameric form comprises nearly 90% of total plasma cholinesterase activity[2]. In a monomer, it consists of a common α/β hydrolase fold, flanked by α helices. The active site gorge volume of BChE is larger than that of AChE[9] and is shaped like a bowl[2]. AChE, though, has nearly 40% more aromatic residues. The active gorge consists of an acylation site for catalysis and pockets for choline-binding. It is rimmed by a peripheral anionic site. The catalytic activity of BChE depends on H-bond stabilization. In simulation studies, inhibitors were shown to reach the catalytic cavity due to the flexible entrance of the gorge[10].

TISSUE DISTRIBUTION

BChE is found in the lungs, plasma, brain, and heart. The highest levels of BChE mRNA are found in the liver, followed by the lung and the brain[4], where it is present at neuromuscular junctions. BChE is also expressed in brain astrocytes. The close association of neurons and glia has been termed the ‘tripartite synapse’, whereby glia exchange information with neurons[11].

BIOLOGICAL ROLES OF BUTYRYLCHOLINESTERASE

The role of BuChE in anaesthetic practice is well-recognized as a degrading enzyme of neuromuscular blockers succinylcholine and mivarurium[12]. Mutant forms of the enzyme have low or absent activity, resulting in prolonged apnea with the use of these muscle relaxants.

BChE levels are low in systemic conditions like liver disease, renal disease, malnutrition, malignancies, and burns[13].

It participates in the first-phase detoxification reaction against natural and exogenous toxins[14] (Table 1).

Table 1 Potential roles of butyrylcholinesterase.
Known functionsMetabolism of drugs and toxins (e.g., succinylcholine, carbamates, glucoalkaloids)
Affected by dietary fats
Influences the expression of metabolic syndrome, via action on lipids
Associations and predictors of outcomes in disease statesNutritional status
Hepatic disorders
Cardiovascular diseaseAcute coronary syndrome
Acute myocardial infarction
Injuries to brain and cerebrovascular and diseaseIschemic brain stroke
Traumatic brain injury
Alzheimer’s diseasePredisposition
Onset
Response to anticholinesterase medications
Pre-eclampsia
Inflammatory and infectionsSepsis
HIV infection
Hansen’s disease
Other conditionsWilsons disease
Chronic obstructive pulmonary disease

In the brain, BChE is present in the glial cells near the hippocampus and amygdala[15]. It may interact with ghrelin in the brain; in mice, increased expression of BChE is associated with low blood levels of ghrelin and reduced aggressive behavior[16].

ASSOCIATION OF BUTYRYLCHOLINESTERASE AND ENVIRONMENTAL TOXINS

BChE activity is principally found in ‘tissues of first contact’, such as the lungs, liver, skin, and blood. It plays a role in the activation of pro-drugs as well as in metabolizing drugs to inactive forms[17]. The biotransformation ability changes due to alterations in the macromolecular structure of the enzyme[17].

Other potential toxins that are inactivated by BChE include carbamates and plants containing glucoalkaloids[18,19].

To derive reference ranges of AChE and BChE, their levels were measured in 387 young and healthy individuals (201 men and 186 women aged between 18 and 45 years)[18], which is useful for comparison in pathological states[20].

Gene polymorphisms of BChE were studied in relation to groundwater fluoride toxicity[21]. In clinically healthy adults from Pakistan, fluorosis was associated with elevated BChE activity[21].

BUTYRYLCHOLINESTERASE, METABOLIC SYNDROME, AND DIABETES MELLITUS

BChE activity in rats was influenced by dietary fat[22], perhaps due to increased release from hepatocytes[23]. Among subjects with type 2 diabetes from southern India, the plasma levels of BChE were inversely related to serum cholesterol (P < 0.05)[24]. The enzyme may not directly cause metabolic syndrome but may serve as a marker for this condition[25].

Similar associations were reported between BChE activity and weight in children[26]. Plasma BChE was proposed as a marker of chronic low-grade inflammation[27]. In Japanese subjects (171 with type 2 diabetes and 88 controls), serum BChE correlated with adiposity, serum lipids, and HOMA-R[28]. To assess the risk of mortality with the levels of BChE, 813 subjects were followed up from 1985-1987 to 1996. Those in the lowest quintile of BChE activity had higher mortality[29]. Body mass index mediates changes in BChE activity in healthy young men and women (age: 18-25 years)[30].

There are other interesting observations. Extracts from the fruit melanocarpa affected BChE activity[31]. Elevated BChE levels predicted the development of type 2 diabetes[32] and its vascular complication[33]. Increased levels of BChE may be associated with lower AChE levels, which play an anti-inflammatory role[34]. Exposure to BChE protects cultured pancreatic cells by reduced formation of toxic amylin oligomer[35].

BUTYRYLCHOLINESTERASE AND COGNITION

Alzheimer’s disease (AD) that often accompanies aging is the most common cause of cognitive decline[36]. According to the cholinergic hypothesis, degeneration of cholinergic neurons in the basal forebrain results in cognitive dysfunction in AD[37]. Other conditions that occur via the cholinergic system include amyloid deposition, tau phosphorylation, neuroinflammation, and vascular damage (Pozzi et al[38], 2022). Acetylcholine, the neurochemical transmitter in the cholinergic synapses, is inactivated by AChE, and to a lesser extent by BuChE. The cholinergic system is part of the cholinergic anti-inflammatory pathway. The cholinergic hypothesis received additional support by the finding that cholinesterase inhibitor drugs which increase acetylcholinesterase at the synaptic cleft are effective in the management of AD[39].

Apart from its role in the cholinergic hypothesis, BuChE has been implicated in the deposition of amyloid. Amyloid hypothesis proposes that abnormal folding of β-amyloid protein may contribute to the pathogenesis of AD[40]. A variant form of the enzyme, called K-variant (Ala567Thr (A539T), may act synergistically with others such as the ε4 allele of apolipoprotein E and iron as a risk factor for AD[41,42].

BUTYRYLCHOLINESTERASE IN RELATION TO OTHER CONDITIONS
Liver disorders and malnutrition

BChE measurement is sometimes included in the panel of liver function tests due to its hepatic origin. It is an indicator of acute hepatitis or cirrhosis of the liver[14].

BChE levels are altered by inflammatory processes: They are low in acute inflammation and normalize once inflammation resolves[43]. BChE levels were low in malnourished children and in subjects with visceral undernutrition[44,45]. It can be used as a marker of nutritional status among the elderly[46].

Coronary artery disease

Acute coronary syndrome encompasses a range of conditions from angina pectoris to irreversible damage of the myocardium. BChE levels could differentiate healthy subjects from those with acute myocardial infarction (AMI). BChE activity was lower in acute myocardial infarction (AMI) (n = 85) compared with controls (n = 45) (P < 0.001)[47,48]. Similar observations were reported by Sulzgruber et al[49] in 2015. Higher BChE levels were associated with greater mortality-free survival in acute coronary syndrome. The strongest effect was observed among people aged 45-65 years. Similar findings of mortality were reported in subjects undergoing veno-arterial extracorporeal membrane oxygenation treatment after cardiac surgery[50].

Disorders of the brain

In ischemic brain stroke, BChE levels were measured in 33 subjects with acute ischemic stroke within 12 h of onset and in 29 controls. Stroke subjects had lower BChE activity compared to controls[51].

Among 188 patients with traumatic brain injury within 72 h of injury, non-survivors (n = 42; 22.3%) had lower levels of BChE activity[52]; they had an acute decrease of enzyme activity.

Pre-eclampsia

Pre-eclampsia, which occurs in pregnancy, is characterized by hypertension, proteinuria, and other maternal-related dysfunctions. BChE levels were measured in 198 unrelated women having pre-eclampsia and 101 unrelated women with normal pregnancy. Pre-eclampsia was associated with lower BChE activity[53].

Sepsis

Sepsis, presenting with acute organ dysfunction, is a common cause of mortality in the intensive care setting. To identify the severity of sepsis, BChE levels were used as a biomarker. Those who died within 90 d of admission had lower levels of BChE. Admission levels of the enzyme could predict those who survived 90 d[54]. Measurement of BChE could complement other ways of predicting the outcome of patients admitted in intensive care units. Using a newer definition of sepsis, ‘life-threatening organ dysfunction due to a dysregulated host response to infection’, Peng et al[55] showed that lower levels of BChE activity are an independent risk factor for the 30-d death rate in sepsis-3 patients.

Infections

The 6-mo outcome of subjects receiving highly active antiretroviral therapy for HIV infection was assessed in relation to the levels of BChE. Low levels of BChE were seen in 25.5% (129/505) of subjects with infection. In the first year, 16.6% of patients died (n = 84). Low BChE levels were associated with a survival of (64.5 +/- 4.5)% at one year compared to (87.6 +/- 1.8)% in those with normal levels[56].

In Hansen’s disease, genotyping of an atypical BChE allele (70G; rs1799807) and five additional single nucleotide polymorphisms (SNPs) reported higher allele (70G) and genotypic (70DG) frequencies in rs1799807. Atypical variants of the enzyme could predispose to infection[57], by interfering with the inflammatory response against the infective agent. Similarly, children with foot and mouth disease caused by enterovirus 71 infection had increased BChE levels[58].

Fertility

BChE was measured in idiopathic unexplained infertility, a day before and a day after intrauterine insemination. A positive correlation between BChE levels and total antioxidant activity on the day before the procedure was observed[59].

Other conditions

BChE was measured in untreated Wilson’s disease and in chronic obstructive pulmonary disease. Pilot studies showed that along with ceruloplasmin, BChE could be used as a biomarker in Wilson’s disease[60]. In chronic obstructive pulmonary disease (n = 153), BChE levels were elevated[61].

Studies in animals

Dogs with hypercortisolism had elevated serum BChE activity, related either to a direct effect of glucocorticoids or to changes in lipid metabolism associated with hypercortisolism[62]. Elevated salivary levels of BChE were reported in dogs with parvovirus infections[63].

The underlying pathogenic mechanism in all these disparate conditions appears to involve dysregulation of the inflammatory response leading to adverse outcome.

Cholinergic control of inflammation

Inflammation is part of a physiological response that is protective against noxious environmental factors. In a recent review, Medzhitov[64] proposed that inflammation ensures that homeostasis is maintained, and tissues retain their functional and structural integrity. It is regulated by the immune system, hormones, and neural signals.

The vagus nerve conveys information from the brain to attenuate the inflammatory process. It integrates signals from the hypothalamic-pituitary-adrenal axis and through the cholinergic anti-inflammatory pathway[65]. Bonaz et al[66] proposed that the anti-inflammatory properties of the vagus nerve may suggest the therapeutic implications of stimulating the nerve.

BChE may also influence the outcomes of coronavirus disease 2019 (COVID-19) via its effect on chronic low-grade inflammation. It could also serve as a biomarker for COVID-19 outcome; subjects with COVID-19 may be studied in subjects with variant forms of BChE[34,64].

BChE activity was predictive of 28-d mortality in critically ill COVID-19 patients[67]. A recent report indicates higher mortality in subjects with low or declining levels of serum BChE during hospitalization[68].

KNOCKOUT ANIMAL MODELS TO ELUCIDATE FUNCTION OF GENES

In functional genomics, gene knockout animal models are used to determine the function of genes. When a specific gene is inactivated, the resultant phenotype can provide information about its function[69]. Humans with loss-of-function genes give better insights than animal models[70]. The differences may relate to the other gene regulators upstream or downstream as well as environmental factors[71].

MacArthur et al[72] reported that a healthy person has an average of 100 inactivated genes, of which 20 are homozygous. A whole-exome sequencing study among European populations (n = 1432) reported that of loss-of-function mutations, nearly 45% (n = 76) were newly identified[73]. Narasimhan et al[74] studied the effects of rare gene knockouts in adults born of consanguineous marriage. Exome sequencing data in 3222 adults of Pakistani origin domiciled in Britain were linked to their lifelong health records. They did not find any relationship between those with loss-of-function genes and their consultation for health issues or prescription medication use. The latter two were taken as surrogate markers for their state of health.

Loss-of-function mutations can result from: (1) Nonsense SNPs leading to a premature stop codon, producing a truncated protein sequence; (2) splicing can be affected by an SNP at a canonical splice site; (3) an insertion or deletion variant located in the gene coding region can disrupt the full-length transcript leading to frameshifts; and (4) loss-of-function mutations can arise from the loss of an initiation codon[75].

Individuals with loss-of-function mutations who are apparently healthy were referred to as ‘experiments of nature’. Studying them could help in the search for new drug targets and in identifying or exploring whether such mutated genes could have beneficial effects[76].

The nascent field of studying natural human knockouts and the genotype-phenotype correlation can provide insights into population genetics and the evolution of genes[69,71].

Butyrylcholinesterase and its variants qualify as natural human knockouts: Other than prolonged apnea following exposure to succinylcholine, individuals with variant forms of BChE are apparently healthy[77].

SIGNIFICANCE OF STUDYING BUTYRYLCHOLINESTERASE VARIANTS

Unlike other gene knockout animal models, variants of BChE have a high prevalence in isolated ethnic groups: Mainly south Indian from the Vysya community, and certain Eskimos in western Alaska[34]. Li et al[78] developed an animal BChE gene knockout model to test drug toxicity. The model had a normal phenotype unless exposed to the drug. Altered cognitive functions were associated with normal nicotinic receptor function, though the muscarinic receptor function was altered in the knockout model. Preliminary studies on the effect of (R)-bambuterol, a specific and reversible inhibitor of BChE, suggested that it may be used in the treatment of early cognitive decline[79].

BUTYRYLCHOLINESTERASE VARIANTS IN HUMANS

Lockridge et al reviewed the naturally occurring genetic variants of BChE[80]. Thirty-four loss-of-function mutations were identified; all of them were tolerated, meaning that having a nonfunctional gene was compatible with life. Humans harboring silent BChE genes are healthy and fertile[77]. Lando et al[81] reported that among healthy blood donors (n = 2609), 59 had low plasma BChE activity.

In the Netherlands Organisation for Applied Research Prins Mauritis Laboratory and Centers for Disease Control and Prevention, the frequency of BChE mutations was 9 out of 121000 alleles. Some of the mutants resulted in a complete absence of enzyme activity[80].

The commonest missense mutation, the K-variant [Ala567Thr (AS39T)], is associated with a 30% lower BChE plasma activity compared to native BChE. It is due to an unknown mutation in a regulatory region[81]. Other variants are less common except in communities such as south Indian Vysyas or Eskimos, where genotyping is not possible. Estimating dibucaine and fluoride numbers could serve as a surrogate. Family studies in the south Indian state of Andhra Pradesh showed various phenotypic forms of BChE deficiency in inbred families (oral presentation at the 12th International Meeting on Cholinesterases-Sixth International Conference on Paraoxonases at Elche (Alicante, Spain) in 2015: GR Sridhar, G Nirmala, Premlata S, Satyanarayana M. Variant butyrylcholinesterase in South India (Figures 1-4).

Figure 1
Figure 1 Phenotype of variant butyrylcholinesterase, families 1-4.
Figure 2
Figure 2 Phenotype of variant butyrylcholinesterase, families 5-8.
Figure 3
Figure 3 Phenotype of variant butyrylcholinesterase, families 9-11. BChE: Butyrylcholinesterase.
Figure 4
Figure 4 Consanguinity (=) in families 7, 8, 9, and 10.

BChE activity increases with the progression of AD and may eventually replace the function of AChE[82]. Individuals with the BChE-K variant could have deleterious outcomes when donepezil is given to patients with mild cognitive impairment. It can therefore serve as a pharmacogenetic marker in the choice of drugs for cognitive impairment[82].

The reasons why non-functional proteins persisted include: (1) The active enzyme can compensate for the absent enzyme; (2) it might have acquired new beneficial functions; and (3) the enzyme may be involved in a pathway regulated by other molecules or enzyme that can substitute for it.

As mentioned earlier, succinylcholine is hydrolyzed by BChE. Administration of succinylcholine to a homozymous knockout mouse model for BChE [BChE-/-] resulted in rapid death; heterozygous model [BChE+/-] had less severe manifestations and recovered within 30 min[78]. AChE-/- animals suffered greater toxicity to succinylcholine exposure than BChE-/- mice.

BChE is a natural drug target in which drug side effects can be minimized by knocking out its gene[83]. Knowledge from knockout models could be used to assess the effects of drugs such as donepezil in cognitive impairment.

CONCLUSION

BChE serves a critical role in the hydrolysis of esters. Unlike AChE with which it shares structural and functional properties, BChE acts on a broader number of substrates, but has lower catalytic efficiency on acetylcholine[83]. Novel ligands and mutants of BChE were developed for use in the treatment of cocaine toxicity and neurological diseases[84]. Its roles extend into cardiovascular health; recent clinical studies suggest a correlation between increased plasma BChE activity and longevity in patients with severe cardiovascular disease[16].

BChE also serves as a prognostic marker for liver and non-liver diseases, protein-energy malnutrition, and obesity by reflecting the availability of amino acidic substrates[46]. It is also involved in neurodegenerative disorders, particularly AD. BChE not only co-regulates cholinergic transmission by hydrolyzing acetylcholine alongside AChE, but potentially interferes with the course of AD. Inhibitors of BChE are therefore used in the treatment of AD and other disorders of cognition by ameliorating cholinergic deficiency[85].

Recent findings show that BChE regulates substrates such as cocaine and ghrelin. Recombinant BChE mutants and viral gene therapy are being developed against cocaine addiction, and in exploring the role of BChE in obesity[86] (Table 2).

Table 2 Potential areas for studies in subjects with variant forms of butyrylcholinesterrase and therapeutic potential of the enzyme.
Follow up of asymptomatic individuals for any protective or predictive role in cardiovascular disease and Alzheimer’s disease
Production from transgenic sources as a pharmacologic agent
Potential drug target

As a therapeutic agent, phase I trials showed the safety of pure BChE, thereby giving an option in preventing nerve agent toxicity. Gene therapy using vectors that allow long-term expression of BChE after a single injection is being explored[87].

Animal studies have illustrated that pretreatment with BChE can prevent adverse effects from lethal doses of nerve agents like soman, sarin, and VX[88]. Other technologies employed to synthesise BChE include glycosylation and PEGylation that can enhance its pharmacokinetics[86].

Not all loss-of-function gene variants manifest in the same way; they may range from being mildly deleterious to neutral and sometimes, even advantageous[89]: In populations where consanguinity and Mendelian disorders are common, population-wide rapid exome sequencing may be beneficial[90,91].

BChE's multi-faceted nature, as a pharmacological target and tool, deepens our understanding of biological pathways in health and disease. Further phenotype-genotype studies will throw light on its potential effects. In this context, subjects with variant forms of BChE serve as critical comparators in such studies.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Emran TB, Bangladesh; Teixeira KN, Brazil S-Editor: Liu JH L-Editor: Wang TQ P-Editor: Yu HG

References
1.  Fukami T, Yokoi T. The emerging role of human esterases. Drug Metab Pharmacokinet. 2012;27:466-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 154]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
2.  Masson P, Carletti E, Nachon F. Structure, activities and biomedical applications of human butyrylcholinesterase. Protein Pept Lett. 2009;16:1215-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
3.  Li B, Stribley JA, Ticu A, Xie W, Schopfer LM, Hammond P, Brimijoin S, Hinrichs SH, Lockridge O. Abundant tissue butyrylcholinesterase and its possible function in the acetylcholinesterase knockout mouse. J Neurochem. 2000;75:1320-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 249]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
4.  Johnson G, Moore SW. Why has butyrylcholinesterase been retained? Structural and functional diversification in a duplicated gene. Neurochem Int. 2012;61:783-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
5.  Sridhar GR, Lakshmi PV, Rao AA. Phylogenetic tree construction of butyrylcholinesterase sequences in life forms. J Assoc Physicians India. 2006;54:122-123.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Pezzementi L, Nachon F, Chatonnet A. Evolution of acetylcholinesterase and butyrylcholinesterase in the vertebrates: an atypical butyrylcholinesterase from the Medaka Oryzias latipes. PLoS One. 2011;6:e17396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
7.  Masson P, Lockridge O. Butyrylcholinesterase for protection from organophosphorus poisons: catalytic complexities and hysteretic behavior. Arch Biochem Biophys. 2010;494:107-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 163]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
8.  Mesulam MM, Guillozet A, Shaw P, Levey A, Duysen EG, Lockridge O. Acetylcholinesterase knockouts establish central cholinergic pathways and can use butyrylcholinesterase to hydrolyze acetylcholine. Neuroscience. 2002;110:627-639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 464]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
9.  Masson P, Lushchekina S, Schopfer LM, Lockridge O. Effects of viscosity and osmotic stress on the reaction of human butyrylcholinesterase with cresyl saligenin phosphate, a toxicant related to aerotoxic syndrome: kinetic and molecular dynamics studies. Biochem J. 2013;454:387-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
10.  De Boer D, Nguyen N, Mao J, Moore J, Sorin EJ. A Comprehensive Review of Cholinesterase Modeling and Simulation. Biomolecules. 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
11.  Perea G, Navarrete M, Araque A. Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci. 2009;32:421-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1045]  [Cited by in F6Publishing: 1126]  [Article Influence: 75.1]  [Reference Citation Analysis (0)]
12.  Delacour H, Dedome E, Courcelle S, Hary B, Ceppa F. Butyrylcholinesterase deficiency. Ann Biol Clin (Paris). 2016;74:279-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
13.  Andersson ML, Møller AM, Wildgaard K. Butyrylcholinesterase deficiency and its clinical importance in anaesthesia: a systematic review. Anaesthesia. 2019;74:518-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
14.  Pohanka M. Butyrylcholinesterase as a biochemical marker. Bratisl Lek Listy. 2013;114:726-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
15.  Darvesh S, Grantham DL, Hopkins DA. Distribution of butyrylcholinesterase in the human amygdala and hippocampal formation. J Comp Neurol. 1998;393:374-390.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Brimijoin S, Chen VP, Pang YP, Geng L, Gao Y. Physiological roles for butyrylcholinesterase: A BChE-ghrelin axis. Chem Biol Interact. 2016;259:271-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 64]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
17.  Sisková K, Bilka F, Adameová A, Balazová A, Mydla M, Pauliková I. Influence of lipid imbalance on butyrylcholinesterase activity and biotransformation efficiency. Pharmazie. 2012;67:345-350.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Nielsen SD, Schmidt JM, Kristiansen GH, Dalsgaard TK, Larsen LB. Liquid Chromatography Mass Spectrometry Quantification of α-solanine, α-chaconine, and Solanidine in Potato Protein Isolates. Foods. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
19.  McGehee DS, Krasowski MD, Fung DL, Wilson B, Gronert GA, Moss J. Cholinesterase inhibition by potato glycoalkaloids slows mivacurium metabolism. Anesthesiology. 2000;93:510-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
20.  Karasova JZ, Maderycova Z, Tumova M, Jun D, Rehacek V, Kuca K, Misik J. Activity of cholinesterases in a young and healthy middle-European population: Relevance for toxicology, pharmacology and clinical praxis. Toxicol Lett. 2017;277:24-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
21.  Bibi S, Habib R, Shafiq S, Abbas SS, Khan S, Eqani SAMAS, Nepovimova E, Khan MS, Kuca K, Nurulain SM. Influence of the chronic groundwater fluoride consumption on cholinergic enzymes, ACHE and BCHE gene SNPs and pro-inflammatory cytokines: A study with Pakistani population groups. Sci Total Environ. 2023;880:163359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Van Lith HA, Haller M, Van Tintelen G, Van Zutphen LF, Beynen AC. Plasma esterase-1 (ES-1) activity in rats is influenced by the amount and type of dietary fat, and butyryl cholinesterase activity by the type of dietary fat. J Nutr. 1992;122:2109-2120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
23.  Van Lith HA, Haller M, Van Tintelen G, Lemmens AG, Van Zutphen LF, Beynen AC. Fat intake and clofibrate administration have interrelated effects on liver cholesterol concentration and serum butyryl cholinesterase activity in rats. J Nutr. 1992;122:2283-2291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
24.  Sridhar GR, Nirmala G, Apparao A, Madhavi AS, Sreelatha S, Rani JS, Vijayalakshmi P. Serum butyrylcholinesterase in type 2 diabetes mellitus: a biochemical and bioinformatics approach. Lipids Health Dis. 2005;4:18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
25.  Han Y, Ma Y, Liu Y, Zhao Z, Zhen S, Yang X, Xu Z, Wen D. Plasma cholinesterase is associated with Chinese adolescent overweight or obesity and metabolic syndrome prediction. Diabetes Metab Syndr Obes. 2019;12:685-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
26.  Rodríguez-Fuentes GA, Arcega-Cabrera Fl, Fargher LF. Plasma and erythrocyte cholinesterase activities in children from Yucatan, Mexico: relationship with anthropometry and obesity. Asian J Pharm Clin Res. 2015;8:224-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Rao AA, Sridhar GR, Das UN. Elevated butyrylcholinesterase and acetylcholinesterase may predict the development of type 2 diabetes mellitus and Alzheimer's disease. Med Hypotheses. 2007;69:1272-1276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 112]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
28.  Iwasaki T, Yoneda M, Nakajima A, Terauchi Y. Serum butyrylcholinesterase is strongly associated with adiposity, the serum lipid profile and insulin resistance. Intern Med. 2007;46:1633-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 102]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
29.  Calderon-Margalit R, Adler B, Abramson JH, Gofin J, Kark JD. Butyrylcholinesterase activity, cardiovascular risk factors, and mortality in middle-aged and elderly men and women in Jerusalem. Clin Chem. 2006;52:845-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
30.  Stojanov M, Stefanović A, Džingalašević G, Mandić-Radić S, Prostran M. Butyrylcholinesterase activity in young men and women: association with cardiovascular risk factors. Clin Biochem. 2011;44:623-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
31.  Duchnowicz P, Ziobro A, Rapacka E, Koter-Michalak M, Bukowska B. Changes in Cholinesterase Activity in Blood of Adolescent with Metabolic Syndrome after Supplementation with Extract from Aronia melanocarpa. Biomed Res Int. 2018;2018:5670145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
32.  Sato KK, Hayashi T, Maeda I, Koh H, Harita N, Uehara S, Onishi Y, Oue K, Nakamura Y, Endo G, Kambe H, Fukuda K. Serum butyrylcholinesterase and the risk of future type 2 diabetes: the Kansai Healthcare Study. Clin Endocrinol (Oxf). 2014;80:362-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
33.  Yu R, Ye X, Wang X, Wu Q, Jia L, Dong K, Zhu Z, Bao Y, Hou X, Jia W. Serum cholinesterase is associated with incident diabetic retinopathy: the Shanghai Nicheng cohort study. Nutr Metab (Lond). 2023;20:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Sridhar GR. Butyrylcholinesterase, variants and metabolic syndrome. Adipobiology. 2018;9:19-27.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Shenhar-Tsarfaty S, Bruck T, Bennett ER, Bravman T, Aassayag EB, Waiskopf N, Rogowski O, Bornstein N, Berliner S, Soreq H. Butyrylcholinesterase interactions with amylin may protect pancreatic cells in metabolic syndrome. J Cell Mol Med. 2011;15:1747-1756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
36.  Sridhar GR, Lakshmi G, Nagamani G. Emerging links between type 2 diabetes and Alzheimer's disease. World J Diabetes. 2015;6:744-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 73]  [Cited by in F6Publishing: 66]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
37.  Jasiecki J, Wasąg B. Butyrylcholinesterase Protein Ends in the Pathogenesis of Alzheimer's Disease-Could BCHE Genotyping Be Helpful in Alzheimer's Therapy? Biomolecules. 2019;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
38.  Pozzi FE, Conti E, Appollonio I, Ferrarese C, Tremolizzo L. Predictors of response to acetylcholinesterase inhibitors in dementia: A systematic review. Front Neurosci. 2022;16:998224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Reference Citation Analysis (0)]
39.  Sridhar GR. Acetylcholinesterase inhibitors (Galantamine, Rivastigmine, and Donepezil). In P. Riederer, G. Laux et al (eds.), NeuroPsychopharmacotherapy. Springer Nature Switzerland AG. 20211;.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Reid GA, Darvesh S. Butyrylcholinesterase-knockout reduces brain deposition of fibrillar β-amyloid in an Alzheimer mouse model. Neuroscience. 2015;298:424-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 105]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
41.  Ratis RC, Dacoregio MI, Simão-Silva DP, Mateus RP, Machado LPB, Bonini JS, da Silva WCFN. Confirmed Synergy Between the ɛ4 Allele of Apolipoprotein E and the Variant K of Butyrylcholinesterase as a Risk Factor for Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis Rep. 2023;7:613-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
42.  Jasiecki J, Targońska M, Wasąg B. The Role of Butyrylcholinesterase and Iron in the Regulation of Cholinergic Network and Cognitive Dysfunction in Alzheimer's Disease Pathogenesis. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
43.  Assis CRD, Linhares AG, Cabrera MP, Oliveira VM, Silva KCC, Marcuschi M, Maciel Carvalho EVM, Bezerra RS, Carvalho LB Jr. Erythrocyte acetylcholinesterase as biomarker of pesticide exposure: new and forgotten insights. Environ Sci Pollut Res Int. 2018;25:18364-18376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
44.  Dabke AT, Pohowalla JN, Inamdar S, Singh SD, Mathur PS. Serum cholinesterase and histopathology of the liver in severe protein calorie malnutrition. Indian J Pediatr. 1972;39:151-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
45.  Camarero González E, Muñoz Leira V, Iglesias Guerrero M, Fernández Alvarez JA, Cabezas-Cerrato J. [Protein-energy malnutrition: its effects on 4 metabolic parameters]. Nutr Hosp. 1995;10:158-160.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Santarpia L, Grandone I, Contaldo F, Pasanisi F. Butyrylcholinesterase as a prognostic marker: a review of the literature. J Cachexia Sarcopenia Muscle. 2013;4:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 168]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
47.  Kocabaş, Ramazan, Erenler, Ali Kemal, Yetim, Mücahit, Doğan, Tolga and Erdemli, Hacı Kemal. Butyrylcholinesterase as an additional marker in the diagnostic network of acute myocardial infarction. Laboratoriums Medizin. 2016;40:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Nechaeva N, Prokopkina T, Makhaeva G, Rudakova E, Boltneva N, Dishovsky C, Eremenko A, Kurochkin I. Quantitative butyrylcholinesterase activity detection by surface-enhanced Raman spectroscopy. Sensors and Actuators B: Chemical. 2018;259:75-82.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Sulzgruber P, Koller L, Reiberger T, El-Hamid F, Forster S, Rothgerber DJ, Goliasch G, Wojta J, Niessner A. Butyrylcholinesterase predicts cardiac mortality in young patients with acute coronary syndrome. PLoS One. 2015;10:e0123948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
50.  Distelmaier K, Winter MP, Rützler K, Heinz G, Lang IM, Maurer G, Koinig H, Steinlechner B, Niessner A, Goliasch G. Serum butyrylcholinesterase predicts survival after extracorporeal membrane oxygenation after cardiovascular surgery. Crit Care. 2014;18:R24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
51.  Vaisi-Raygani A, Tavilani H, Zahrai M, Rahimi Z, Sheikh N, Aminian M, Pourmotabbed T. Serum butyrylcholinesterase activity and phenotype associations with lipid profile in stroke patients. Clin Biochem. 2009;42:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
52.  Zhang QH, Li AM, He SL, Yao XD, Zhu J, Zhang ZW, Sheng ZY, Yao YM. Serum Total Cholinesterase Activity on Admission Is Associated with Disease Severity and Outcome in Patients with Traumatic Brain Injury. PLoS One. 2015;10:e0129082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
53.  Rahimi Z, Ahmadi R, Vaisi-Raygani A, Rahimi Z, Bahrehmand F, Parsian A. Butyrylcholinesterase (BChE) activity is associated with the risk of preeclampsia: influence on lipid and lipoprotein metabolism and oxidative stress. J Matern Fetal Neonatal Med. 2013;26:1590-1594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
54.  Zivkovic AR, Decker SO, Zirnstein AC, Sigl A, Schmidt K, Weigand MA, Hofer S, Brenner T. A Sustained Reduction in Serum Cholinesterase Enzyme Activity Predicts Patient Outcome following Sepsis. Mediators Inflamm. 2018;2018:1942193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
55.  Peng ZL, Huang LW, Yin J, Zhang KN, Xiao K, Qing GZ. Association between early serum cholinesterase activity and 30-day mortality in sepsis-3 patients: A retrospective cohort study. PLoS One. 2018;13:e0203128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
56.  Xu L, Zhu B, Huang Y, Yang Z, Sun J, Xu Y, Zheng J, Kinloch S, Yin MT, Weng H, Wu N. Butyrylcholinesterase Levels on Admission Predict Severity and 12-Month Mortality in Hospitalized AIDS Patients. Mediators Inflamm. 2018;2018:5201652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
57.  Gomes HJ, Souza RL, Prevedello FC, Mira MT, Chautard-Freire-Maia EA. Investigation of Association between Susceptibility to Leprosy and SNPs inside and near the BCHE Gene of Butyrylcholinesterase. J Trop Med. 2012;2012:184819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
58.  Cheng BN, Jin YL, Chen BQ, Zhu LY, Xu ZC, Shen T. Serum cholinesterase: a potential assistant biomarker for hand, foot, and mouth disease caused by enterovirus 71 infection. Infect Dis Poverty. 2016;5:27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
59.  Haghnazari L, Vaisi-Raygani A, Keshvarzi F, Ferdowsi F, Goodarzi M, Rahimi Z, Baniamerian H, Tavilani H, Vaisi-Raygani H, Pourmotabbed T. Effect of Acetylcholinesterase and Butyrylcholinesterase on Intrauterine Insemination, Contribution to Inflammations, Oxidative Stress and Antioxidant Status; A Preliminary Report. J Reprod Infertil. 2016;17:157-162.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Hefter H, Arslan M, Kruschel TS, Novak M, Rosenthal D, Meuth SG, Albrecht P, Hartmann CJ, Samadzadeh S. Pseudocholinesterase as a Biomarker for Untreated Wilson's Disease. Biomolecules. 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
61.  Ben Anes A, Ben Nasr H, Garrouch A, Bennour S, Bchir S, Hachana M, Benzarti M, Tabka Z, Chahed K. Alterations in acetylcholinesterase and butyrylcholinesterase activities in chronic obstructive pulmonary disease: relationships with oxidative and inflammatory markers. Mol Cell Biochem. 2018;445:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
62.  Tvarijonaviciute A, Caldin M, Martinez-Subiela S, Tecles F, Pastor J, Ceron JJ. Serum paraoxonase 1 and butyrylcholinesterase in dogs with hyperadrenocorticism. Vet J. 2015;203:262-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
63.  Kocatürk M, Tecles F, Yalçın E, Cihan H, Tural M, Levent P, Cansev M, Cerón JJ, Yilmaz Z. Changes in choline and cholinesterase in saliva of dogs with parvovirus infection. Res Vet Sci. 2021;134:147-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
64.  Medzhitov R. The spectrum of inflammatory responses. Science. 2021;374:1070-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 185]  [Article Influence: 61.7]  [Reference Citation Analysis (0)]
65.  Sridhar G, Lakshmi G. Influence of butyrylcholinesterase on the course of COVID-19. Biomedical Reviews. 2021;32:37-46.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Bonaz B, Sinniger V, Pellissier S. Anti-inflammatory properties of the vagus nerve: potential therapeutic implications of vagus nerve stimulation. J Physiol. 2016;594:5781-5790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 307]  [Article Influence: 38.4]  [Reference Citation Analysis (0)]
67.  Espeter F, Künne D, Garczarek L, Kuhlmann H, Skarabis A, Zivkovic AR, Brenner T, Schmidt K. Critically Ill COVID-19 Patients Show Reduced Point of Care-Measured Butyrylcholinesterase Activity-A Prospective, Monocentric Observational Study. Diagnostics (Basel). 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Markuskova L, Javorova Rihova Z, Fazekas T, Martinkovicova A, Havrisko M, Dingova D, Solavova M, Rabarova D, Hrabovska A. Serum butyrylcholinesterase as a marker of COVID-19 mortality: Results of the monocentric prospective observational study. Chem Biol Interact. 2023;381:110557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
69.  Borger P. Natural Knockouts: Natural Selection Knocked Out. Biology (Basel). 2017;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
70.  Alkuraya FS. Natural human knockouts and the era of genotype to phenotype. Genome Med. 2015;7:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
71.  Narasimhan VM, Xue Y, Tyler-Smith C. Human Knockout Carriers: Dead, Diseased, Healthy, or Improved? Trends Mol Med. 2016;22:341-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
72.  MacArthur DG, Balasubramanian S, Frankish A, Huang N, Morris J, Walter K, Jostins L, Habegger L, Pickrell JK, Montgomery SB, Albers CA, Zhang ZD, Conrad DF, Lunter G, Zheng H, Ayub Q, DePristo MA, Banks E, Hu M, Handsaker RE, Rosenfeld JA, Fromer M, Jin M, Mu XJ, Khurana E, Ye K, Kay M, Saunders GI, Suner MM, Hunt T, Barnes IH, Amid C, Carvalho-Silva DR, Bignell AH, Snow C, Yngvadottir B, Bumpstead S, Cooper DN, Xue Y, Romero IG; 1000 Genomes Project Consortium, Wang J, Li Y, Gibbs RA, McCarroll SA, Dermitzakis ET, Pritchard JK, Barrett JC, Harrow J, Hurles ME, Gerstein MB, Tyler-Smith C. A systematic survey of loss-of-function variants in human protein-coding genes. Science. 2012;335:823-828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 998]  [Cited by in F6Publishing: 874]  [Article Influence: 72.8]  [Reference Citation Analysis (0)]
73.  Kaiser VB, Svinti V, Prendergast JG, Chau YY, Campbell A, Patarcic I, Barroso I, Joshi PK, Hastie ND, Miljkovic A, Taylor MS; Generation Scotland;  UK10K, Enroth S, Memari Y, Kolb-Kokocinski A, Wright AF, Gyllensten U, Durbin R, Rudan I, Campbell H, Polašek O, Johansson Å, Sauer S, Porteous DJ, Fraser RM, Drake C, Vitart V, Hayward C, Semple CA, Wilson JF. Homozygous loss-of-function variants in European cosmopolitan and isolate populations. Hum Mol Genet. 2015;24:5464-5474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
74.  Narasimhan VM, Hunt KA, Mason D, Baker CL, Karczewski KJ, Barnes MR, Barnett AH, Bates C, Bellary S, Bockett NA, Giorda K, Griffiths CJ, Hemingway H, Jia Z, Kelly MA, Khawaja HA, Lek M, McCarthy S, McEachan R, O'Donnell-Luria A, Paigen K, Parisinos CA, Sheridan E, Southgate L, Tee L, Thomas M, Xue Y, Schnall-Levin M, Petkov PM, Tyler-Smith C, Maher ER, Trembath RC, MacArthur DG, Wright J, Durbin R, van Heel DA. Health and population effects of rare gene knockouts in adult humans with related parents. Science. 2016;352:474-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 202]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
75.  Xu YC, Guo YL. Less Is More, Natural Loss-of-Function Mutation Is a Strategy for Adaptation. Plant Commun. 2020;1:100103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
76.  Kaiser J. The hunt for missing genes. Science. 2014;344:687-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
77.  Manoharan I, Boopathy R, Darvesh S, Lockridge O. A medical health report on individuals with silent butyrylcholinesterase in the Vysya community of India. Clin Chim Acta. 2007;378:128-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 77]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
78.  Li B, Duysen EG, Carlson M, Lockridge O. The butyrylcholinesterase knockout mouse as a model for human butyrylcholinesterase deficiency. J Pharmacol Exp Ther. 2008;324:1146-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
79.  Liu W, Cao Y, Lin Y, Tan KS, Zhao H, Guo H, Tan W. Enhancement of Fear Extinction Memory and Resistance to Age-Related Cognitive Decline in Butyrylcholinesterase Knockout Mice and (R)-Bambuterol Treated Mice. Biology (Basel). 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
80.  Lockridge O, Norgren RB Jr, Johnson RC, Blake TA. Naturally Occurring Genetic Variants of Human Acetylcholinesterase and Butyrylcholinesterase and Their Potential Impact on the Risk of Toxicity from Cholinesterase Inhibitors. Chem Res Toxicol. 2016;29:1381-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
81.  Lando G, Mosca A, Bonora R, Azzario F, Penco S, Marocchi A, Panteghini M, Patrosso MC. Frequency of butyrylcholinesterase gene mutations in individuals with abnormal inhibition numbers: an Italian-population study. Pharmacogenetics. 2003;13:265-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 37]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
82.  Sokolow S, Li X, Chen L, Taylor KD, Rotter JI, Rissman RA, Aisen PS, Apostolova LG. Deleterious Effect of Butyrylcholinesterase K-Variant in Donepezil Treatment of Mild Cognitive Impairment. J Alzheimers Dis. 2017;56:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
83.  Ha ZY, Mathew S, Yeong KY. Butyrylcholinesterase: A Multifaceted Pharmacological Target and Tool. Curr Protein Pept Sci. 2020;21:99-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
84.  Sridhar GR  Proteins of the Esterase Family: Patents for Some Proteins in Search of Metabolic Functions. Recent Patents on Biomarkers, 2011; 1: 205-212. Available from: https://www.ingentaconnect.com/content/ben/rpbm/2011/00000001/00000003/art00004.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Geula C, Darvesh S. Butyrylcholinesterase, cholinergic neurotransmission and the pathology of Alzheimer's disease. Drugs Today (Barc). 2004;40:711-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 55]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
86.  Lockridge O. Review of human butyrylcholinesterase structure, function, genetic variants, history of use in the clinic, and potential therapeutic uses. Pharmacol Ther. 2015;148:34-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 273]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
87.  Lockridge O, Duysen EG, Masson P. Butyrylcholinesterase: overview, structure, and function. Anticholinesterase Pesticides. 2011;10:25-41.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Zhang P, Jain P, Tsao C, Sinclair A, Sun F, Hung HC, Bai T, Wu K, Jiang S. Butyrylcholinesterase nanocapsule as a long circulating bioscavenger with reduced immune response. J Control Release. 2016;230:73-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
89.  MacArthur DG, Tyler-Smith C. Loss-of-function variants in the genomes of healthy humans. Human Molecular Genetics. 2010;19:R125-R130.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Monies D, Abouelhoda M, AlSayed M, Alhassnan Z, Alotaibi M, Kayyali H, Al-Owain M, Shah A, Rahbeeni Z, Al-Muhaizea MA, Alzaidan HI, Cupler E, Bohlega S, Faqeih E, Faden M, Alyounes B, Jaroudi D, Goljan E, Elbardisy H, Akilan A, Albar R, Aldhalaan H, Gulab S, Chedrawi A, Al Saud BK, Kurdi W, Makhseed N, Alqasim T, El Khashab HY, Al-Mousa H, Alhashem A, Kanaan I, Algoufi T, Alsaleem K, Basha TA, Al-Murshedi F, Khan S, Al-Kindy A, Alnemer M, Al-Hajjar S, Alyamani S, Aldhekri H, Al-Mehaidib A, Arnaout R, Dabbagh O, Shagrani M, Broering D, Tulbah M, Alqassmi A, Almugbel M, AlQuaiz M, Alsaman A, Al-Thihli K, Sulaiman RA, Al-Dekhail W, Alsaegh A, Bashiri FA, Qari A, Alhomadi S, Alkuraya H, Alsebayel M, Hamad MH, Szonyi L, Abaalkhail F, Al-Mayouf SM, Almojalli H, Alqadi KS, Elsiesy H, Shuaib TM, Seidahmed MZ, Abosoudah I, Akleh H, AlGhonaium A, Alkharfy TM, Al Mutairi F, Eyaid W, Alshanbary A, Sheikh FR, Alsohaibani FI, Alsonbul A, Al Tala S, Balkhy S, Bassiouni R, Alenizi AS, Hussein MH, Hassan S, Khalil M, Tabarki B, Alshahwan S, Oshi A, Sabr Y, Alsaadoun S, Salih MA, Mohamed S, Sultana H, Tamim A, El-Haj M, Alshahrani S, Bubshait DK, Alfadhel M, Faquih T, El-Kalioby M, Subhani S, Shah Z, Moghrabi N, Meyer BF, Alkuraya FS. The landscape of genetic diseases in Saudi Arabia based on the first 1000 diagnostic panels and exomes. Hum Genet. 2017;136:921-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 196]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
91.  Monies D, Goljan E; Rapid Exome Consortium, Assoum M, Albreacan M, Binhumaid F, Subhani S, Boureggah A, Hashem M, Abdulwahab F, Abuyousef O, Temsah MH, Alsohime F, Kelaher J, Abouelhoda M, Meyer BF, Alkuraya FS. The clinical utility of rapid exome sequencing in a consanguineous population. Genome Med. 2023;15:44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]