Next Article in Journal
Identification of Cellular Factors Required for SARS-CoV-2 Replication
Next Article in Special Issue
Therapeutic Benefit of Galectin-1: Beyond Membrane Repair, a Multifaceted Approach to LGMD2B
Previous Article in Journal
Neuromolecular Underpinnings of Negative Cognitive Bias in Depression
Previous Article in Special Issue
Towards Splicing Therapy for Lysosomal Storage Disorders: Methylxanthines and Luteolin Ameliorate Splicing Defects in Aspartylglucosaminuria and Classic Late Infantile Neuronal Ceroid Lipofuscinosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage

by
Tomáš Zárybnický
1,
Anne Heikkinen
2,3,
Salla M. Kangas
2,4,5,
Marika Karikoski
6,
Guillermo Antonio Martínez-Nieto
6,7,
Miia H. Salo
2,4,5,
Johanna Uusimaa
4,5,8,
Reetta Vuolteenaho
2,
Reetta Hinttala
2,4,5,*,
Petra Sipilä
6,7,* and
Satu Kuure
1,9,*
1
Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland
2
Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
3
Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland
4
PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland
5
Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
6
Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland
7
Turku Center for Disease Modelling (TCDM), Institute of Biomedicine, University of Turku, 20520 Turku, Finland
8
Clinic for Children and Adolescents, Division of Pediatric Neurology, Oulu University Hospital, P.O. Box 20, 90029 Oulu, Finland
9
GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, 00790 Helsinki, Finland
*
Authors to whom correspondence should be addressed.
Cells 2021, 10(11), 3158; https://doi.org/10.3390/cells10113158
Submission received: 27 September 2021 / Revised: 4 November 2021 / Accepted: 6 November 2021 / Published: 13 November 2021
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Rare Diseases)

Abstract

:
The modification of genes in animal models has evidently and comprehensively improved our knowledge on proteins and signaling pathways in human physiology and pathology. In this review, we discuss almost 40 monogenic rare diseases that are enriched in the Finnish population and defined as the Finnish disease heritage (FDH). We will highlight how gene-modified mouse models have greatly facilitated the understanding of the pathological manifestations of these diseases and how some of the diseases still lack proper models. We urge the establishment of subsequent international consortiums to cooperatively plan and carry out future human disease modeling strategies. Detailed information on disease mechanisms brings along broader understanding of the molecular pathways they act along both parallel and transverse to the proteins affected in rare diseases, therefore also aiding understanding of common disease pathologies.

1. Introduction

Understanding and treating human diseases requires thorough knowledge of disease-causing molecular and pathophysiological mechanisms. Despite the recent advances in induced pluripotent stem cell techniques, many of these aspects remain challenging to study, especially under physiological conditions in patient-derived material [1,2]. In order to be reliable and valuable, the disease model should recapitulate if not the entire human disease phenotype, then at least the key features of each specific disease under study [3]. For precision medicine, a good understanding of the genetic bases of variation in phenotypes and their interaction with the environment in health and disease are required [4,5]. Thus, animal models, and especially genetically modified (GM) mice, offer great potential to serve as precious preclinical models that facilitate basic understanding of disease pathomechanisms and provide clues for the development of treatment options and novel strategies to follow a treatment’s response.

Mice as a Genetically Modified Model for Diseases

Mice have been used as an animal model in biomedical studies for decades and for many reasons, including their small size, efficient reproduction, relatively reasonable expenses and similarities in anatomy and physiology to humans [4]. The fact that mice are housed in controlled environment and that its genome was the first that was sequenced among the rodents makes it the quintessential and by far most extensively used animal employed in genetically modified models [6,7]. The possibility to derive and successfully culture mouse embryonic stem (ES) cells together with the importance of similarities between human and mouse genomes significantly contributed to the dominance of the mouse in genetic modeling [8,9].
Almost 15 years ago, an international knockout (KO) mouse consortium (IKMC) with the aim to inactivate all known mouse genes was established [10,11]. It has been extremely successful in generating and providing mouse gene inactivation models either in the form of targeted ES cells or mouse lines as, at least two thirds of the protein coding genes have been knocked out [12,13]. Together with the research community and systematic phenotyping conducted by the International Mouse Phenotyping Consortium (IMPC, https://www.mousephenotype.org, accessed on 15 September 2021), these models have produced a wealth of novel information on gene functions and facilitated the understanding of essential genetic requirements for life [12,14,15]. Detailed phenotyping of KO mice has also identified various full inactivation models that correlate or associate with Mendelian diseases [14,15]. However, straightforward full-gene inactivation appears to be less successful at modeling monogenic diseases than originally thought, and thus more precise models are urged.
One of the big challenges at hand is to model the approximately 6500 different human monogenic diseases in mice [16]. Although typically classified as rare diseases, many monogenic diseases are relatively common, either in general or in certain geographical regions and among certain ethnicities. In this review, we discuss a group of monogenic rare diseases enriched in the Finnish population and defined as the Finnish disease heritage (FDH) [17]. We will highlight how appropriate mouse models have greatly facilitated the understanding of disease manifestation in certain monogenic diseases and, on the other hand, emphasize the barriers in other diseases, as studies have been carried out mainly in cell culture models.

2. The Finnish Disease Heritage

The first scientific reports of FDH were published in early 1970s by Perheentupa et al. [18] and Norio et al. [19] who referred to a group of inherited clinical phenotypes that are, in proportion to population size, more common in Finland than elsewhere in the world. Today, 36 mostly autosomal recessive diseases (Table 1) are defined as FDH, varying in severity from embryonic lethal to adult-onset milder phenotypes [20]. The collection of monogenic FDH is constantly evolving (Table 1), as new diseases following the same patterns of enriched founder mutations are still being identified [21].
The enrichment of some monogenic diseases in Finland (especially in the north and east) was caused by geographical, linguistic and cultural isolation of this population in history [22]. The resultant characteristic features of population isolation (e.g., the founder effect, genetic drift and genetic isolation) have shaped the gene pool of Finns over the centuries, leading to the enrichment of certain disease-causing gene variants [23]. Although FDH diseases are rare, many of them share similarities with more common diseases, and this may sometimes delay a correct diagnosis. Thus, the importance of consultation of national expert centers that are linked to international networks is of ultimate importance for rare disease patients (https://ec.europa.eu/health/ern/, accessed on 1 September 2021). One example of such networks is European Reference Networks on rare diseases, which help professionals and expert centers to share knowledge on rare diseases requiring special care, serve as research and knowledge centers for treating patients from other EU countries, ensure the availability of treatment facilities and provide high-quality training for students and members of multidisciplinary teams.

2.1. State of the Art in Modeling the FDH in Mice

Thanks to the IKMC, many of the FDH genes have already been knocked out in mice (Table 1). However, only a few of the existing KO mouse lines recapitulate the full spectrum of FDH disease symptoms (Table 2). Moreover, as described in Table 2 for all FDH diseases, KO and rare transgenic (TG) and knock-in (KI) mouse models that have been generated for certain diseases selectively manifest only some of the disease features.
The selected FDH diseases and their animal model statuses are discussed in detail in the following section. These examples were chosen to highlight the great variation in disease manifestation and underline the importance of disease-specific knock-in mouse models for advancing knowledge on the molecular networks involved in normal development, physiology and disease pathogenesis. Specific attention was given to diseases with possible advances or suitability in preclinical treatments (AGU and HOGA), with severe developmental complications leading to devastating death of the fetus (HLS1), with severe infancy- or childhood-onset neurodegenerative storage disorders (Northern epilepsy and Salla disease) and with complex symptom spectrums (CHH and LCHAD). As exemplified below, strategies to generate and utilize in vivo models that faithfully genocopy their disease-specific variants and phenocopy major FDH disease symptoms are needed.

2.1.1. Aspartylglucosaminuria (AGU)

AGU (OMIM 208400) represents one of the rare FDH diseases that has been subjected to some treatment strategies and preclinical development (see below). AGU is a recessive neurodegenerative disease which is characterized by progressive intellectual disability, skeletal and connective tissue abnormalities, behavioral changes (e.g., hyperactivity, tantrums and violence) and disruptive sleep patterns followed by premature death, usually before the age of 50 [95,96,97,98]. Developmental delay is the first typical sign of neurological defects, which become evident at 15–18 months of age. Children may manifest macrocephaly and early growth spurts in the infantile phase, but adult affected individuals end up having small brains and lower than average heights [99,100,101,102,103].
AGU is the most common autosomal recessive disease in Finland, and its prevalence is 1.7–5 per 100,000 live births [101,104]. AGU is caused by defects in the lysosomal aspartylglucosaminidase (AGA) enzyme. The AGUFin major variant, which consists of two nucleotide changes (c.482G > A and c.488G > C), covers 98% of cases in Finland, while the AGUFin minor variant (two base pairs deleted) is causative for 1.5% of the cases [105,106,107]. Although being enriched in Finland, AGU affects all ethnicities, and approximately 40 different AGA variants have been identified worldwide (A. Banning and R. Tikkanen, personal communication). Roughly half of the variants are missense mutations, while the rest represent many different aberrations without proper understanding of the genotype–phenotype causalities [98]. Notably, recent findings have demonstrated several patients with high residual AGA activity and a milder phenotype [108]. This is an important finding for development of treatment options, since it means that even less than 50% AGA activity (i.e., the carrier level) may result in a significant improvement of the phenotype.
The pathogenic AGA variants result in decreased AGA activity [109,110,111,112]. This causes a failure to break down the N-glycosidic bond of glycoproteins and leads to the progressive accumulation of AGA substrates, including glycoasparagine in lysosomes [113]. The KO mice of Aga recapitulated the biochemical defects of AGU, as glycoasparagine accumulation was evident in the studied tissues, but the model failed to manifest the early onset and progressive nature of specific disease symptoms [24,25].
Although there are no approved curative therapies currently available for AGU, some disease-modifying strategies have been and are currently being tested. Enzyme replacement therapy was shown to work in human cells and preclinical KO mouse models but has not been transferred to clinical trials, due to challenges in the required large-scale protein production and concerns for the blood–brain barrier transport [29,30,114,115]. Adenoviral gene therapy in Aga KO mice restored the enzyme activity [116,117]. The latest experiments with serotype-specific adeno-associated viral vectors safely targeting the central nervous system (AAV9) alleviated the neurological phenotype of Aga KO mice [26] and suggested clinical translatability due to good adherence with the dosage and blood–brain barrier transport requirements. Hematopoietic stem cell transplantation as a potential treatment of AGU showed promising results in the Aga KO mice but failed to improve the neuropsychologic and other clinical symptoms of AGU in clinical trials [114,115,118,119]. While promising, it is possible that previous enzyme replacement and gene and stem cell transplantation preclinical trials provoked unmerited hope due to the use of an inappropriate KO mouse model, which does not recapitulate the disease pathomechanisms caused by misfolded AGA protein.
Currently one AGU treatment option being tested is pharmacological chaperone therapy, which aims at facilitating proper folding of the mutant AGA protein. It can restore AGA activity in patient-derived fibroblasts and is currently in phase I or II clinical trials for efficacy and safety testing [108] (clinical trial). Of note, this clinical trial is conducted without preclinical animal testing because the Aga KO mouse is not a suitable model for this type of therapeutic strategy, and animal testing is not required for drug repurposing with approved agents such as anhydrous betaine (Cystadane), the chaperone used for AGU treatment, which is already approved for the treatment of homocystinuria. Important for future studies of targeted therapies in AGU is that a mouse model mimicking human disease-causing variant(s) is still lacking but would be highly desirable to facilitate drug development.

2.1.2. Cartilage Hair Hypoplasia (CHH)

CHH (OMIM 250250), originally described by McKusick et al. in 1965, is metaphyseal chondrodysplasia characterized by a short stature, sparse hypoplastic hair and immune deficiency. Patients with CHH may also suffer from gastrointestinal dysfunctions, anemia and impaired spermatogenesis [120,121,122]. Especially due to malignancies and diseases of the respiratory system, patients with CHH have an increased risk of early mortality [123]. CHH is enriched among the Old Order Amish and Finnish populations, and the incidence in Finland is 1 in 23,000 births [124]. CHH is caused by defects in the long non-coding RNA gene known as RMRP [125]. The most common pathogenic variant is n.71A > G (previously known as n.70A > G) substitution, representing 92% of the disease-causing variants among Finnish CHH patients and also being the most frequent, if not the only one, in the Amish population [126,127].
RMRP encodes the RNA component of mitochondrial RNA processing endoribonuclease. It has well-characterized roles in many cancers, possibly due to its recently recognized function to inhibit p53 [128,129]. The RMRP KO mouse model indicates that it is essential for early embryonic development, as homozygous null mice die in utero [31]. Due to the missing mammalian CHH model, the pathophysiological mechanisms leading to the disease symptom spectrum remains poorly understood.

2.1.3. Hydrolethalus Syndrome 1 (HLS1)

HLS1 (OMIM 236680) is a lethal disease with multiple developmental anomalies at the fetal stage, and it leads to stillbirth or death of the affected child soon after birth. The characteristic findings of HLS1 in the central nervous system are hydrocephalus, missing midline structures of the brain and a keyhole-shaped foramen magnum [130,131,132]. The incidence of this syndrome is 1 in 20,000 births in Finland [133,134,135]. Patients with similar or somewhat milder clinical features have also been reported outside Finland [133,136,137,138]. However, no causative variants have been confirmed in these cases.
An autosomal recessive variant of the HYLS1 gene carrying the c.1416A > G transition has been identified as the disease-causing variant in HLS1. This A-to-G nucleotide transition in exon 6 leads to substitution of the conserved aspartate-211 to glycine (p.D211G) in the HYLS protein [139]. Hydrolethalus syndrome 2 (HLS2, OMIM 614120) has similar features to HLS1, but the causative variant resides in the KIF7 gene [134]. Both HLS1 and HLS2 belong to primary ciliopathies, a diverse spectrum of neurodevelopmental disorders [135].
HYLS1 encodes centriolar- and ciliogenesis-associated protein, which has been shown to play an important role in ciliogenesis in Caenorhabditis elegans and Drosophila melanogaster [140,141,142]. In vitro studies on human-derived cells and the examination of tissues from aborted HLS1 fetuses suggest that HYLS1 is a transcriptional regulator essential for fetal development [143]. However, the exact cellular and molecular mechanisms behind severe malformations of the brain and other organs in HLS1 are yet unknown.
Currently, there are no publications on mouse models lacking HYLS1 or replicating the disease-causing variant of HLS1. The severity of the syndrome and the utmost significance of HYLS1 protein to fetal development highlight the importance of further studies to advance our knowledge on molecular networks involved in embryo development both in health and disease.

2.1.4. Hyperornithinemia with Gyrate Atrophy of the Choroid and Retina (HOGA)

HOGA (also known as GACR, OMIM 258870) is a rare autosomal recessive disorder characterized by progressive chorioretinal degeneration showing clinical symptoms during the first and second decades of life. It leads to visual impairment and blindness in adolescence or adulthood and early cataract formation, and it also often leads to neurological abnormalities and type II muscle fiber atrophy [144,145].
HOGA has an estimated global incidence of 1 in 1,500,000, being the highest in the Finnish population with an incidence of 1 in 50,000 [145,146]. This rare genetic disease is caused by defects in the ornithine-degrading mitochondrial enzyme, ornithine delta-aminotransferase (OAT, OMIM 613349), which leads to hyperornithinemia in the plasma [144]. To date, more than 60 variants have been identified in the OAT gene that cause HOGA, 90% of which account for missense or frameshift changes [145]. From all the different variants, the C-terminal domain leucine-402 to the proline variant (L402P) and the catalytic site variant arginine-180 to the threonine (R180T) are the most frequent ones among the Finnish population [145].
The OAT enzyme function is bidirectional; in the early neonatal period it is more active in ornithine production, but later, this is reversed. Therefore, the adolescence-onset clinical HOGA manifestation can be delayed by a rigorous arginine-restricted diet, since this amino acid is the main source of ornithine. Several of the human variants have only been studied in patient-derived cells, human cell lines or in yeast [147,148,149]. In addition, there are two mouse models available for studying the function of the Oat and HOGA disease: a full KO line (Oattm1Dva) and a mouse line with a spontaneous recessive mutation, called retarded hair growth (Oatrhg), that harbors glycine-353 to alanine (G353A) substitution in OAT protein [150]. Studies with KO mice showed that the loss of Oat leads to neonatal lethality due to hypoornithinemia and subsequent arginine synthesis failure in the small intestine [66]. Notably, neonatal lethality has not been observed in HOGA patients, although one asymptomatic individual was reported to have transient neonatal hypoornithinemia. This suggests that in human infants, the biochemical phenotype might be similar, albeit milder than in KO mice. The neonatal lethality observed in KO mice can be rescued by arginine supplementation, and adult mice develop hyperornithinemia and retinal degeneration comparable to humans. The Oatrhg mice model the classical gyrate atrophy disease with chorioretinal deterioration and hyperornithinemia [151].
Despite the existence of two mouse models, the HOGA pathogenesis and the exact molecular mechanism of the different OAT variants remain elusive, which reflects the current lack of treatments. This highlights the necessity for generating animal models that recapitulate the human variants to facilitate understanding of the HOGA disease and the development of efficient therapies.

2.1.5. Northern Epilepsy (EPMR)

Northern epilepsy (progressive epilepsy with mental retardation (EPMR), OMIM 610003) is a neurodegenerative storage disease and a form of neuronal ceroid lipofuscinosis (NCL) [152]. The disease onset is at 5–10 years of age and marked by the appearance of generalized tonic-clonic seizures. The frequency of epileptic seizures increases toward puberty, after which the epileptic activity decreases. Mental deterioration is typically observed 2–5 years after the onset of epilepsy, and it is progressive despite the decline in the frequency of epileptic seizures toward adulthood. Other features associated with Northern epilepsy are pubertal behavioral difficulties as well as problems in fine motor tasks and equilibrium [153]. Progressive brain atrophy [147] and the accumulation of lipopigment in the cytoplasm of neurons and other cell types are observed at the tissue level [152].
Northern epilepsy is caused by a missense variant (c.70C > G) in the CLN8 gene encoding the CLN8 transmembrane ER and ERGIC protein [154], which is involved in the transport of newly synthesized lysosomal enzymes from the endoplasmic reticulum (ER) to the Golgi apparatus [148]. This pathogenic variant results in arginine to glycine substitution at amino acid 24 (R24G) of CLN8 [154]. While the variant causing Northern epilepsy has only been found in the Finnish population, other pathogenic variants in CLN8 have been found elsewhere with different NCL disease phenotypes (OMIM 600143). In addition to its role in the ER for Golgi trafficking of lysosomal enzymes, CLN8 has recently been suggested to function in the regulation of endo-lysosomal dynamics and dendritic morphology [149].
Motor neuron degeneration (mnd) in mice, caused by spontaneous mutation in Cln8, is the most characterized murine model for CLN8 deficiency. This naturally occurring variant (c.267–268insC at codon 90 of Cln8) results in frameshift mutation and a premature stop codon [154]. While the Cln8mnd/mnd mouse recapitulates many NCL features [57,155], the genotype, intracellular targeting and some of the phenotypic features do not correspond to those observed specifically in Northern epilepsy [156,157,158]. Recently, AAV9 gene therapy was tested in Cln8mnd mice, with promising NCL-alleviating results [58]. To date, no specific KI models of Northern epilepsy have been published.

2.1.6. Salla Disease (SD)

Salla disease (OMIM 604369) is a slowly progressing neurodegenerative lysosomal sialic acid storage disorder. Its first clinical signs, such as muscular hypotonia, ataxia, transient nystagmus and retarded motor development, are usually observed during the first year of life. The life expectancy of the patients is considered to have slightly decreased. The severity of the clinical manifestations may vary, but all patients are intellectually disabled [159], and characteristic MRI findings include delayed myelination in the brain [160]. Salla disease manifests with the accumulation of sialic acid in lysosomes due to deficient sialic acid transportation out of the lysosomal membrane, and one of the key clinical characteristics of the patients includes excessive amounts of secreted sialic acid in their urine [159].
Salla disease and other related and often more severe sialic acid storage diseases (SASD) are caused by pathogenic variants in the SLC17A5 protein, which functions as a sialic acid transporter in lysosomes [161]. Most of the Finnish Salla disease patients are homozygous for missense variant R39C [162]. SLC17A5 also has nitrate transporter activity [156], and the extralysosomal localization in the CNS indicates that it may have other functional roles in addition to lysosomal sialic acid transport [163].
The SLC17A5 gene is highly conserved across species. The mouse Slc17A5 sequence is an 86.26% match with its human ortholog (Ensembl release 104, May 2021 [164]). The Slc17A5 KO mouse model has a severe hypomyelination phenotype leading to death after 3 weeks of life [89,90]. Even though the KO phenotype in mice resembles a human disease in the terms of hypomyelination and lysosomal accumulation, the short life span and total loss of the protein do not facilitate studies of the disease mechanisms that could be causative for human patients. Different SLC17A5 variants cause different disease phenotypes, varying from mild to severe due to the amount of residual functional activity [162]. Interestingly, SLC17A5 variants have also been recognized as candidates for Parkinson’s disease susceptibility genes [165]. Therefore, specific KI mouse models that recapitulate the phenotype of Salla disease are needed to better understand the versatile functions of the SLC17A5 protein and pathogenetic processes behind the phenotypic spectrum.

2.1.7. Evolving FDH: An Example of Long-Chain 3-hydroxyacyl-CoA Dehydrogenase Deficiency (LCHAD)

Although the definition for FDH has remained the same for decades, new diseases have been identified and are constantly included in this definition. One of the diseases currently under consideration for FDH is long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHAD, OMIM 609016), which is a mitochondrial disorder of long-chain fatty acid oxidation characterized by infancy- or early childhood-onset metabolic acidosis, hypoketotic hypoglycemia, hypotonia, liver dysfunction, cardiomyopathy and arrhythmias [157,158]. Other symptoms of LCHAD deficiency include later-onset chronic peripheral neuropathy and pigmentary retinopathy. Furthermore, LCHAD deficiency carriers have an increased risk of pregnancy complications, including acute fatty liver; hemolysis, elevated liver enzymes and low platelets (HELLP) syndrome; and pre-eclampsia [166,167,168]. Treatment of LCHAD deficiency comprises a low-fat and high-carbohydrate diet, as well as avoidance of fasting. To obviate the fasting period during the nighttime in infants and small children, a nasogastric tube or gastrostomy is beneficial. During metabolic stress, like infections such as gastroenteritis, an infusion of intravenous glucose is used [169].
LCHAD deficiency in Finland is typically caused by the c.1528G > C founder mutation in the hydroxyacyl-coenzyme A dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) gene encoding the α-subunit of the mitochondrial trifunctional protein (MPT) complex. The missense variant leads to the substitution of glutamate-510 to glutamine (E510Q) [170]. This results in the accumulation of long-chain 3-hydroxy fatty acids and long-chain 3-hydroxyacylcarnitines in the patients’ tissues, but the damaging mechanisms causing the spectrum of symptoms manifesting at very different stages of life are not fully understood [171,172]. Early studies suggested that defects in mitochondrial energy metabolism might underlay the skeletal muscle defects, and this has been supported by in vitro studies with mitochondria isolated from rat skeletal muscle [173,174,175]. Homozygote Hadha KO mice with exon 15 deletion are reported to result in either early postnatal or embryonic lethality, while heterozygosity leads to hepatic steatosis at a young age (3 months) and hepatocellular carcinoma without cirrhosis at an older age (>13 months) [70,176].
Although dietary restrictions such as prevention of fasting and supplementation of carbohydrates and medium-chain triacylglycerols, together with acute infection avoidance, form the basis of LCHAD deficiency management, they insufficiently protect patients from long-term adverse effects. Better in vivo understanding of the consequences of metabolite accumulation in different tissues is needed to improve the life quality and expectancy of LCHAD deficiency patients.

3. Conclusions

For practical and ethical reasons, model organisms have been used as simplified models of humans to study the genetic, molecular and physiological basis of complex traits and to find therapeutic targets for human diseases [4]. At the same time, the use of animal experiments is ethically controversial and needs to be thoroughly justified. Non-animal approaches, based mainly on cell or tissue culture and in silico computational methods, may help to reduce the number of animals used for experimentation and predict clinical outcomes in a limited manner. Many rare hereditary diseases have a multi-organic clinical presentation. Cell and organ culture or computational models are incapable of modeling such biological complexity. Thus, deciphering the physiological functions, pathological processes and interactions between tissues necessitates the use of the whole organism. Consequently, mouse models continue to have a crucial role in biomedical research as well as drug discovery and development [177].
Although traditional KO mice have been valuable tools for studying gene functions, in many cases, the use of full inactivation of gene function has not successfully modeled the human monogenic diseases caused by point mutations in the corresponding gene (Table 2). The accurate recapitulation of disease-causing variants in mice could be the key to providing valuable tools for studying rare diseases. In recent years, the clustered and regularly interspaced palindromic sequences and CAS9 endonuclease (CRISPR/Cas9) genome editing technique have significantly improved the efficiency of generating animal models and been proven especially useful for the generation of point mutation models. In addition, CRISPR/Cas9 technology may alleviate some of the ethical concerns of animal use, especially during model creation. The three Rs principle—replacement, reduction and refinement [178]—could be tackled in several ways when using the CRISPR/Cas9 method, which increases the precision in genome editing and may help to refine and reduce mice, especially when generating complex models or engineering a mutation to the existing mutant background [179].
Importantly, in addition to providing valuable tools for diagnostic, prognostic and therapeutic strategies for rare diseases, precision mouse models facilitate our understanding of the pathomechanisms in the more common diseases affecting the same signaling pathways or cell and tissue types. A good example of this is a rare inherited Tangier disease, named after the location in which it was initially discovered and characterized by significantly reduced levels of high-density lipoproteins (HDLs) in the blood. Tangier disease is caused by a mutation in the ABCA1 gene encoding the ATP-binding cassette transporter A1 (ABCA1), leading to impaired cholesterol efflux capacity. Research on Tangier disease and ABCA1 has had a tremendous impact on the understanding of HDL cholesterol metabolism and atherosclerosis [180,181]. Furthermore, the involvement of ABCA1 has been demonstrated in the pathophysiology of Alzheimer’s disease, for which it is now studied as a novel therapeutic target [182].
Owing to pioneer work by clinicians and geneticists working with FDH patients, the clinical implications and the genetic etiologies of the FDH are well known. However, the consequences of pathogenic variants and their contribution to disease progression at the molecular, cellular and tissue levels remain to be resolved for many diseases. CRISPR/Cas9 technology now allows the generation of disease models for the FDH with exact correspondence of disease-causing human variants in the mouse genome. From a total of 36 FDH diseases, 8 completely lack a mouse model, and in 14 diseases with the existing mouse model(s), only part of the disease symptoms is recapitulated (Table 1 and Table 2). In 2020, the corresponding authors of this review established a national FinnDisMice research consortium with the aim of generating mouse models that recapitulate the human disease-causing variants of the equivalent disease. The overall goal of this effort is to facilitate understanding of the disease pathomechanisms, provide preclinical tools for the development of novel treatment strategies and increase understanding of the molecular mechanisms behind similar common diseases, such as amyotrophic lateral sclerosis (ALS), Parkinson’s disease and other degenerative disorders.
Future technical developments in genome engineering will likely simplify the generation of humanized mouse models, with whole mouse genes substituted with human orthologs with or without disease-causing mutations [183]. Humanized models would be especially useful in the cases where there is low homology between the mouse and human orthologs, as well as in therapeutic development. Humanizing whole genes can be performed either by replacing the mouse gene with the human protein coding region and intervening introns or by excluding some or all the introns to reduce the size of the genomic fragment to be inserted. However, the latter option can result in unexpected surprises, such as in splicing [183]. Thus, when humanizing entire genes, the size of the genomic fragment inserted tends to be dozens or even hundreds of kilobases. The traditional targeting via homologous recombination in mouse ES cells is very inefficient, especially for inserting such long sequences. CRISPR/Cas9 in combination with the vectors accepting large genomic inserts (bacterial artificial chromosomes (BACs)) or single-stranded oligodeoxynucleotides has been proven to improve the targeted insertion of human TERT and SIRPA genes [184,185]. With the use of CRISPR/Cas9, the generation of humanized mice has become easier; however, it is still far from routine work. The current and future developments in the technique are likely to provide easier and more efficient ways of humanizing mice. While waiting for those technical advances, point mutation models of monogenic diseases provide a great preclinical tool for a wide research community.
Interestingly, a recent work with so-called “wildling” mice revealed that C57Bl/6 laboratory mice, which represent a much-used wild type strain where genetic engineering is carried out, with natural microbiota and pathogens phenocopy human immune responses better than the normally used pathogen-free C57Bl/6 mice [186]. Wildling mice were generated by transferring C57Bl/6NTac embryos into the oviducts of wild female mice. The resulting wildling mice had bacterial microbiomes in the gut, skin and vagina similar to those of wild mice, and the microbiota remained stable during several further generations (tested until F5 generation). Using a similar strategy in genetic manipulations could provide better translational value for precision medicine mouse models, especially in the cases where disease pathomechanisms involve the immune system (CHH) or neurodegeneration (Northern epilepsy and Salla disease).
Animal studies remain indispensable for understanding the complex biology and physiology of living organisms, and many regulatory authorities require them as a safety checkpoint in testing new treatments, whether based on drugs, genetic solutions or regenerative processes. Additionally, animal-based research often plays a crucial role in providing new insight into the understanding of diseases, associated pathologies and the identification of targets to which the treatment is directed [6]. Thus, animal experiments remain the best—but yet imperfect—model to predict and characterize human diseases [187].

Author Contributions

Conceptualization, T.Z., R.H., P.S. and S.K.; writing—original draft preparation, T.Z., A.H., S.M.K., M.K., G.A.M.-N., M.H.S., J.U., R.V., R.H., P.S. and S.K.; writing—review and editing, R.H., P.S. and S.K.; supervision, R.H., P.S. and S.K.; project administration, R.H., P.S. and S.K.; funding acquisition, R.H., P.S. and S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Jane and Aatos Erkko Foundation, FinnDisMice consortium; Helsinki Institute of Life Science (HiLIFE), University of Helsinki for S.K.; Foundation for Pediatric Research; the Academy of Finland project grant (#331436) for J.U., and profiling program (grant #311934), University of Oulu for R.H. Open access funding provided by University of Helsinki.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Jukka Kallijärvi (Folkhälsän Research Center, Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland), Ritva Tikkanen (Institute of Biochemistry, Medical Faculty, University of Giessen, Germany) and Kirmo Wartiovaara (Clinical Genetics, Helsinki University Hospital, Finland) for their valuable comments on LCHAD, AGU and HOGA, respectively.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Little, M.H.; Combes, A.N. Kidney organoids: Accurate models or fortunate accidents. Genes Dev. 2019, 33, 1319–1345. [Google Scholar] [CrossRef] [Green Version]
  2. Schutgens, F.; Clevers, H. Human Organoids: Tools for Understanding Biology and Treating Diseases. Annu. Rev. Pathol. Mech. Dis. 2020, 15, 211–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Milne, T.A. Mouse models of MLL leukemia: Recapitulating the human disease. Blood 2017, 129, 2217–2223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Li, H.; Auwerx, J. Mouse Systems Genetics as a Prelude to Precision Medicine. Trends Genet. 2020, 36, 259–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Tadenev, A.L.D.; Burgess, R.W. Model validity for preclinical studies in precision medicine: Precisely how precise do we need to be? Mamm. Genome 2019, 30, 111–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Whitelaw, C.B.A.; Sheets, T.P.; Lillico, S.; Telugu, B.P. Engineering large animal models of human disease. J. Pathol. 2015, 238, 247–256. [Google Scholar] [CrossRef]
  7. Gurumurthy, C.B.; Lloyd, K.C.K. Generating mouse models for biomedical research: Technological advances. Dis. Model. Mech. 2019, 12, dmm029462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Pettitt, S.J.; Liang, Q.; Rairdan, X.Y.; Moran, J.L.; Prosser, H.M.; Beier, D.R.; Lloyd, K.C.; Bradley, A.; Skarnes, W.C. Agouti C57BL/6N embryonic stem cells for mouse genetic resources. Nat. Methods 2009, 6, 493–495. [Google Scholar] [CrossRef] [Green Version]
  9. Waterston, R.H.; Lindblad-Toh, K.; Birney, E.; Rogers, J.; Abril, J.F.; Agarwal, P.; Agarwala, R.; Ainscough, R.; Alexandersson, M.; An, P.; et al. Initial sequencing and comparative analysis of the mouse genome. Nature 2002, 420, 520–562. [Google Scholar] [CrossRef]
  10. International Mouse Knockout Consortium; Collins, F.S.; Rossant, J.; Wurst, W. A Mouse for All Reasons. Cell 2007, 128, 9–13. [Google Scholar] [CrossRef] [Green Version]
  11. Bradley, A.; Anastassiadis, K.; Ayadi, A.; Battey, J.F.; Bell, C.; Birling, M.-C.; Bottomley, J.; Brown, S.D.; Bürger, A.; Bult, C.J.; et al. The mammalian gene function resource: The international knockout mouse consortium. Mamm. Genome 2012, 23, 580–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Skarnes, W.C.; Rosen, B.; West, A.P.; Koutsourakis, M.; Bushell, W.; Iyer, V.; Mujica, A.O.; Thomas, M.; Harrow, J.; Cox, T.; et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 2011, 474, 337–342. [Google Scholar] [CrossRef] [Green Version]
  13. Birling, M.C.; Yoshiki, A.; Adams, D.J.; Ayabe, S.; Beaudet, A.L.; Bottomley, J.; Bradley, A.; Brown, S.D.M.; Bürger, A.; Bushell, W.; et al. A resource of targeted mutant mouse lines for 5,061 genes. Nat. Genet. 2021, 53, 416–419. [Google Scholar] [CrossRef]
  14. Meehan, T.F.; Conte, N.; West, D.B.; Jacobsen, J.O.; Mason, J.; Warren, J.; Chen, C.K.; Tudose, I.; Relac, M.; Matthews, P.; et al. Disease model discovery from 3,328 gene knockouts by The International Mouse Phenotyping Consortium. Nat. Genet. 2017, 49, 1231–1238. [Google Scholar] [CrossRef] [Green Version]
  15. Cacheiro, P.; Haendel, M.A.; Smedley, D.; Meehan, T.; Mason, J.; Mashhadi, H.H.; Muñoz-Fuentes, V.; Tocchini, G.; Lloyd, K.K.C.; McKerlie, C.; et al. New models for human disease from the International Mouse Phenotyping Consortium. Mamm. Genome 2019, 30, 143–150. [Google Scholar] [CrossRef] [Green Version]
  16. Lee, C.E.; Singleton, K.S.; Wallin, M.; Faundez, V. Rare Genetic Diseases: Nature’s Experiments on Human Development. iScience 2020, 23, 101123. [Google Scholar] [CrossRef]
  17. Norio, R. Finnish Disease Heritage I. Hum. Genet. 2003, 112, 441–456. [Google Scholar] [CrossRef] [PubMed]
  18. Perheentupa, J. Hereditary diseases in Finland—From the clinician’s and scientist’s point of view. Duodecim 1972, 88, 1–3. [Google Scholar]
  19. Norio, R.; Nevanlinna, H.R.; Perheentupa, J. Hereditary diseases in Finland; rare flora in rare soul. Ann. Clin. Res. 1973, 5, 109–141. [Google Scholar] [PubMed]
  20. Norio, R. The Finnish disease heritage III: The individual diseases. Hum. Genet. 2003, 112, 470–526. [Google Scholar] [CrossRef]
  21. Trotta, L.; Hautala, T.; Hämäläinen, S.; Syrjänen, J.; Viskari, H.; Almusa, H.; Lepistö, M.H.; Kaustio, M.; Porkka, K.; Palotie, A.; et al. Enrichment of rare variants in population isolates: Single AICDA mutation responsible for hyper-IgM syndrome type 2 in Finland. Eur. J. Hum. Genet. 2016, 24, 1473–1478. [Google Scholar] [CrossRef]
  22. Griffey, M.; Bible, E.; Vogler, C.; Levy, B.; Gupta, P.; Cooper, J.; Sands, M.S. Adeno-associated virus 2-mediated gene therapy decreases autofluorescent storage material and increases brain mass in a murine model of infantile neuronal ceroid lipofuscinosis. Neurobiol. Dis. 2004, 16, 360–369. [Google Scholar] [CrossRef]
  23. Peltonen, L.; Jalanko, A.; Varilo, T. Molecular genetics of the Finnish disease heritage. Hum. Mol. Genet. 1999, 8, 1913–1923. [Google Scholar] [CrossRef] [Green Version]
  24. Kaartinen, V.; Mononen, I.; Voncken, J.W.; Noronkoski, T.; Gonzalez-Gomez, I.; Heisterkamp, N.; Groffen, J. A mouse model for the human lysosomal disease aspartylglycosaminuria. Nat. Med. 1996, 2, 1375–1378. [Google Scholar] [CrossRef] [PubMed]
  25. Jalanko, A.; Tenhunen, K.; McKinney, C.E.; Lamarca, M.E.; Rapola, J.; Autti, T.; Joensuu, R.; Manninen, T.; Sipilä, I.; Ikonen, S.; et al. Mice with an aspartylglucosaminuria mutation similar to humans replicate the pathophysiology in patients. Hum. Mol. Genet. 1998, 7, 265–272. [Google Scholar] [CrossRef] [Green Version]
  26. Chen, X.; Snanoudj-Verber, S.; Pollard, L.; Hu, Y.; Cathey, S.S.; Tikkanen, R.; Gray, S.J. Pre-clinical Gene Therapy with AAV9/AGA in Aspartylglucosaminuria Mice Provides Evidence for Clinical Translation. Mol. Ther. 2021, 29, 989–1000. [Google Scholar] [CrossRef]
  27. Dunder, U.; Kaartinen, V.; Valtonen, P.; Väänänen, E.; Kosma, V.-M.; Heisterkamp, N.; Groffen, J.; Mononen, I. Enzyme replacement therapy in a mouse model of aspartylglycosaminuria. FASEB J. 2000, 14, 361–367. [Google Scholar] [CrossRef]
  28. Dunder, U.; Mononen, I. Human leukocyte glycosylasparaginase: Cell-to-cell transfer and properties in correction of aspartylglycosaminuria. FEBS Lett. 2001, 499, 77–81. [Google Scholar] [CrossRef] [Green Version]
  29. Ramsey, C.; Winqvist, O.; Puhakka, L.; Halonen, M.; Moro, A.; Kämpe, O.; Eskelin, P.; Pelto-Huikko, M.; Peltonen, L. Aire deficient mice develop multiple features of APECED phenotype and show altered immune response. Hum. Mol. Genet. 2002, 11, 397–409. [Google Scholar] [CrossRef] [PubMed]
  30. Anderson, M.S.; Venanzi, E.S.; Klein, L.; Chen, Z.; Berzins, S.P.; Turley, S.J.; von Boehmer, H.; Bronson, R.; Dierich, A.; Benoist, C.; et al. Projection of an Immunological Self Shadow Within the Thymus by the Aire Protein. Science 2002, 298, 1395–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Rosenbluh, J.; Nijhawan, D.; Chen, Z.; Wong, K.-K.; Masutomi, K.; Hahn, W.C. RMRP Is a Non-Coding RNA Essential for Early Murine Development. PLoS ONE 2011, 6, e26270. [Google Scholar] [CrossRef] [Green Version]
  32. Tolmachova, T.; Anders, R.; Abrink, M.; Bugeon, L.; Dallman, M.; Futter, C.; Ramalho, J.; Tonagel, F.; Tanimoto, N.; Seeliger, M.W.; et al. Independent degeneration of photoreceptors and retinal pigment epithelium in conditional knockout mouse models of choroideremia. J. Clin. Investig. 2006, 116, 386–394. [Google Scholar] [CrossRef] [Green Version]
  33. Wavre-Shapton, S.T.; Tolmachova, T.; Lopes da Silva, M.; Futter, C.E.; Seabra, M.C. Conditional Ablation of the Choroideremia Gene Causes Age-Related Changes in Mouse Retinal Pigment Epithelium. PLoS ONE 2013, 8, e57769. [Google Scholar] [CrossRef]
  34. Gupta, P.; Soyombo, A.A.; Atashband, A.; Wisniewski, K.E.; Shelton, J.M.; Richardson, J.A.; Hammer, R.E.; Hofmann, S.L. Disruption of PPT1 or PPT2 causes neuronal ceroid lipofuscinosis in knockout mice. Proc. Natl. Acad. Sci. USA 2001, 98, 13566–13571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bible, E.; Gupta, P.; Hofmann, S.L.; Cooper, J.D. Regional and cellular neuropathology in the palmitoyl protein thioesterase-1 null mutant mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol. Dis. 2004, 16, 346–359. [Google Scholar] [CrossRef]
  36. Griffey, M.; Macauley, S.L.; Ogilvie, J.M.; Sands, M.S. AAV2-mediated ocular gene therapy for infantile neuronal ceroid lipofuscinosis. Mol. Ther. 2005, 12, 413–421. [Google Scholar] [CrossRef]
  37. Kielar, C.; Maddox, L.; Bible, E.; Pontikis, C.C.; Macauley, S.; Griffey, M.A.; Wong, M.; Sands, M.; Cooper, J.D. Successive neuron loss in the thalamus and cortex in a mouse model of infantile neuronal ceroid lipofuscinosis. Neurobiol. Dis. 2007, 25, 150–162. [Google Scholar] [CrossRef] [Green Version]
  38. Macauley, S.L.; Wozniak, D.F.; Kielar, C.; Tan, Y.; Cooper, J.D.; Sands, M.S. Cerebellar pathology and motor deficits in the palmitoyl protein thioesterase 1-deficient mouse. Exp. Neurol. 2009, 217, 124–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Jalanko, A.; Vesa, J.; Manninen, T.; von Schantz, C.; Minye, H.; Fabritius, A.-L.; Salonen, T.; Rapola, J.; Gentile, M.; Kopra, O.; et al. Mice with Ppt1Δex4 mutation replicate the INCL phenotype and show an inflammation-associated loss of interneurons. Neurobiol. Dis. 2005, 18, 226–241. [Google Scholar] [CrossRef]
  40. Staropoli, J.F.; Haliw, L.; Biswas, S.; Garrett, L.; Hölter, S.M.; Becker, L.; Skosyrski, S.; Da Silva-Buttkus, P.; Calzada-Wack, J.; Neff, F.; et al. Large-Scale Phenotyping of an Accurate Genetic Mouse Model of JNCL Identifies Novel Early Pathology Outside the Central Nervous System. PLoS ONE 2012, 7, e38310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Mitchison, H.M.; Bernard, D.J.; Greene, N.D.E.; Cooper, J.D.; Junaid, M.A.; Pullarkat, R.K.; de Vos, N.; Breuning, M.H.; Owens, J.W.; Mobley, W.C.; et al. Targeted Disruption of the Cln3 Gene Provides a Mouse Model for Batten Disease. Neurobiol. Dis. 1999, 6, 321–334. [Google Scholar] [CrossRef] [Green Version]
  42. Cotman, S.L.; Vrbanac, V.; Lebel, L.-A.; Lee, R.L.; Johnson, K.A.; Donahue, L.-R.; Teed, A.M.; Antonellis, K.; Bronson, R.T.; Lerner, T.J.; et al. Cln3 Deltaex7/8 knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum. Mol. Genet. 2002, 11, 2709–2721. [Google Scholar] [CrossRef]
  43. Kovács, A.D.; Pearce, D.A. Finding the most appropriate mouse model of juvenile CLN3 (Batten) disease for therapeutic studies: The importance of genetic background and gender. Dis. Model. Mech. 2015, 8, 351–361. [Google Scholar] [CrossRef] [Green Version]
  44. Kopra, O.; Vesa, J.; Von Schantz, C.; Manninen, T.; Minye, H.; Fabritius, A.-L.; Rapola, J.; Van Diggelen, O.P.; Saarela, J.; Jalanko, A.; et al. A mouse model for Finnish variant late infantile neuronal ceroid lipofuscinosis, CLN5, reveals neuropathology associated with early aging. Hum. Mol. Genet. 2004, 13, 2893–2906. [Google Scholar] [CrossRef] [Green Version]
  45. Schmiedt, M.-L.; Blom, T.; Blom, T.; Kopra, O.; Wong, A.; von Schantz-Fant, C.; Ikonen, E.; Kuronen, M.; Jauhiainen, M.; Cooper, J.D.; et al. Cln5-deficiency in mice leads to microglial activation, defective myelination and changes in lipid metabolism. Neurobiol. Dis. 2011, 46, 19–29. [Google Scholar] [CrossRef]
  46. Choi, K.M.; McMahon, L.P.; Lawrence, J.C., Jr. Two Motifs in the Translational Repressor PHAS-I Required for Efficient Phosphorylation by Mammalian Target of Rapamycin and for Recognition by Raptor. J. Biol. Chem. 2003, 278, 19667–19673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Putaala, H.; Soininen, R.; Kilpeläinen, P.; Wartiovaara, J.; Tryggvason, K. The murine nephrin gene is specifically expressed in kidney, brain and pancreas: Inactivation of the gene leads to massive proteinuria and neonatal death. Hum. Mol. Genet. 2001, 10, 1–8. [Google Scholar] [CrossRef]
  48. Rantanen, M.; Palmén, T.; Pätäri, A.; Ahola, H.; Lehtonen, S.; Åström, E.; Floss, T.; Vauti, F.; Wurst, W.; Ruiz, P.; et al. Nephrin TRAP Mice Lack Slit Diaphragms and Show Fibrotic Glomeruli and Cystic Tubular Lesions. J. Am. Soc. Nephrol. 2002, 13, 1586–1594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Schweinfest, C.W.; Spyropoulos, D.D.; Henderson, K.W.; Kim, J.-H.; Chapman, J.M.; Barone, S.; Worrell, R.T.; Wang, Z.; Soleimani, M. slc26a3 (dra)-deficient Mice Display Chloride-losing Diarrhea, Enhanced Colonic Proliferation, and Distinct Up-regulation of Ion Transporters in the Colon. J. Biol. Chem. 2006, 281, 37962–37971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Forlino, A.; Piazza, R.; Tiveron, C.; Della Torre, S.; Tatangelo, L.; Bonafè, L.; Gualeni, B.; Romano, A.; Pecora, F.; Superti-Furga, A.; et al. A diastrophic dysplasia sulfate transporter (SLC26A2) mutant mouse: Morphological and biochemical characterization of the resulting chondrodysplasia phenotype. Hum. Mol. Genet. 2005, 14, 859–871. [Google Scholar] [CrossRef] [PubMed]
  51. Monti, L.; Paganini, C.; Lecci, S.; De Leonardis, F.; Hay, E.; Cohen-Solal, M.; Villani, S.; Superti-Furga, A.; Tenni, R.; Forlino, A.; et al. N -acetylcysteine treatment ameliorates the skeletal phenotype of a mouse model of diastrophic dysplasia. Hum. Mol. Genet. 2015, 24, 5570–5580. [Google Scholar] [CrossRef] [Green Version]
  52. Pennacchio, L.A.; Bouley, D.M.; Higgins, K.M.; Scott, M.P.; Noebels, J.L.; Myers, R.M. Progressive ataxia, myoclonic epilepsy and cerebellar apoptosis in cystatin B-deficient mice. Nat. Genet. 1998, 20, 251–258. [Google Scholar] [CrossRef]
  53. Lehtinen, M.K.; Tegelberg, S.; Schipper, H.; Su, H.; Zukor, H.; Manninen, O.; Kopra, O.; Joensuu, T.; Hakala, P.; Bonni, A.; et al. Cystatin B Deficiency Sensitizes Neurons to Oxidative Stress in Progressive Myoclonus Epilepsy, EPM1. J. Neurosci. 2009, 29, 5910–5915. [Google Scholar] [CrossRef] [PubMed]
  54. Messer, A.; Plummer, J.; Wong, V.; Lavail, M.M. Retinal Degeneration in Motor Neuron Degeneration (mnd) Mutant Mice. Exp. Eye Res. 1993, 57, 637–641. [Google Scholar] [CrossRef]
  55. Messer, A.; Flaherty, L. Autosomal Dominance in a Late-Onset Motor Neuron Disease in the Mouse. J. Neurogenet. 1986, 3, 345–355. [Google Scholar] [CrossRef] [PubMed]
  56. Messer, A.; Strominger, N.L.; Mazurkiewicz, J.E. Histopathology of the late-onset motor neuron degeneration (Mnd) mutant in the mouse. J. Neurogenet. 1987, 4, 201–213. [Google Scholar] [CrossRef] [PubMed]
  57. Pardo, C.A.; Rabin, B.A.; Palmer, D.N.; Price, D.L. Accumulation of the adenosine triphosphate synthase subunit C in the mnd mutant mouse. A model for neuronal ceroid lipofuscinosis. Am. J. Pathol. 1994, 144, 829–835. [Google Scholar]
  58. Johnson, T.B.; White, K.A.; Brudvig, J.J.; Cain, J.T.; Langin, L.; Pratt, M.A.; Booth, C.D.; Timm, D.J.; Davis, S.S.; Meyerink, B.; et al. AAV9 Gene Therapy Increases Lifespan and Treats Pathological and Behavioral Abnormalities in a Mouse Model of CLN8-Batten Disease. Mol. Ther. 2021, 29, 162–175. [Google Scholar] [CrossRef]
  59. Page, L.J.; Suk, J.Y.; Bazhenova, L.; Fleming, S.M.; Wood, M.; Jiang, Y.; Guo, L.T.; Mizisin, A.P.; Kisilevsky, R.; Shelton, G.D.; et al. Secretion of amyloidogenic gelsolin progressively compromises protein homeostasis leading to the intracellular aggregation of proteins. Proc. Natl. Acad. Sci. USA 2009, 106, 11125–11130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Van Overbeke, W.; Verhelle, A.; Everaert, I.; Zwaenepoel, O.; Vandekerckhove, J.; Cuvelier, C.; Derave, W.; Gettemans, J. Chaperone Nanobodies Protect Gelsolin Against MT1-MMP Degradation and Alleviate Amyloid Burden in the Gelsolin Amyloidosis Mouse Model. Mol. Ther. 2014, 22, 1768–1778. [Google Scholar] [CrossRef] [Green Version]
  61. Pai, Y.J.; Leung, K.-Y.; Savery, D.; Hutchin, T.; Prunty, H.; Heales, S.; Brosnan, M.E.; Brosnan, J.T.; Copp, A.J.; Greene, N.D.E. Glycine decarboxylase deficiency causes neural tube defects and features of non-ketotic hyperglycinemia in mice. Nat. Commun. 2015, 6, 6388. [Google Scholar] [CrossRef]
  62. Santos, C.; Pai, Y.J.; Mahmood, M.R.; Leung, K.-Y.; Savery, D.; Waddington, S.N.; Copp, A.J.; Greene, N.D.E. Impaired folate 1-carbon metabolism causes formate-preventable hydrocephalus in glycine decarboxylase–deficient mice. J. Clin. Investig. 2020, 130, 1446–1452. [Google Scholar] [CrossRef]
  63. Leung, K.-Y.; Pai, Y.J.; Chen, Q.; Santos, C.; Calvani, E.; Sudiwala, S.; Savery, D.; Ralser, M.; Gross, S.S.; Copp, A.J.; et al. Partitioning of One-Carbon Units in Folate and Methionine Metabolism Is Essential for Neural Tube Closure. Cell Rep. 2017, 21, 1795–1808. [Google Scholar] [CrossRef] [Green Version]
  64. Levéen, P.; Kotarsky, H.; Mörgelin, M.; Karikoski, R.; Elmér, E.; Fellman, V. The GRACILE mutation introduced into Bcs1l causes postnatal complex III deficiency: A viable mouse model for mitochondrial hepatopathy. Hepatology 2011, 53, 437–447. [Google Scholar] [CrossRef]
  65. Kotarsky, H.; Keller, M.; Davoudi, M.; Levéen, P.; Karikoski, R.; Enot, D.P.; Fellman, V. Metabolite Profiles Reveal Energy Failure and Impaired Beta-Oxidation in Liver of Mice with Complex III Deficiency Due to a BCS1L Mutation. PLoS ONE 2012, 7, e41156. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, T.; Lawler, A.M.; Steel, G.; Sipilä, I.; Milam, A.H.; Valle, D. Mice lacking ornithine-delta-amino-transferase have paradoxical neonatal hypoornithinaemia and retinal degeneration. Nat. Genet. 1995, 11, 185–190. [Google Scholar] [CrossRef]
  67. Smith, B.T.; Mussell, J.C.; Fleming, P.A.; Barth, J.L.; Spyropoulos, D.D.; Cooley, M.A.; Drake, C.J.; Argraves, W.S. Targeted disruption of cubilin reveals essential developmental roles in the structure and function of endoderm and in somite formation. BMC Dev. Biol. 2006, 6, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Nikkanen, J.; Forsström, S.; Euro, L.; Paetau, I.; Kohnz, R.A.; Wang, L.; Chilov, D.; Viinamäki, J.; Roivainen, A.; Marjamäki, P.; et al. Mitochondrial DNA Replication Defects Disturb Cellular dNTP Pools and Remodel One-Carbon Metabolism. Cell Metab. 2016, 23, 635–648. [Google Scholar] [CrossRef] [Green Version]
  69. Buzkova, J.; Nikkanen, J.; Ahola, S.; Hakonen, A.H.; Sevastianova, K.; Hovinen, T.; Yki-Järvinen, H.; Pietiläinen, K.H.; Lönnqvist, T.; Velagapudi, V.; et al. Metabolomes of mitochondrial diseases and inclusion body myositis patients: Treatment targets and biomarkers. EMBO Mol. Med. 2018, 10, e9091. [Google Scholar] [CrossRef] [PubMed]
  70. Khare, T.; Khare, S.; Angdisen, J.J.; Zhang, Q.; Stuckel, A.; Mooney, B.P.; Ridenhour, S.E.; Gitan, R.S.; Hammoud, G.M.; Ibdah, J.A. Defects in long-chain 3-hydroxy acyl-CoA dehydrogenase lead to hepatocellular carcinoma: A novel etiology of hepatocellular carcinoma. Int. J. Cancer 2020, 147, 1461–1473. [Google Scholar] [CrossRef]
  71. Sperandeo, M.P.; Annunziata, P.; Bozzato, A.; Piccolo, P.; Maiuri, L.; D’Armiento, M.; Ballabio, A.; Corso, G.; Andria, G.; Borsani, G.; et al. Slc7a7 disruption causes fetal growth retardation by downregulating Igf1 in the mouse model of lysinuric protein intolerance. Am. J. Physiol.-Cell Physiol. 2007, 293, C191–C198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Stroup, B.M.; Marom, R.; Li, X.; Hsu, C.-W.; Chang, C.-Y.; Truong, L.D.; Dawson, B.; Grafe, I.; Chen, Y.; Jiang, M.-M.; et al. A global Slc7a7 knockout mouse model demonstrates characteristic phenotypes of human lysinuric protein intolerance. Hum. Mol. Genet. 2020, 29, 2171–2184. [Google Scholar] [CrossRef]
  73. Liu, J.; Yang, Y.; Li, X.; Zhang, P.; Qi, Y.; Hu, H. Cellular and Molecular Characterization of Abnormal Brain Development in Protein O-Mannose N-Acetylglucosaminyltransferase 1 Knockout Mice. Methods Enzymol. 2010, 479, 353–366. [Google Scholar] [CrossRef]
  74. Liu, J.; Ball, S.L.; Yang, Y.; Mei, P.; Zhang, L.; Shi, H.; Kaminski, H.J.; Lemmon, V.P.; Hu, H. A genetic model for muscle–eye–brain disease in mice lacking protein O-mannose 1,2-N-acetylglucosaminyltransferase (POMGnT1). Mech. Dev. 2006, 123, 228–240. [Google Scholar] [CrossRef] [PubMed]
  75. Miyagoe-Suzuki, Y.; Masubuchi, N.; Miyamoto, K.; Wada, M.R.; Yuasa, S.; Saito, F.; Matsumura, K.; Kanesaki, H.; Kudo, A.; Manya, H.; et al. Reduced proliferative activity of primary POMGnT1-null myoblasts in vitro. Mech. Dev. 2009, 126, 107–116. [Google Scholar] [CrossRef] [PubMed]
  76. Cui, C.; Chatterjee, B.; Francis, D.; Yu, Q.; SanAgustin, J.T.; Francis, R.; Tansey, T.; Henry, C.; Wang, B.; Lemley, B.; et al. Disruption of Mks1 localization to the mother centriole causes cilia defects and developmental malformations in Meckel-Gruber syndrome. Dis. Model. Mech. 2011, 4, 43–56. [Google Scholar] [CrossRef] [Green Version]
  77. Kettunen, K.M.; Karikoski, R.; Hämäläinen, R.H.; Toivonen, T.T.; Antonenkov, V.D.; Kulesskaya, N.; Voikar, V.; Hölttä-Vuori, M.; Ikonen, E.; Sainio, K.; et al. Trim37-deficient mice recapitulate several features of the multi-organ disorder Mulibrey nanism. Biol. Open 2016, 5, 584–595. [Google Scholar] [CrossRef] [Green Version]
  78. Dierich, A.; Sairam, M.R.; Monaco, L.; Fimia, G.M.; Gansmuller, A.; LeMeur, M.; Sassone-Corsi, P. Impairing follicle-stimulating hormone (FSH) signaling in vivo: Targeted disruption of the FSH receptor leads to aberrant gametogenesis and hormonal imbalance. Proc. Natl. Acad. Sci. USA 1998, 95, 13612–13617. [Google Scholar] [CrossRef] [Green Version]
  79. Krishnamurthy, H.; Danilovich, N.; Morales, C.R.; Sairam, M.R. Qualitative and quantitative decline in spermatogenesis of the follicle-stimulating hormone receptor knockout (FORKO) mouse. Biol. Reprod. 2000, 62, 1146–1159. [Google Scholar] [CrossRef] [Green Version]
  80. Abel, M.H.; Wootton, A.N.; Wilkins, V.; Huhtaniemi, I.; Knight, P.G.; Charlton, H.M. The Effect of a Null Mutation in the Follicle-Stimulating Hormone Receptor Gene on Mouse Reproduction. Endocrinology 2000, 141, 1795–1803. [Google Scholar] [CrossRef]
  81. Kaifu, T.; Nakahara, J.; Inui, M.; Mishima, K.; Momiyama, T.; Kaji, M.; Sugahara, A.; Koito, H.; Ujike-Asai, A.; Nakamura, A.; et al. Osteopetrosis and thalamic hypomyelinosis with synaptic degeneration in DAP12-deficient mice. J. Clin. Investig. 2003, 111, 323–332. [Google Scholar] [CrossRef] [Green Version]
  82. Hayashi, M.; Nakashima, T.; Taniguchi, M.; Kodama, T.; Kumanogoh, A.; Takayanagi, H. Osteoprotection by semaphorin 3A. Nature 2012, 485, 69–74. [Google Scholar] [CrossRef]
  83. Ichikawa, K.; Noda, T.; Furuichi, Y. Preparation of the gene targeted knockout mice for human premature aging diseases, Werner syndrome, and Rothmund-Thomson syndrome caused by the mutation of DNA helicases. Nihon Yakurigaku Zasshi 2002, 119, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mann, M.B.; Hodges, C.A.; Barnes, E.; Vogel, H.; Hassold, T.J.; Luo, G. Defective sister-chromatid cohesion, aneuploidy and cancer predisposition in a mouse model of type II Rothmund-Thomson syndrome. Hum. Mol. Genet. 2005, 14, 813–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hoki, Y.; Araki, R.; Fujimori, A.; Ohhata, T.; Koseki, H.; Fukumura, R.; Nakamura, M.; Takahashi, H.; Noda, Y.; Kito, S.; et al. Growth retardation and skin abnormalities of the Recql4-deficient mouse. Hum. Mol. Genet. 2003, 12, 2293–2299. [Google Scholar] [CrossRef] [PubMed]
  86. Weber, B.H.; Schrewe, H.; Molday, L.L.; Gehrig, A.; White, K.L.; Seeliger, M.W.; Jaissle, G.B.; Friedburg, C.; Tamm, E.; Molday, R.S. Inactivation of the murine X-linked juvenile retinoschisis gene, Rs1h, suggests a role of retinoschisin in retinal cell layer organization and synaptic structure. Proc. Natl. Acad. Sci. USA 2002, 99, 6222–6227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Apaolaza, P.; del Pozo-Rodríguez, A.; Solinís, M.A.; Rodríguez, J.M.; Friedrich, U.; Torrecilla, J.; Weber, B.H.; Rodríguez-Gascón, A. Structural recovery of the retina in a retinoschisin-deficient mouse after gene replacement therapy by solid lipid nanoparticles. Biomaterials 2016, 90, 40–49. [Google Scholar] [CrossRef]
  88. Walia, S.; Fishman, G.A.; Molday, R.S.; Dyka, F.M.; Kumar, N.M.; Ehlinger, M.A.; Stone, E.M. Relation of Response to Treatment with Dorzolamide in X-Linked Retinoschisis to the Mechanism of Functional Loss in Retinoschisin. Am. J. Ophthalmol. 2009, 147, 111–115.e1. [Google Scholar] [CrossRef] [Green Version]
  89. Prolo, L.M.; Vogel, H.; Reimer, R.J. The Lysosomal Sialic Acid Transporter Sialin Is Required for Normal CNS Myelination. J. Neurosci. 2009, 29, 15355–15365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Stroobants, S.; Van Acker, N.G.; Verheijen, F.W.; Goris, I.; Daneels, G.F.; Schot, R.; Verbeek, E.; Knaapen, M.W.; De Bondt, A.; Göhlmann, H.W.; et al. Progressive leukoencephalopathy impairs neurobehavioral development in sialin-deficient mice. Exp. Neurol. 2017, 291, 106–119. [Google Scholar] [CrossRef]
  91. May, S.R.; Stewart, N.J.; Chang, W.; Peterson, A.S. A Titin mutation defines roles for circulation in endothelial morphogenesis. Dev. Biol. 2004, 270, 31–46. [Google Scholar] [CrossRef]
  92. Peng, J.; Raddatz, K.; Labeit, S.; Granzier, H.; Gotthardt, M. Muscle atrophy in Titin M-line deficient mice. J. Muscle Res. Cell Motil. 2005, 26, 381–388. [Google Scholar] [CrossRef] [PubMed]
  93. Geng, R.; Geller, S.F.; Hayashi, T.; Ray, C.A.; Reh, T.A.; Bermingham-McDonogh, O.; Jones, S.M.; Wright, C.G.; Melki, S.; Imanishi, Y.; et al. Usher syndrome IIIA gene clarin-1 is essential for hair cell function and associated neural activation. Hum. Mol. Genet. 2009, 18, 2748–2760. [Google Scholar] [CrossRef] [Green Version]
  94. Geng, R.; Omar, A.; Gopal, S.R.; Chen, D.H.-C.; Stepanyan, R.; Basch, M.L.; Dinculescu, A.; Furness, D.N.; Saperstein, D.; Hauswirth, W.; et al. Modeling and Preventing Progressive Hearing Loss in Usher Syndrome III. Sci. Rep. 2017, 7, 13480. [Google Scholar] [CrossRef]
  95. Pollitt, R.J.; Jenner, F.A.; Merskey, H. Aspartylglycosaminuria: An Inborn Error of Metabolism Associated with Mental Defect. Lancet 1968, 292, 253–255. [Google Scholar] [CrossRef]
  96. Arvio, M.; Mononen, I. Aspartylglycosaminuria: A review. Orphanet J. Rare Dis. 2016, 11, 162. [Google Scholar] [CrossRef] [Green Version]
  97. Palo, J.; Mattsson, K. Eleven new cases of aspartylglucosaminuria. J. Ment. Defic. Res. 1970, 14, 168–173. [Google Scholar] [CrossRef]
  98. Goodspeed, K.; Feng, C.; Laine, M.; Lund, T.C. Aspartylglucosaminuria: Clinical Presentation and Potential Therapies. J. Child Neurol. 2021, 36, 403–414. [Google Scholar] [CrossRef]
  99. Arvio, M.; Autio, S.; Louhiala, P. Early clinical symptoms and incidence of aspartylglucosaminuria in Finland. Acta Paediatr. 1993, 82, 587–589. [Google Scholar] [CrossRef]
  100. Arvio, M.; Arvio, P.; Hurmerinta, K.; Pirinen, S.; Sillanpää, M. Reduction in head size in patients with aspartylglucosaminuria. Acta Neurol. Scand. 2005, 112, 335–337. [Google Scholar] [CrossRef] [PubMed]
  101. Anna, T.; Nina, B.; Antti, H.; Eero, S.; Laura, Å.; Taina, A. White Matter Microstructure and Subcortical Gray Matter Structure Volumes in Aspartylglucosaminuria; a 5-Year Follow-Up Brain MRI Study of an Adolescent with Aspartylglucosaminuria and His Healthy Twin Brother. JIMD Rep. 2017, 35, 105–115. [Google Scholar] [CrossRef] [Green Version]
  102. Sairanen, V.; Tokola, A.; Tikkanen, R.; Laine, M.; Autti, T. Statistical Permutation Test Reveals Progressive and Region-Specific Iron Accumulation in the Thalami of Children with Aspartylglucosaminuria. Brain Sci. 2020, 10, 677. [Google Scholar] [CrossRef]
  103. Tokola, A.; Laine, M.; Tikkanen, R.; Autti, T. Susceptibility-Weighted Imaging Findings in Aspartylglucosaminuria. AJNR Am. J. Neuroradiol. 2019, 40, 1850–1854. [Google Scholar] [CrossRef] [PubMed]
  104. Arvio, P.; Arvio, M. Progressive nature of aspartylglucosaminuria. Acta Paediatr. 2002, 91, 255–257. [Google Scholar] [CrossRef] [PubMed]
  105. Fisher, K.J.; Tollersrud, O.K.; Aronson, N.N., Jr. Cloning and sequence analysis of a cDNA for human glycosylasparaginase. A single gene encodes the subunits of this lysosomal amidase. FEBS Lett. 1990, 269, 440–444. [Google Scholar] [CrossRef] [Green Version]
  106. Ikonen, E.; Baumann, M.; Grön, K.; Syvänen, A.C.; Enomaa, N.; Halila, R.; Aula, P.; Peltonen, L. Aspartylglucosaminuria: cDNA encoding human aspartylglucosaminidase and the missense mutation causing the disease. EMBO J. 1991, 10, 51–58. [Google Scholar] [CrossRef] [PubMed]
  107. Mononen, I.; Heisterkamp, N.; Kaartinen, V.; Williams, J.C.; Yates, J.R., 3rd; Griffin, P.R.; Hood, L.E.; Groffen, J. Aspartylglycosaminuria in the Finnish population: Identification of two point mutations in the heavy chain of glycoasparaginase. Proc. Natl. Acad. Sci. USA 1991, 88, 2941–2945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Banning, A.; Schiff, M.; Tikkanen, R. Amlexanox provides a potential therapy for nonsense mutations in the lysosomal storage disorder Aspartylglucosaminuria. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2018, 1864, 668–675. [Google Scholar] [CrossRef] [PubMed]
  109. Fisher, K.J.; Aronson, N.N., Jr. Characterization of the mutation responsible for aspartylglucosaminuria in three Finnish patients. Amino acid substitution Cys163—Ser abolishes the activity of lysosomal glycosylasparaginase and its conversion into subunits. J. Biol. Chem. 1991, 266, 12105–12113. [Google Scholar] [CrossRef]
  110. McCormack, A.L.; Mononen, I.; Kaartinen, V.; Yates, J.R., 3rd. Localization of the Disulfide Bond Involved in Post-translational Processing of Glycosylasparaginase and Disrupted by a Mutation in the Finnish-type Aspartylglycosaminuria. J. Biol. Chem. 1995, 270, 3212–3215. [Google Scholar] [CrossRef] [Green Version]
  111. Sui, L.; Lakshminarasimhan, D.; Pande, S.; Guo, H.-C. Structural Basis of a Point Mutation that Causes the Genetic Disease Aspartylglucosaminuria. Structure 2014, 22, 1855–1861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Pande, S.; Bizilj, W.; Guo, H.-C. Biochemical and structural insights into an allelic variant causing the lysosomal storage disorder–aspartylglucosaminuria. FEBS Lett. 2018, 592, 2550–2561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Mononen, I.; Fisher, K.J.; Kaartinen, V.; Aronson, N.N., Jr. Aspartylglycosaminuria: Protein chemistry and molecular biology of the most common lysosomal storage disorder of glycoprotein degradation. FASEB J. 1993, 7, 1247–1256. [Google Scholar] [CrossRef]
  114. Arvio, M.; Sauna-Aho, O.; Peippo, M. Bone marrow transplantation for aspartylglucosaminuria: Follow-up study of transplanted and non-transplanted patients. J. Pediatr. 2001, 138, 288–290. [Google Scholar] [CrossRef] [PubMed]
  115. Ringdén, O.; Remberger, M.; Svahn, B.-M.; Barkholt, L.; Mattsson, J.; Aschan, J.; Le Blanc, K.; Gustafsson, B.; Hassan, Z.; Omazic, B.; et al. Allogeneic Hematopoietic Stem Cell Transplantation for Inherited Disorders: Experience in a Single Center. Transplantation 2006, 81, 718–725. [Google Scholar] [CrossRef] [PubMed]
  116. Virta, S.; Rapola, J.; Jalanko, A.; Laine, M. Use of nonviral promoters in adenovirus-mediated gene therapy: Reduction of lysosomal storage in the aspartylglucosaminuria mouse. J. Gene Med. 2006, 8, 699–706. [Google Scholar] [CrossRef]
  117. Peltola, M.; Kyttälä, A.; Heinonen, O.; Rapola, J.; Paunio, T.; Revah, F.; Peltonen, L.; Jalanko, A. Adenovirus-mediated gene transfer results in decreased lysosomal storage in brain and total correction in liver of aspartylglucosaminuria (AGU) mouse. Gene Ther. 1998, 5, 1314–1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Laine, M.; Richter, J.; Fahlman, C.; Rapola, J.; Renlund, M.; Peltonen, L.; Karlsson, S.; Jalanko, A. Correction of peripheral lysosomal accumulation in mice with aspartylglucosaminuria by bone marrow transplantation. Exp. Hematol. 1999, 27, 1467–1474. [Google Scholar] [CrossRef]
  119. Malm, G.; Månsson, J.-E.; Winiarski, J.; Mosskin, M.; Ringdén, O. Five-Year Follow-Up of Two Siblings with Aspartylglucosaminuria Undergoing Allogeneic Stem-Cell Transplantation from Unrelated Donors. Transplantation 2004, 78, 415–419. [Google Scholar] [CrossRef]
  120. McKusick, V.A.; Eldridge, R.; Hostetler, J.A.; Ruangwit, U.; Egeland, J.A. Dwarfism in the Amish. II. Cartilage-hair hypoplasia. Bull. Johns Hopkins Hosp. 1965, 116, 285–326. [Google Scholar]
  121. Mäkitie, O.; Kaitila, I. Cartilage-hair hypoplasia—Clinical manifestations in 108 Finnish patients. Eur. J. Pediatr. 1993, 152, 211–217. [Google Scholar] [CrossRef]
  122. Mäkitie, O.M.; Tapanainen, P.J.; Dunkel, L.; Siimes, M.A. Impaired spermatogenesis: An unrecognized feature of cartilage-hair hypoplasia. Ann. Med. 2001, 33, 201–205. [Google Scholar] [CrossRef]
  123. Vakkilainen, S.; Taskinen, M.; Klemetti, P.; Pukkala, E.; Mäkitie, O. A 30-Year Prospective Follow-Up Study Reveals Risk Factors for Early Death in Cartilage-Hair Hypoplasia. Front. Immunol. 2019, 10, 1581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mäkitie, O. Cartilage-hair hypoplasia in Finland: Epidemiological and genetic aspects of 107 patients. J. Med. Genet. 1992, 29, 652–655. [Google Scholar] [CrossRef] [Green Version]
  125. Ridanpää, M.; van Eenennaam, H.; Pelin, K.; Chadwick, R.; Johnson, C.; Yuan, B.; Vanvenrooij, W.; Pruijn, G.; Salmela, R.; Rockas, S.; et al. Mutations in the RNA Component of RNase MRP Cause a Pleiotropic Human Disease, Cartilage-Hair Hypoplasia. Cell 2001, 104, 195–203. [Google Scholar] [CrossRef] [Green Version]
  126. Ridanpää, M.; Sistonen, P.; Rockas, S.; Rimoin, D.L.; Mäkitie, O.; Kaitila, I. Worldwide mutation spectrum in cartilage-hair hypoplasia: Ancient founder origin of the major70A→G mutation of the untranslated RMRP. Eur. J. Hum. Genet. 2002, 10, 439–447. [Google Scholar] [CrossRef] [Green Version]
  127. Ridanpää, M.; Jain, P.; McKusick, V.A.; Francomano, C.A.; Kaitila, I. The major mutation in the RMRP gene causing CHH among the Amish is the same as that found in most Finnish cases. Am. J. Med. Genet. Part C Semin. Med. Genet. 2003, 121C, 81–83. [Google Scholar] [CrossRef]
  128. Hussen, B.M.; Azimi, T.; Hidayat, H.J.; Taheri, M.; Ghafouri-Fard, S. Long Non-coding RNA RMRP in the Pathogenesis of Human Disorders. Front. Cell Dev. Biol. 2021, 9, 1130. [Google Scholar] [CrossRef]
  129. Chen, Y.; Hao, Q.; Wang, S.; Cao, M.; Huang, Y.; Weng, X.; Wang, J.; Zhang, Z.; He, X.; Lu, H.; et al. Inactivation of the tumor suppressor p53 by long noncoding RNA RMRP. Proc. Natl. Acad. Sci. USA 2021, 118, e2026813118. [Google Scholar] [CrossRef]
  130. Salonen, R.; Herva, R.; Norio, R. The hydrolethalus syndrome: Delineation of a “new”, lethal malformation syndrome based on 28 patients. Clin. Genet. 2008, 19, 321–330. [Google Scholar] [CrossRef] [PubMed]
  131. Payan, P.; Girard, J.P. Evidence for a Na+/NH4+ exchange in the gill of trout adapted to sea water: Adrenergic control. C. R. Acad. Hebd. Seances Acad. Sci. D 1978, 286, 335–338. [Google Scholar]
  132. Paetau, A.; Honkala, H.; Salonen, R.; Ignatius, J.; Kestilä, M.; Herva, R. Hydrolethalus syndrome: Neuropathology of 21 cases confirmed by HYLS1 gene mutation analysis. J. Neuropathol. Exp. Neurol. 2008, 67, 750–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Shotelersuk, V.; Punyavoravud, V.; Phudhichareonrat, S.; Kukulprasong, A. An Asian girl with a ‘milder’ form of the Hydrolethalus syndrome. Clin. Dysmorphol. 2001, 10, 51–55. [Google Scholar] [CrossRef] [PubMed]
  134. Putoux, A.; Thomas, S.; Coene, K.L.; Davis, E.E.; Alanay, Y.; Ogur, G.; Uz, E.; Buzas, D.; Gomes, C.; Patrier, S.; et al. KIF7 mutations cause fetal hydrolethalus and acrocallosal syndromes. Nat. Genet. 2011, 43, 601–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Valente, E.M.; Rosti, R.O.; Gibbs, E.; Gleeson, J.G. Primary cilia in neurodevelopmental disorders. Nat. Rev. Neurol. 2014, 10, 27–36. [Google Scholar] [CrossRef] [Green Version]
  136. Bachman, H.; Clark, R.D.; Salahi, W. Holoprosencephaly and polydactyly: A possible expression of the hydrolethalus syndrome. J. Med. Genet. 1990, 27, 50–52. [Google Scholar] [CrossRef] [Green Version]
  137. de Ravel, T.J.; van der Griendt, M.C.; Evan, P.; Wright, C.A. Hydrolethalus syndrome in a non-Finnish family: Confirmation of the entity and early prenatal diagnosis. Prenat. Diagn. 1999, 19, 279–281. [Google Scholar] [CrossRef]
  138. Chan, B.C.; Shek, T.W.; Lee, C.P. First-trimester diagnosis of hydrolethalus syndrome in a Chinese family. Prenat. Diagn. 2004, 24, 587–590. [Google Scholar] [CrossRef]
  139. Mee, L.; Honkala, H.; Kopra, O.; Vesa, J.; Finnilä, S.; Visapää, I.; Sang, T.-K.; Jackson, G.R.; Salonen, R.; Kestilä, M.; et al. Hydrolethalus syndrome is caused by a missense mutation in a novel gene gene HYLS1. Hum. Mol. Genet. 2005, 14, 1475–1488. [Google Scholar] [CrossRef] [Green Version]
  140. Dammermann, A.; Pemble, H.; Mitchell, B.J.; McLeod, I.; Yates, J.R., 3rd; Kintner, C.; Desai, A.B.; Oegema, K. The hydrolethalus syndrome protein HYLS-1 links core centriole structure to cilia formation. Genes Dev. 2009, 23, 2046–2059. [Google Scholar] [CrossRef] [Green Version]
  141. Wei, Q.; Zhang, Y.; Schouteden, C.; Zhang, Y.; Zhang, Q.; Dong, J.; Wonesch, V.; Ling, K.; Dammermann, A.; Hu, J. The hydrolethalus syndrome protein HYLS-1 regulates formation of the ciliary gate. Nat. Commun. 2016, 7, 12437. [Google Scholar] [CrossRef] [Green Version]
  142. Hou, Y.; Wu, Z.; Zhang, Y.; Chen, H.; Hu, J.; Guo, Y.; Peng, Y.; Wei, Q. Functional Analysis of Hydrolethalus Syndrome Protein HYLS1 in Ciliogenesis and Spermatogenesis in Drosophila. Front. Cell Dev. Biol. 2020, 8, 301. [Google Scholar] [CrossRef]
  143. Honkala, H.; Lahtela, J.; Fox, H.; Gentile, M.; Pakkasjärvi, N.; Salonen, R.; Wartiovaara, K.; Jauhiainen, M.; Kestilä, M. Unraveling the disease pathogenesis behind lethal hydrolethalus syndrome revealed multiple changes in molecular and cellular level. PathoGenetics 2009, 2, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Elnahry, A.G.; Tripathy, K. Gyrate Atrophy of the Choroid and Retina. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  145. Montioli, R.; Bellezza, I.; Desbats, M.A.; Borri Voltattorni, C.; Salviati, L.; Cellini, B. Deficit of human ornithine aminotransferase in gyrate atrophy: Molecular, cellular, and clinical aspects. Biochim. Biophys. Acta (BBA) Proteins Proteom. 2021, 1869, 140555. [Google Scholar] [CrossRef] [PubMed]
  146. Jalali, H.; Najafi, M.; Khoshaeen, A.; Mahdavi, M.R.; Mahdavi, M. First report of c.425-1G>A mutation in ornithine aminotransferase gene causing gyrate atrophy of the choroid and retina with hyperornithinemia. Eur. J. Ophthalmol. 2020. [Google Scholar] [CrossRef] [PubMed]
  147. Hirvasniemi, A.; Karumo, J. Neuroradiological findings in the northern epilepsy syndrome. Acta Neurol. Scand. 2009, 90, 388–393. [Google Scholar] [CrossRef]
  148. Di Ronza, A.; Bajaj, L.; Sharma, J.; Sanagasetti, D.; Lotfi, P.; Adamski, C.J.; Collette, J.; Palmieri, M.; Amawi, A.; Popp, L.; et al. CLN8 is an endoplasmic reticulum cargo receptor that regulates lysosome biogenesis. Nat. Cell Biol. 2018, 20, 1370–1377. [Google Scholar] [CrossRef] [PubMed]
  149. Pesaola, F.; Quassollo, G.; Venier, A.C.; De Paul, A.L.; Noher, I.; Bisbal, M. The neuronal ceroid lipofuscinosis-related protein CLN8 regulates endo-lysosomal dynamics and dendritic morphology. Biol. Cell 2021, 113, 419–437. [Google Scholar] [CrossRef]
  150. Brody, L.C.; Mitchell, G.A.; Obie, C.; Michaud, J.; Steel, G.; Fontaine, G.; Robert, M.F.; Sipila, I.; Kaiser-Kupfer, M.; Valle, D. Ornithine delta-aminotransferase mutations in gyrate atrophy. Allelic heterogeneity and functional consequences. J. Biol. Chem. 1992, 267, 3302–3307. [Google Scholar] [CrossRef]
  151. Bisaillon, J.J.; Radden, L.A., 2nd; Szabo, E.T.; Hughes, S.R.; Feliciano, A.M.; Nesta, A.V.; Petrovic, B.; Palanza, K.M.; Lancinskas, D.; Szmurlo, T.A.; et al. The retarded hair growth (rhg) mutation in mice is an allele of ornithine aminotransferase (Oat). Mol. Genet. Metab. Rep. 2014, 1, 378–390. [Google Scholar] [CrossRef]
  152. Herva, R.; Tyynelä, J.; Hirvasniemi, A.; Syrjäkallio-Ylitalo, M.; Haltia, M. Northern Epilepsy: A Novel Form of Neuronal Ceroid-Lipofuscinosis. Brain Pathol. 2000, 10, 215–222. [Google Scholar] [CrossRef]
  153. Hirvasniemi, A.; Lang, H.; Lehesjoki, A.E.; Leisti, J. Northern epilepsy syndrome: An inherited childhood onset epilepsy with associated mental deterioration. J. Med. Genet. 1994, 31, 177–182. [Google Scholar] [CrossRef] [Green Version]
  154. Ranta, S.; Zhang, Y.; Ross, B.M.; Lonka, L.; Takkunen, E.; Messer, A.; Sharp, J.D.; Wheeler, R.B.; Kusumi, K.; Mole, S.; et al. The neuronal ceroid lipofuscinoses in human EPMR and mnd mutant mice are associated with mutations in CLN8. Nat. Genet. 1999, 23, 233–236. [Google Scholar] [CrossRef] [PubMed]
  155. Bronson, R.T.; Lake, B.D.; Cook, S.; Taylor, S.; Davisson, M.T. Motor neuron degeneration of mice is a model of neuronal ceroid lipofuscinosis (Batten’s disease). Ann. Neurol. 1993, 33, 381–385. [Google Scholar] [CrossRef] [PubMed]
  156. Qin, L.; Liu, X.; Sun, Q.; Fan, Z.; Xia, D.; Ding, G.; Ong, H.L.; Adams, D.; Gahl, W.A.; Zheng, C.; et al. Sialin (SLC17A5) functions as a nitrate transporter in the plasma membrane. Proc. Natl. Acad. Sci. USA 2012, 109, 13434–13439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Ribas, G.S.; Vargas, C.R. Evidence that Oxidative Disbalance and Mitochondrial Dysfunction are Involved in the Pathophysiology of Fatty Acid Oxidation Disorders. Cell. Mol. Neurobiol. 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  158. Ii, J.L.M.; Norris, M.; Kanungo, S. Fatty acid oxidation disorders. Ann. Transl. Med. 2018, 6, 473. [Google Scholar] [CrossRef]
  159. Varho, T.T.; Alajoki, L.E.; Posti, K.M.; Korhonen, T.T.; Renlund, M.G.; Nyman, S.R.; Sillanpää, M.; Aula, P.P. Phenotypic spectrum of Salla disease, a free sialic acid storage disorder. Pediatr. Neurol. 2002, 26, 267–273. [Google Scholar] [CrossRef]
  160. Morse, R.P.; Kleta, R.; Alroy, J.; Gahl, W.A. Novel Form of Intermediate Salla Disease: Clinical and Neuroimaging Features. J. Child Neurol. 2005, 20, 814–816. [Google Scholar] [CrossRef]
  161. Verheijen, F.W.; Verbeek, E.; Aula, N.; Beerens, C.E.; Havelaar, A.C.; Joosse, M.; Peltonen, L.; Aula, P.; Galjaard, H.; Van Der Spek, P.J.; et al. A new gene, encoding an anion transporter, is mutated in sialic acid storage diseases. Nat. Genet. 1999, 23, 462–465. [Google Scholar] [CrossRef]
  162. Aula, N.; Salomäki, P.; Timonen, R.; Verheijen, F.; Mancini, G.; Månsson, J.-E.; Aula, P.; Peltonen, L. The Spectrum of SLC17A5-Gene Mutations Resulting in Free Sialic Acid–Storage Diseases Indicates Some Genotype-Phenotype Correlation. Am. J. Hum. Genet. 2000, 67, 832–840. [Google Scholar] [CrossRef] [Green Version]
  163. Aula, N.; Kopra, O.; Jalanko, A.; Peltonen, L. Sialin expression in the CNS implicates extralysosomal function in neurons. Neurobiol. Dis. 2004, 15, 251–261. [Google Scholar] [CrossRef] [PubMed]
  164. Howe, K.L.; Achuthan, P.; Allen, J.; Allen, J.; Alvarez-Jarreta, J.; Amode, M.R.; Armean, I.M.; Azov, A.G.; Bennett, R.; Bhai, J.; et al. Ensembl 2021. Nucleic Acids Res. 2020, 49, D884–D891. [Google Scholar] [CrossRef] [PubMed]
  165. Robak, L.A.; Jansen, I.E.; Van Rooij, J.; Uitterlinden, A.G.; Kraaij, R.; Jankovic, J.; Heutink, P.; Shulman, J.M.; A Nalls, M.; Plagnol, V.; et al. Excessive burden of lysosomal storage disorder gene variants in Parkinson’s disease. Brain 2017, 140, 3191–3203. [Google Scholar] [CrossRef]
  166. Wilcken, B.; Leung, K.-C.; Hammond, J.; Kamath, R.; Leonard, J.V. Pregnancy and fetal long-chain 3-hydroxyacyl coenzyme A dehydrogenase deficiency. Lancet 1993, 341, 407–408. [Google Scholar] [CrossRef]
  167. Sims, H.F.; Brackett, J.C.; Powell, C.K.; Treem, W.R.; Hale, D.E.; Bennett, M.J.; Gibson, B.; Shapiro, S.; Strauss, A.W. The molecular basis of pediatric long chain 3-hydroxyacyl-CoA dehydrogenase deficiency associated with maternal acute fatty liver of pregnancy. Proc. Natl. Acad. Sci. USA 1995, 92, 841–845. [Google Scholar] [CrossRef] [Green Version]
  168. Treem, W.R.; Shoup, M.E.; Hale, D.E.; Bennett, M.J.; Rinaldo, P.; Millington, D.S.; Stanley, C.A.; Riely, C.A.; Hyams, J.S. Acute fatty liver of pregnancy, hemolysis, elevated liver enzymes, and low platelets syndrome, and long chain 3-hydroxyacyl-coenzyme A dehydrogenase deficiency. Am. J. Gastroenterol. 1996, 91, 2293–2300. [Google Scholar] [PubMed]
  169. Immonen, T.; Turanlahti, M.; Paganus, A.; Keskinen, P.; Tyni, T.; Lapatto, R. Earlier diagnosis and strict diets improve the survival rate and clinical course of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Acta Paediatr. 2016, 105, 549–554. [Google Scholar] [CrossRef]
  170. Ijlst, L.; Wanders, R.J.A.; Ushikubo, S.; Kamijo, T.; Hashimoto, T. Molecular basis of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency: Identification of the major disease-causing mutation in the alpha-subunit of the mitochondrial trifunctional protein. Biochim. Biophys. Acta (BBA) Lipids Lipid Metab. 1994, 1215, 347–350. [Google Scholar] [CrossRef]
  171. Olpin, S.E.; Clark, S.; Andresen, B.S.; Bischoff, C.; Olsen, R.; Gregersen, N.; Chakrapani, A.; Downing, M.; Manning, N.J.; Sharrard, M.; et al. Biochemical, clinical and molecular findings in LCHAD and general mitochondrial trifunctional protein deficiency. J. Inherit. Metab. Dis. 2005, 28, 533–544. [Google Scholar] [CrossRef]
  172. Tyni, T.; Rapola, J.; Paetau, A.; Palotie, A.; Pihko, H. Pathology of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency caused by the G1528C mutation. Pediatr. Pathol. Lab. Med. 1997, 17, 427–447. [Google Scholar] [CrossRef]
  173. Tyni, T.; Majander, A.; Kalimo, H.; Rapola, J.; Pihko, H. Pathology of skeletal muscle and impaired respiratory chain function in long-chain 3-hydroxyacyl-coa dehydrogenase deficiency with the G1528C mutation. Neuromuscul. Disord. 1996, 6, 327–337. [Google Scholar] [CrossRef]
  174. Cecatto, C.; Dos Santos Godoy, K.; da Silva, J.C.; Amaral, A.U.; Wajner, M. Disturbance of mitochondrial functions provoked by the major long-chain 3-hydroxylated fatty acids accumulating in MTP and LCHAD deficiencies in skeletal muscle. Toxicol. In Vitro 2016, 36, 1–9. [Google Scholar] [CrossRef]
  175. Hickmann, F.H.; Cecatto, C.; Kleemann, D.; Monteiro, W.O.; Castilho, R.F.; Amaral, A.U.; Wajner, M. Uncoupling, metabolic inhibition and induction of mitochondrial permeability transition in rat liver mitochondria caused by the major long-chain hydroxyl monocarboxylic fatty acids accumulating in LCHAD deficiency. Biochim. Biophys. Acta 2015, 1847, 620–628. [Google Scholar] [CrossRef] [Green Version]
  176. Ibdah, J.A.; Paul, H.; Zhao, Y.; Binford, S.; Salleng, K.; Cline, M.; Matern, D.; Bennett, M.J.; Rinaldo, P.; Strauss, A.W. Lack of mitochondrial trifunctional protein in mice causes neonatal hypoglycemia and sudden death. J. Clin. Investig. 2001, 107, 1403–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Barré-Sinoussi, F.; Montagutelli, X. Animal models are essential to biological research: Issues and perspectives. Future Sci. OA 2015, 1, Fso63. [Google Scholar] [CrossRef] [Green Version]
  178. Russell, W.M.S.; Burch, R.L. The Principles of Humane Experimental Technique; Methuen: London, UK, 1959; reprinted in 1992. [Google Scholar]
  179. Mehravar, M.; Shirazi, A.; Nazari, M.; Banan, M. Mosaicism in CRISPR/Cas9-mediated genome editing. Dev. Biol. 2019, 445, 156–162. [Google Scholar] [CrossRef] [PubMed]
  180. Oram, J.F. Tangier disease and ABCA1. Biochim. Biophys. Acta 2000, 1529, 321–330. [Google Scholar] [CrossRef]
  181. Hooper, A.J.; Hegele, R.A.; Burnett, J.R. Tangier disease: Update for 2020. Curr. Opin. Lipidol. 2020, 31, 80–84. [Google Scholar] [CrossRef]
  182. Jacobo-Albavera, L.; Domínguez-Pérez, M.; Medina-Leyte, D.; González-Garrido, A.; Villarreal-Molina, T. The Role of the ATP-Binding Cassette A1 (ABCA1) in Human Disease. Int. J. Mol. Sci. 2021, 22, 1593. [Google Scholar] [CrossRef]
  183. Zhu, F.; Nair, R.R.; Fisher, E.M.C.; Cunningham, T.J. Humanising the mouse genome piece by piece. Nat. Commun. 2019, 10, 1845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Zhang, F.; Cheng, D.; Wang, S.; Zhu, J. Crispr/Cas9-mediated cleavages facilitate homologous recombination during genetic engineering of a large chromosomal region. Biotechnol. Bioeng. 2020, 117, 2816–2826. [Google Scholar] [CrossRef] [PubMed]
  185. Yoshimi, K.; Kunihiro, Y.; Kaneko, T.; Nagahora, H.; Voigt, B.; Mashimo, T. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat. Commun. 2016, 7, 10431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Rosshart, S.P.; Herz, J.; Vassallo, B.G.; Hunter, A.; Wall, M.K.; Badger, J.H.; McCulloch, J.A.; Anastasakis, D.G.; Sarshad, A.A.; Leonardi, I.; et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 2019, 365. [Google Scholar] [CrossRef] [PubMed]
  187. Murillo-Cuesta, S.; Artuch, R.; Asensio, F.; de la Villa, P.; Dierssen, M.; Enríquez, J.A.; Fillat, C.; Fourcade, S.; Ibáñez, B.; Montoliu, L.; et al. The Value of Mouse Models of Rare Diseases: A Spanish Experience. Front. Genet. 2020, 11, 583932. [Google Scholar] [CrossRef]
Table 1. Finnish disease heritage diseases and the affected genes with the major Finnish mutation.
Table 1. Finnish disease heritage diseases and the affected genes with the major Finnish mutation.
DiseaseGeneMajor MutationMouse Model Recapitulating Disease
Aspartylglucosaminuria (AGU)Aspartylglucosaminidase (AGA)c.488G > C p.C163SAga KO +
Autoimmune polyendocrinopathy -candidiasis-ectodermal dystrophy (APECED)Autoimmune regulator (AIRE)c.769C > T p.R257XAire KO +/−
Cartilage hair hypoplasia (CHH)RNA component of mitochondrial RNA processing endoribonuclease (RMRP)n.71A > GRmrp KO
Choroideremia (CHM)Rab escort protein 1 (REP1)c.1639 + 2insTRep1 cKO +
Lactase deficiency, congenital (CLD)Lactase (LCT)c.4170T > A p.Y1390XN/A
Ceroid lipofuscinosis, neuronal, 1 (CLN1)Palmitoyl-protein thioesterase 1 (PPT1)c.364A > T p.R122WPpt1 KO +
Ceroid lipofuscinosis, neuronal, 3 (CLN3)CLN3, battenin (CLN3)g.462-677del p.G154Afs*29Cln3 KO +/−
Cln3(Δex7/8) KI +/
Ceroid lipofuscinosis, neuronal, 5 (CLN5)CLN5-intracellular trafficking protein (CLN5)c.1175_1176delAT p.Tyr392*Cln5 KO+/−
Cornea plana 2 (Cornea plana congenital, CNA2)Keratocan (KERA)c.740A > G p.N247SKera KO+/−
Finnish congenital nephrosis (CNF)Nephrin (NPHS1)c.121_122delCT p.R1109XNphs1 KO +
Cohen syndrome (COH1)Vacuolar protein sorting 13 homolog B (VPS13B)c.3348_3349delCT p.C1117fsN/A
Diarrhea, secretory chloride, congenital (DIAR1)Solute carrier family 26, member 3 (SLC26A3)c.-26 + 2T > C p.V317delSlc26a3 KO +/−
Diastrophic dysplasia (DTD)Solute carrier family 26 member 2 (SLC26A2)c.-26 + 2T > CSlc26a2 KI +
Epilepsy, progressive myoclonic, 1 (EPM1)Cystatin B (CSTB)12 nucleotide expansion in promoterCstb KO +
Epilepsy, progressive, with mental retardation (EPMR)CLN8 transmembrane ER and ERGIC protein (CLN8)c.70C > G p.R24GCln8mnd+/−
Amyloidosis, Finnish type (FAF)Gelsolin (GSN)c.654G > A p.D187NhGSN Tg +/−
Glycine encephalopathy (GCE)Glycine decarboxylase (GLDC)c.1691G > T p.S564IGldc KO +
Gracile syndrome (GRACILE)BCS1 homolog, ubiquinol-cytochrome c reductase complex chaperone (BCS1L)c.232A > G p.S78GBcs1l KI+
Hydrolethalus syndrome 1 (HLS1)HYLS1 centriolar and ciliogenesis associated (HYLS1)c.1416A > G p.D211GN/A
Hyperornithinemia with gyrate atrophy of choroid and retina (HOGA)Ornithine aminotransferase (OAT)c.1205T > C p.L402POat KO +/−
Imerslund-Grasbeck syndrome 1 (IGS1)Cubilin (CUBN)c.3891G > A p.P1297LCubn KO
Infantile onset spinocerebellar ataxia (IOSCA)C10ORF2-chromosome 10 open reading frame 2 (C10ORF2)c.1708A > G p.Y508CC10orf2 KI +
Lethal arthrogryposis with anterior horn cell disease (LAAHD)RNA transport mediator (GLE1)c.432-10A > G p.T144_E145insPFQN/A
Compound heterozygote
Lethal congenital contracture syndrome 1 (LCCS1)RNA transport mediator (GLE1)c.432-10A > G p.T144_E145insPFQN/A
Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency *Hydroxyacyl-coenzyme A dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA)c.1528G > C p.E510QHadha KO +/−
Lysinuric protein intolerance (LPI)Solute carrier family 7 member 7 (SLC7A7)c.895-2A > T p.T299IfsX128Slc7a7 KO1 + Slc7a7 KO2
Muscular dystrophy-dystroglycanopathy (congenital with brain and eye anomalies), type a, 3 (MDDGA3)Protein O-linked mannose N-acetylglucosaminyltransferase 1 (beta 1,2-) (POMGNT1)c.1539+1G > A p.L472_H513delPomgnt1 KO1 + Pomgnt1 KO2 +/
Meckel syndrome type 1 (MKS1)MKS transition zone complex subunit 1 (MKS1)c.1408-7_35del p.G470fsMks1 KO +
Mulibrey nanism (MUL)Tripartite motif containing 37 (TRIM37)c.493-2A > G p.R166fsTrim37 KO +
Ovarian dysgenesis 1 (ODG1)Follicle stimulating hormone receptor (FSHR)c.566C > T p.A189VFshr KO +
Progressive encephalopathy with edema, hypsarrhythmia and optic atrophy (PEHO)Zinc finger HIT-type containing 3 (ZNHIT3)c.92C > T p.S31LN/A
Polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy 1 (PLOSL1)Transmembrane immune signaling adaptor TYROBP (TYROBP)Ex1-4del: 5,3 kb deletionTyrobp KO +
RAPADILINO syndromeRecQ like helicase 4 (RECQL4)c.1390+2delTRecql4 KO1-3+/−
Retinoschisis (RS1)Retinoschisin 1 (RS1)c.214G > A p.E72KRs1 KO +/−
Salla disease (SD)Solute carrier family 17 member 5 (SLC17A5)c.115C > T p.R39CSlc17a5 KO +/−
Tibial muscular dystrophy (TMD)Titin (TTN)11-bp change in the last exonTtn cKO +/−
Glu → Val
Val → Lys
Thr → Glu
Trp → Lys
Usher syndrome, type III (USH3)Clarin (CLRN1)c.528T > G p.Tyr176TerClrn1 KO +/−
* Currently under consideration for FDH. c. = coding DNA variant; cKO = conditional knockout; KI = knock-in; KO = knockout; n. = non-coding DNA variant; p. = protein variant; + = mouse model(s) exists and recapitulates the majority of the human disease symptoms; +/− = mouse model(s) exists and recapitulates some of the disease symptoms; N/A = mouse model does not exist or recapitulate disease symptoms.
Table 2. Description of characteristic clinical features of FDH diseases and respective studies in possible knockout (KO), transgenic (TG) or knock-in (KI) mouse model(s).
Table 2. Description of characteristic clinical features of FDH diseases and respective studies in possible knockout (KO), transgenic (TG) or knock-in (KI) mouse model(s).
DiseaseOMIMDisease ManifestationMouse ModelModel Utility
Aspartyl-
glucosaminuria (AGU)
208,400Lysosomal storage disease with infantile growth spurt, progressive mental retardation in
children, abnormalities in the
central nervous system and skeleton and connective tissue lesions
AgaKO: recapitulates well
lysosomal storage disease, but manifests symptoms only
in aged animals [24,25]
Enzyme replacement and gene therapies successfully used in KO model [26,27,28]
Autoimmune
polyendocrinopathy,
candidiasis and
ectodermal dystrophy (APECED)
240,300Multi-symptomatic
endocrinopathy with fungal
infections and ectodermal changes
Aire KO: recapitulates
autoimmune symptoms of APECED [29,30]
Cartilage hair
hypoplasia
(CHH)
250,250Metaphyseal chondrodysplasia, short stature, sparse hair, immune deficiency, gastrointestinal
dysfunctions, anemia,
increased risk for lymphoma and impaired spermatogenesis
RmrpKO: embryonic lethal [31]
Choroideremia (CHM)303,100X-linked progressive degeneration of the retinal pigment
epithelium, photoreceptors
and choroid leading to vision
loss of affected males
Rep1 cKO: conditional knockouts showed the early onset and progressive retinal degeneration, patchy depigmentation of the retinal pigment epithelium and Rab prenylation defects, leading to premature accumulation of deposits in retinal pigment epithelium [32,33]
Lactase deficiency, congenital (CLD)223,000Infantile-onset severe diarrhea and failure to thriveN/A
Ceroid
lipofuscinosis,
neuronal, 1
(CLN1)
256,730Infantile-onset, lethal neurodegenerative disease leading
to psychomotor deterioration,
muscular hypotonia, ataxia,
myoclonia, microcephaly,
progressive epilepsy and visual impairment causing blindness
Ppt1KO: (exon 9 or exon 4): similar CLN1-like phenotypes with blindness, seizures and
myoclonic jerks; progressive
motor difficulties leading
to hind limb paralysis
and death [34,35,36,37,38,39]
Gene therapy testing conducted using the Ppt1 KO mice [24,38,40]
Ceroid
lipofuscinosis,
neuronal, 3
(CLN3)
204,200Fatal neurodegenerative disorder with childhood-onset vision impairment, intellectual disability, movement problems, speech
difficulties and seizures,
which worsen over time
Cln3KO: neuronal storage
disorder and other
neuropathologies [41]
Cln3(Δex7/8) KI: degenerative changes in retina, cerebral
cortex and cerebellum;
neurological deficits and
premature death [40,42]
Although both mouse models recapitulate the aspects of CLN3, they either also show non-neuronal or genetic background-dependent phenotypes, thus not being good models for interventional studies [40,43]
Ceroid
lipofuscinosis,
neuronal, 5
(CLN5)
256,731Childhood-onset developmental regression, myoclonic epilepsy, ataxia, vision loss, speech
problems and a decline in
intellectual function with
varied life expectancy
Cln5 KO: progressive pathology
of the brain mimics the CLN5
symptoms, and Cln5 deficiency leads to microglial activation,
defective myelination and changes in lipid metabolism [44,45]
Cornea plana 2
(cornea plana
congenital)
(CNA2)
217,300Congenital visual impairment,
reduced curvature and hazy
limbus of the cornea, opacities
in the corneal stroma and
marked corneal arcus at early age
Kera KO: structural alterations
recapitulate disease phenotype,
but corneal transparency
is normal [46]
Finnish congenital
nephrosis
(CNF)
256,300Prenatal onset of massive
proteinuria, severe steroid-
resistant nephrotic syndrome
at birth and rapid progression
to end-stage renal failure
Nphs1KO: severe proteinuria
associated with kidney defects
and leading to postnatal
lethality [47,48]
Cohen syndrome (COH1)216,550Non-progressive psychomotor
retardation and microcephaly, characteristic facial features,
retinal dystrophy, cardiac
dysfunction, hyperlaxity of joints and intermittent neutropenia
Vsp13b KO (IMPC)
Diarrhea, secretory chloride, congenital (DIAR1)214,700Fetal-onset watery diarrhea,
polyhydramnion and chronic
diarrhea due to chloride
absorption defect
Slc26a3KO: inpenetrant postnatal lethality, and survivors suffer from growth retardation and acidic
chloridorrhea [49]
Diastrophic
dysplasia (DTD)
222,600Chondrodysplasia causing
severe growth retardation
and structural and functional
abnormalities of joints
Slc26a2KI (hypomorph):
recapitulates essential aspects of DTD such as growth retardation, skeletal dysplasia and joint
contractures [50]
Therapeutic approaches to improve skeletal deformity and short stature in DTD successfully tested using Slc26a2 KI mice [51]
Epilepsy, progressive myoclonic, 1 (EPM1)254,800Childhood- or juvenile-onset
progressive myoclonic
epilepsy with variable severity
CstbKO: phenocopies
progressive ataxia
and myoclonic seizures [52,53]
Epilepsy, progressive, with mental
retardation
(EPMR)
610,003A neurodegenerative, lysosomal storage disease characterized by childhood-onset epilepsy and progressive mental retardation Cln8mnd (267–268insC; frameshift, predicted truncated protein): early onset retinal degeneration and adult-onset hindlimb weakness and ataxia, progressing to spastic paralysis of all limbs and death by 9–14 months; accumulation of intracytoplasmic and lipopigment immunoreactive to ATP synthase subunit c [54,55,56,57] Cln8 KO (IMPC)Gene therapy testing conducted using Cln8mnd mice [58]
Amyloidosis,
Finnish type
(FAF)
105,120Amyloidogenic disease
characterized by lattice corneal dystrophy, cranial neuropathy, bulbar signs, and
dermatologic changes.
Peripheral neuropathy and
renal failure are less common
symptoms
hGSNTg: transgenic line expressing human D187N gelsolin modeling the pathogenic endoproteolytic cascade that leads to gelsolin amyloidogenic peptides and accumulation with amyloidogenesis is
restricted to muscle tissue [59]
Mouse model was used to test D187N gelsolin-targeting nanobodies with positive results [60]
Glycine
encephalopathy (GCE)
605,899Accumulation of glycine in
neonates. Disease varies from
attenuated to fatal form and
presents with lethargy,
hypotonia, myoclonic jerks
and apneas
GldcKO: neonatal disease
features with increased
glycine levels, premature
lethality and hydrocephalus,
in addition to neural tube
defects [61]
Abnormalities of folate metabolism and hydrocephalus were prevented by maternal supplementation of carbon donor to normalize folate
cycle [62,63]
Gracile syndrome (GRACILE)603,358A mitochondrial disease characterized by severe growth retardation, lactic acidosis, nonspecific amino aciduria, cholestasis and abnormalities in iron metabolism, resulting neonatal or early infancy lethalityBcs1l KI: similar phenotype to
human diseases such as growth
restriction (>4 wk), progressive liver disease, renal tubulopathy and premature death (<6 wk) [64,65]
Hydrolethalus
syndrome 1
(HLS1)
236,680A lethal condition of fetus with
hydramnion and multiple
developmental anomalies, including central nervous system malformation, micrognathia, polydactyly, congenital heart defects and abnormal lung lobuli
Hyls1 KO (IMPC)
Hyperornithinemia with gyrate atrophy
of the choroid and
retina
(HOGA)
258,870Hyperornithinemia presumably due to OAT deficiency; triad of progressive chorioretinal
degeneration, early cataract
formation and type II muscle
fiber atrophy; progressive
vision loss
OatKO: neonatal
hypoornithinemia and lethality rescuable by short-term arginine supplementation; postweaning
hyperornithinemia; retinal degeneration in aged mice recapitulating the HOGA phenotype [66]
Imerslund-Grasbeck syndrome 1
(IGS1)
261,100Infancy- or early childhood-onset proteinuria and megaloblastic anemia due to vitamin B12
(cobalamin, Cbl) deficiency caused by vitamin B12
malabsorption
CubnKO: no disease
recapitulation,
embryonic lethality [67]
Infantile onset spinocerebellar ataxia (IOSCA)271,245Severe progressive neurodegenerative disorder characterized
primarily by hypotonia, ataxia, ophthalmoplegia, hearing
impairment, epilepsy and
sensory axonal neuropathy
C10orf2KI: IOSCA mice
manifest a mitochondrial
epileptic
encephalohepatopathy
replicating the key findings
of IOSCA patients [68,69]
Suitable model for testing metabolic interventions as treatment options for mitochondrial diseases
Lethal arthrogryposis with anterior horn cell disease
(LAAHD)
611,890Prenatal onset of diminished fetal mobility and contractures and postnatal respiratory failure
resulting in perinatal death
Gle1KO (IMPC)
Lethal congenital
contracture
syndrome 1
(LCCS1)
253,310A lethal condition of fetus with lack of movements,
hydrops, micrognathia,
pulmonary hypoplasia and
multiple joint contractures
Gle1KO (IMPC)
Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD)
deficiency *
609,016A mitochondrial disorder of long-chain fatty acid oxidation characterized by infancy- or early childhood-onset hypoglycemia, metabolic acidosis, hypotonia, liver disease, cardiomyopathy and
arrhythmias, as well as a later onset of chronic peripheral neuropathy and pigmentary retinopathy
HadhaKO: embryonic lethality
in homozygotes, hepatic steatosis at a young age (3 mo) and
hepatocellular carcinoma
without cirrhosis at an
older age (>13 mo) in
heterozygotes [70]
Lysinuric protein
intolerance
(LPI)
222,700Inborn error of amino acid
metabolism resulting in growth failure, renal disease,
hyperammonemia,
pulmonary
alveolar proteinosis,
autoimmune disorders
and osteoporosis
Slc7a7KO1: growth restriction and very early embryonic lethality [71]
Slc7a7KO2: deletions do not recapitulate precisely the variants that have been reported in humans; key features of human LPI such as
intrauterine growth restriction and proximal tubular dysfunction
are present [72]
Muscular dystrophy–dystroglycanopathy (congenital with brain and eye anomalies), type A, 3
(MDDGA3)
253,280Brain and eye malformations,
severe, congenital muscular
dystrophy, mental retardation and survival up to
more than 70 years
Pomgnt1KO1: viable mice developmental defects in muscle, eye and brain, similar to the phenotypes observed in humans [73,74] Pomgnt1 KO2: increased postnatal lethality, mild dystrophy with reduction in muscle mass and muscle fibers and impaired muscle regeneration [75]
Meckel syndrome type 1,
(MKS1)
249,000Genetically heterogenous disease with the main features being
central nervous system malformation, polycystic kidneys,
fibrotic changes in the liver,
congenital heart malformation and polydactyly
Mks1KO: 259 amino acid deletion resulted in craniofacial defects,
polydactyly, congenital heart
defects, polycystic kidneys and randomized left-right patterning, quite similar to the human MKS1
phenotype [76]
Mulibrey nanism (MUL)253,250Multi-organ disorder with
prenatal onset growth failure,
cardiomyopathy, characteristic craniofacial features, infertility, insulin resistance with type 2
diabetes and an increased risk for tumors
Trim37KO: recapitulates several features of the multi-organ
human disorder, including
infertility, increased risk for
tumors, fatty liver and
cardiomyopathy [77]
Ovarian
dysgenesis 1
(ODG1)
233,300Hypergonadotropic
hypogonadism with poorly
developed streak ovaries in
females and smaller testes and from low to normal sperm
counts in men
FshrKO: recapitulates human phenotype quite well; females have small ovaries due to a blockage of folliculogenesis, and male mice have smaller testes and reduced sperm counts [78,79,80]
PEHO syndrome260,565Early infancy-onset hypotonia, delayed psychomotor development, infantile spasms, optic
atrophy, progressive atrophy
of the cerebellum and
brainstem, dysmyelination
and profound mental retardation
N/A
Polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy 1 (PLOSL1)221,770Adult-onset disorder of bones and central nervous system,
leading to early dementia
and death
TyrobpKO: recapitulates
skeletal and psychotic
characteristics of
PLOSL1 [81]
Promising drug
therapy testing conducted using Tyrobp KO mice [82]
RAPADILINO
syndrome
266,280Radial and patellar aplasia, cleft or highly arched palate,
diarrhea, dislocated joints,
small size and limb
malformations, long slender
nose, cancer predisposition
and normal intelligence
Recql4 ex 5-8 KO: embryonic
lethality; Recql4 ex 13 KO: neo- and postnatal lethality with growth retardation, skin, hair and bone
defects; Recql4 ex 9-13 KO: palate and limb defects and cancer
predisposition [83,84,85]
Retinoschisis
(RS1)
312,700X-linked childhood-onset
reduced visual acuity due to
retinal dystrophy leading to
retinoschisis (splitting) of the
neural retina in affected men
Rs1KO: not exactly
recapitulating the human
phenotype as disrupted
organization of the retina was
in all cell layers [86]
Successful gene replacement therapy in KO model [87], and dorzolamide treatment improved morphological features in 6 of 7 patients [88]
Salla disease
(SD)
604,369Hypotonia and delayed development in infancy, cerebellar ataxia, progressive cerebellar atrophy and dysmyelination leading to mental retardation; viscero-
megaly and coarse features,
enlarged lysosomes and high amounts of free sialic acid
excreted in the urine
Slc17a5 KO: recapitulates
hypomyelinating and
lysosomal accumulation
phenotype in CNS; in
addition to premature
death, poor coordination
and seizures [89,90]
Tibial muscular
dystrophy
(TMD)
600,334Late adult-onset tibial
muscular dystrophy
TtnKO: embryonic or postnatal death; Ttn muscle-specific KO:
adolescent death [91,92]
Usher syndrome,
type III
(USH3)
276,902Post-lingual,
progressive hearing loss
and loss of central
visual acuity later in life
Clrn1KO: phenocopies
early onset hearing loss,
but not visual
impairment [93,94]
Transgene strategy used for possible therapeutic intervention for Usher syndrome [94]
* Currently under consideration for FDH. KO (IMPC): mouse knockout has been generated by the International Mouse Knockout Consortium, and preliminary screening has been performed by the International Mouse Phenotyping Consortium (https://www.mousephenotype.org, accessed on 25 September 2021), but no scientific publication exists yet. CNS: central nervous system; KO: knockout; N/A: not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zárybnický, T.; Heikkinen, A.; Kangas, S.M.; Karikoski, M.; Martínez-Nieto, G.A.; Salo, M.H.; Uusimaa, J.; Vuolteenaho, R.; Hinttala, R.; Sipilä, P.; et al. Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage. Cells 2021, 10, 3158. https://doi.org/10.3390/cells10113158

AMA Style

Zárybnický T, Heikkinen A, Kangas SM, Karikoski M, Martínez-Nieto GA, Salo MH, Uusimaa J, Vuolteenaho R, Hinttala R, Sipilä P, et al. Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage. Cells. 2021; 10(11):3158. https://doi.org/10.3390/cells10113158

Chicago/Turabian Style

Zárybnický, Tomáš, Anne Heikkinen, Salla M. Kangas, Marika Karikoski, Guillermo Antonio Martínez-Nieto, Miia H. Salo, Johanna Uusimaa, Reetta Vuolteenaho, Reetta Hinttala, Petra Sipilä, and et al. 2021. "Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage" Cells 10, no. 11: 3158. https://doi.org/10.3390/cells10113158

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop