Skip to main content

The double whammy of ER-retention and dominant-negative effects in numerous autosomal dominant diseases: significance in disease mechanisms and therapy

Abstract

The endoplasmic reticulum (ER) employs stringent quality control mechanisms to ensure the integrity of protein folding, allowing only properly folded, processed and assembled proteins to exit the ER and reach their functional destinations. Mutant proteins unable to attain their correct tertiary conformation or form complexes with their partners are retained in the ER and subsequently degraded through ER-associated protein degradation (ERAD) and associated mechanisms. ER retention contributes to a spectrum of monogenic diseases with diverse modes of inheritance and molecular mechanisms. In autosomal dominant diseases, when mutant proteins get retained in the ER, they can interact with their wild-type counterparts. This interaction may lead to the formation of mixed dimers or aberrant complexes, disrupting their normal trafficking and function in a dominant-negative manner. The combination of ER retention and dominant-negative effects has been frequently documented to cause a significant loss of functional proteins, thereby exacerbating disease severity. This review aims to examine existing literature and provide insights into the impact of dominant-negative effects exerted by mutant proteins retained in the ER in a range of autosomal dominant diseases including skeletal and connective tissue disorders, vascular disorders, neurological disorders, eye disorders and serpinopathies. Most crucially, we aim to emphasize the importance of this area of research, offering substantial potential for understanding the factors influencing phenotypic variability associated with genetic variants. Furthermore, we highlight current and prospective therapeutic approaches targeted at ameliorating the effects of mutations exhibiting dominant-negative effects. These approaches encompass experimental studies exploring treatments and their translation into clinical practice.

Background

The exploration of molecular and cellular mechanisms underlying many genetic diseases typically starts by evaluating the early stages of the protein biogenesis as well as its subsequent processes including trafficking, interactions, the execution of its biological function and even its disposal. A detailed understanding of defects and aberrations in these processes provides key insights into the molecular foundation of the pathogenesis of genetic diseases and the consequent manifestations of the pathological phenotypes. In eukaryotic cells, secretory and endomembrane proteins destined for many cellular organelles typically enter the endoplasmic reticulum (ER) in their unfolded states, where they undergo their initial and crucial processes to acquire their proper tertiary conformations [1, 2]. In particular, this is where these ER-targeted proteins undergo essential post-translational modifications including glycosylation, proline isomerization, lipidation and disulfide bond formation, which are often crucial for guiding proper folding, stability and the performance of their biological functions [3, 4]. To ensure efficiency and fidelity, cells have adapted extensive ER quality control (ERQC) mechanisms that allow only properly folded proteins to reach their functional destination [5]. Due to the extensive and rigorous cellular mechanisms dedicated to maintaining protein fidelity and proper conformation, it is estimated that 12–15% of newly synthesized proteins do not successfully attain their intended conformation, leading to their subsequent elimination via one or more of the cellular degradation pathways within the secretory pathway [6, 7]. This percentage is significantly increased when proteins harbor mutations that lead to their mis- or mal-folding, and often the removal of the mutant protein quantitatively [8]. Disease-causing mutations, including point mutations, insertions, deletions and repeat expansions impact protein structure and function in diverse ways, leading to three primary disease cellular mechanisms: loss-of-function, gain-of-function or dominant-negative effects [9]. Loss-of-function mutations may cause decreased or total loss of protein function that consequently leads to failure or reduction in performing its normal physiological function [10]. Conversely, gain-of-function mutations occur when the mutant protein acquires a new or abnormal function such as increased or uncontrolled activities leading to dysregulation in the normal cellular activities [11]. In autosomal dominant diseases where one allele expresses the mutant protein, while the other allele preserves its WT expression, a range of disease mechanisms can be demonstrated including loss-of-function of one allele (haploinsufficiency), gain-of-function, dominant-negative effects or a combination of two mechanisms [11]. The dominant-negative effects exerted by the mutant protein on the WT protein may exacerbate the haploinsufficiency state in autosomal dominant diseases [12]. This occurs through interference with the function or trafficking of the normally functioning protein. It is common for proteins to function as homodimers, oligomers or part of multi-subunit complexes. As a consequence, when a mutant protein is expressed from the mutant allele, this can lead to the formation of abnormal dimers, heteromers or multi-subunit complexes that often negatively impact the function or stability of their functioning unit. Numerous studies, including our own, indicate that ER-targeted mutant proteins that are unable to attain their correct conformation, often experience defective trafficking, leading to their entrapment in the ER and subsequent degradation via the ER-associated protein degradation (ERAD) and other associated mechanisms [13,14,15,16,17,18,19,20]. Therefore, exploring whether certain ER-retained mutant proteins exert dominant-negative effects on their WT counterparts, or their complexes has become crucial. Such effects may exacerbate the disease pathological state, potentially explaining some of the broad and variable spectrum of phenotypic expressivity observed for many monogenic diseases [12]. Despite extensive research efforts directed towards comprehending loss of function and gain of function mutations in dominant conditions, there has been relatively limited exploration into the involvement of dominant-negative effects mechanisms. ERAD is a complex mechanism that plays a crucial role in protein quality control in the secretory pathway, involving the recognition of mutant, orphaned and misfolded proteins then targeting them for re-translocation for degradation by the proteasomal or lysosomal machineries. This mechanism has been implicated in the pathology of numerus human genetic condition and the number is expected to keep rising due to the central importance of this quality control mechanism in the monitoring almost a third of the cellular proteins [21,22,23].

In this manuscript, we aim to review the literature and include our perspectives on the impact of various dominant-negative effects implicated in the pathology of a spectrum of autosomal dominant diseases caused by mutations in secretory and membrane proteins (Table 1). We will focus on both the currently acknowledged and potential dominant-negative effects displayed by mutant variants retained in the ER as a result of the ER quality control (ERQC) mechanisms. In addition, we will also highlight existing and potential therapeutic interventions aimed at mitigating the impact of mutations exhibiting dominant-negative effects, spanning from experimental research on therapies to their application in patient care.

Table 1 Autosomal dominant monogenic diseases caused by mutant secretory and membranal proteins

The dominant-negative effects: Concept and mechanisms

Dominant-negative mutations have been defined in 1987 by Ira Herskovitz as “those leading to mutant polypeptides that disrupt the activity of the WT gene when overexpressed” [83, 84]. This current review is focused on the combinational mechanism of ER retention and dominant-negative effects exerted by the mutant proteins on their WT counterpart, resulting in the entrapment and defective trafficking of WT proteins from the ER to their functional destinations. `It is crucial, however, to recognize that dominant-negative mechanisms extend far beyond those caused by ER-retained mutants. Here is an overview of some of these mechanisms:

  1. 1.

    Altered protein trafficking: Mutant proteins might alter the trafficking of the WT proteins causing their entrapment in cellular compartments, thus preventing their trafficking to their functional destination. For example, some ER-retained endoglin mutants associated with hereditary haemorrhagic telangiectasia type 1(HHT1) form heterodimers with WT endoglin in the ER and thus preventing its trafficking to the plasma membrane [47]

  2. 2.

    The formation of inactive protein complexes: Trafficking-competent mutant proteins can form heterodimers with their WT counterpart expressed by the unaffected allele or with other WT partner proteins expressed by different genes, which renders the heterodimeric complex inactive [85]. This type of dominant-negative effect is best represented in collagen disorders such as Osteogenesis Imperfecta (OI), in which a mutant collagen protein forms inactive complexes with its WT partners negatively impacting the function of the whole collagen matrix [39].

  3. 3.

    Competitive binding inhibition: In this case, the mutant proteins compete with the WT counterparts for binding to shared substrates or ligands, thereby limiting proper binding interactions, leading to a potential inhibitory effect. For example, Von Willebrand disease (VWD) is a bleeding disease caused by mutants von Willebrand factor (VWF), a glycoprotein expressed by endothelial cells [86]. Mutants VWF interfere with the binding of WT protein to platelets and sub-endothelium in a dominant-negative manner, resulting in reduced clotting function and increased risk of bleeding [87].

  4. 4.

    Protein destabilizing effect: The dominant-negative effect exerted by the mutant protein results in WT reduced stability or even premature degradation. Certain p53 tumor suppressor mutant proteins adversely affect the stability of WT p53, leading to its reduced function and effectiveness [88].

  5. 5.

    Conformational effects: Secreted misfolded mutant proteins with structural defects may give rise to conformational defects in the WT protein complex, impeding its physiological function. For example, mutant fibrillin-1 proteins encoded by FBN1 gene, associated with Marfan disease exhibit a dominant-negative effect on the WT protein via disruption of the normal assembly of the extracellular matrix [89].

Protein quality control mechanisms in the early secretory pathway: Components and involvement in human diseases

Newly synthesized secretory and membrane proteins are transported to the ER where they undergo various interactions, posttranslational modifications and assembly to generate mature proteins in fully folded three-dimensional states. While protein folding can occur co- or posttranslationally, its accuracy and efficiency are important factors for the protein’s functionality and cellular homeostasis. Protein folding in the ER is mediated via a complex ER-resident chaperoning and folding machineries, consisting of the heat shock protein (Hsp) chaperones and the calnexin/calreticulin (CNX/CRT) lectin-like chaperones [90]. Among the major molecular chaperones involved in preventing protein aggregations and incorrect folding is the Hsp70 chaperone; BiP (Fig. 1) [91, 92]. Dysfunction in the chaperoning machinery contributes to various diseases, including Alzheimer's and Parkinson diseases [93].

Fig. 1
figure 1

Protein synthesis and quality control mechanisms in the ER. Newly synthesized proteins in the ER undergo quality control checks to ensure correct folding. Misfolded proteins form aggregates in the ER lumen and activate the UPR response that leads to transcriptional activation of various ER stress genes and are degraded via the proteasomal ERAD machinery based on their lesioned domain

In addition to the failure of the chaperoning systems, several other factors contribute to defective protein folding in the ER, including transcriptional or translational errors, abnormal chemical protein modifications, oxidative stress, and genetic mutations [18, 94]. Consequently, incorrect protein folding results in defective trafficking of the proteins to their destination associated with ER retention and the formation of protein aggregates in the ER lumen. Accumulation of misfolded proteins in the ER triggers an ER stress signaling cascade known as the unfolded protein response (UPR). The UPR employs cytoprotective strategies aimed at preserving ER homeostasis, thereby alleviating ER stress caused by the burden of misfolded proteins. This is accomplished by activating downstream ER quality control activities. The UPR cascade is induced by three established arms: protein kinase RNA like endoplasmic reticulum kinase (PERK), inositol requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6) (Fig. 1) [20, 95]. The coordinated activation of the three UPR arms collectively targets the attenuation of protein synthesis within the ER, induction of molecular chaperones’ gene expression aiding in protein folding, and ultimately the removal of misfolded proteins via ERAD mechanism [96]. ER stress and the disruption of the UPR have been linked to the development of various diseases, spanning neurodegenerative conditions like Alzheimer's and Parkinson's diseases [97], metabolic disorders such as diabetes and obesity [98,99,100], inflammatory diseases [101], cancer [102, 103], and rare genetic disorders like cystic fibrosis (CF) [104], Gaucher disease [105, 106], Acromesomelic Dysplasia 1, Maroteaux type [18] and many others [107,108,109,110]. Understanding the mechanisms underlying ER stress and the UPR activation is crucial for elucidating disease pathogenesis and developing therapeutic strategies. CF has been recognized as a pioneering disease in the field of ER stress research due to its well-defined genetic basis, thoroughly investigated pathophysiology, and the availability of animal models and cell culture systems for studying disease mechanisms [111]. CF is an autosomal recessive disorder caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene, which encodes a chloride channel primarily found in the apical membrane of epithelial cells [112]. Mutations in CFTR result in defective chloride ion transport across cell membranes, leading to sticky mucus production in the lungs and digestive system. Studies have revealed that most CF disease-causing mutations, including the most common CF variant (F508del), result in defective protein folding and trafficking, leading to ER retention and degradation of misfolded CFTR protein [113,114,115,116,117]. This accumulation of misfolded CFTR protein in the ER triggers ER stress and activates the the UPR mechanism aimed at restoring ER homeostasis [118].

In eukaryotic cells, nascently synthesized proteins are subjected to unique quality control assessments. This step involves the UDP-glucose: glycoprotein glucosyltransferase (UGGT) interaction with partially or misfolded proteins providing a further folding attempt mediated by the CNX/CRT chaperones cycle [119]. Proteins failing to meet the stringent quality checks are destined for degradation via various pathways including the ERAD machinery. Misfolded proteins are recognized and processed by distinct ERAD sub-pathways: ERAD-L, ERAD-M, and ERAD-C, based on the site of the defective domain within the protein—luminal (ERAD-L), membrane-bound (ERAD-M), or cytosolic (ERAD-C) (Fig. 1) [120]. ERAD substrates are exported and tagged with ubiquitin that serves as a degradation signal. These proteins are retrotranslocated to the cytosol for degradation by the large protein complex, the proteasome or the lysosomes, via a pathway known as the ER-to-lysosome associated degradation (ERLAD) [121]. Moreover, some ER retained mutants might escape the ERQC mechanism and avoid being degraded. These mutants might form aggregates in the ER lumen or interact in a dominant-negative manner with their WT counterparts and possibly affect their normal physiological function. These mechanisms and indeed defects in their components have been implicated in numerous human conditions, which is the focus of this review.

ER-retained mutant proteins exhibit dominant-negative effects in a range of autosomal dominant disorders

As indicated earlier, the entrapment of secretory proteins may result in a spectrum of monogenic diseases with varied modes of inheritance and molecular mechanisms. These encompass a range from loss of function, gain of function, dominant-negative effects, or combinations thereof. Here, we emphasize the dominant-negative effects demonstrated or predicted to play significant roles in the pathology of numerous autosomal dominant diseases. In these conditions, both the WT and mutant variants are expressed and anticipated to interact during the initial stages of the protein biogenesis and assembly within the ER, particularly in cases where the protein functions as a dimer, oligomer or part of a multi-subunit complex (Fig. 2). The broad spectrum of phenotypic severity observed across various autosomal dominant diseases constitutes a significant area of ambiguity and concern in biomedical research. The exploration of mechanisms, or combinations thereof, that cause phenotypic heterogeneity among affected individuals is essential in understanding the complex genotype–phenotype interplay. Extensive literature highlights examples of dominant-negative effects instigated by mutant variants, wherein these mutants impair the function of the WT counterpart at the functional location. In this review, we specifically explore the dominant-negative effects exerted by ER-retained mutants on the WT proteins, thereby initiating a double effect that entails dominant-negative effects on top of haploinsufficiency leading to the entrapment and possibly premature degradation of WT protein. Consequently, this combinatorial mechanism causes an excessive loss of protein function, thereby exacerbating the severity of disease phenotypes for some mutants exhibiting ER retention. It is important to note that our aim in this review is not to exhaustively cover every condition exhibiting dominant-negative effects within the field, but rather to highlight the widespread involvement of these mechanisms in disease pathogenesis in some autosomal dominant conditions.

Fig. 2
figure 2

Dominant-negative effects exerted by ER-retained mutant on the WT protein expressed by the functional allele. Monomeric Plasma membrane proteins or secretory proteins that fail to attain their normal conformation get targeted by the ERAD machinery for proteasomal degradation. In the case of dimeric or oligomeric proteins, ER-retained mutants are likely to interfere with the WT counterpart via a formation of hetero dimers/oligomers complexes. This interreference causes the entrapment of the WT within these complexes impeding its normal trafficking and leading to premature degradation through the ERAD mechanism

Skeletal and connective tissue disorders

Marfan Syndrome (MFS, MIM #154700) is an autosomal dominant genetic disease caused by heterozygous mutations in the FBN1 gene that encodes Fibrillin-1, a crucial protein component in the extracellular matrix (ECM) [122]. The disease is broadly classified as a connective tissue disorder with clinical manifestations impacting various organs including the heart, the blood vessels and the eyes [123]. The most distinctives features include a long face, high-arched palate, elongated limbs, tall and slender physique, chest deformities, lens dislocation, aortic root dilation and potential aneurysms [122]. MFS is characterized by a wide spectrum of phenotypic variabilities among affected individuals, including those carrying different genetic variants or even those with the same variant [123]. Fibrillin family of proteins encompasses three main types of fibrillin, fibrillin-1, 2 and 3. Fibrillin-2 and 3 are predominantly expressed during development, whereas fbrillin-1 is expressed throughout adulthood, as it provides strength and elasticity to connective tissues in major organs [124]. Fibrillin-1 is a 250 kDa glycoprotein characterized by multi-modular organization and is considered as the major structural component of the connective tissues microfibrils [24]. However, the mechanism by which fibrillin-1 assembles into microfibrils, remains to be fully elucidated. The majority of disease-causing variants in FBN-1 involve the substitution of cysteine amino acids, which play a critical role in forming disulphide bridges and maintaining proper protein conformation. For instance, severely misfolded variants like C1117Y and C1129Y, where cysteine is replaced by tyrosine, exhibit defective trafficking and become trapped and accumulate in the ER [24]. Conversely, the disease-causing variant G1127S is secreted normally, similar to the WT. These conclusions were drawn by examining their glycosylation profiles. Variants C1117Y and C1129Y exhibit a simple N-linked glycosylation pattern characteristic of ER acquisition. On the other hand, they lack the complex N-glycosylation typical of Golgi apparatus processing, resulting in a lower molecular weight presentation on SDS-PAGE, confirming their retention in the ER. In contrast, variant G1127S appears in a mature, fully glycosylated form, mirroring WT fibrillin-1 [24]. As a result, it was proposed that normally-trafficked fibrillin-1 mutants such as G1127S are likely to exert dominant-negative effects via misincorporation into the normal microfibril, however, no clear evidence was presented. In addition, intracellular dominant-negative was postulated as a probable disease mechanism attributed to the ER-retained variants such as C1117Y and C1129Y in addition to haploinsufficiency. This occurs because ER-retained mutants are prone to interact with WT fibrillin-1, forming heterodimers during the initial stages of protein dimerization within the ER. This interaction hampers the trafficking and secretion of fibrillin-1 to the cell surface [24]. The concept of a dominant-negative effect was previously proposed by Dietz and colleagues, supported by their demonstration that patients expressing the least amount of the nonsense mutant variants of fibrillin-1 exhibited the mildest disease phenotype. Conversely, patients with fully expressed variants exhibited the usual moderate to severe manifestation of the diseases [25].

Recent advances in high-throughput genomic analysis have revealed that idiopathic short stature (ISS, MIM # 300582) in children is linked to various genes that regulate growth plate function, including heterozygous mutations in NPR2 [29]. NPR-B encoded by NPR2 functions as a homodimer that catalyzes the conversion of GTP to cGMP upon binding of its ligand, C-type natriuretic peptide (CNP) [30]. Recently, co-immunoprecipitation assays have revealed that diseases-causing heterozygous missense NPR-B variants (R110C, R495C and Y598N) identified in ISS subjects exhibit dominant-negative effects on the WT receptor [29, 31]. These studies have demonstrated that intracellular cGMP levels significantly increase upon cell transfection with the WT NPR-B. On the other hand, a significant decrease is observed when a mutant variant is co-expressed with the WT receptor. Notably, a prior investigation has also identified variant R110C as an ISS-causing mutation. This study demonstrated that this particular variant displays defective trafficking from the ER to the Golgi apparatus, evidenced by an immature glycosylation profile characteristic of ER-retained mutants [28]. Conversely, variant Q417E, also identified in the same study, trafficked normally to the plasma membrane. Interestingly, both variants exhibited dominant-negative effects, as evidenced by a decrease in cGMP production capacities. Specifically, R110C showed a negligible cGMP response, while Q417E displayed a significantly reduced response. These findings underscore the importance of further investigating the implications of dominant-negative effects and ER retention across a broader spectrum of variants. This approach will enable researchers to gain a more comprehensive understanding of the molecular pathology associated with variable variants.

Autosomal dominant Limb-girdle Muscular Dystrophy (LGMD-1CI, MIM # 609115) is a specific subtype of LGMD that is associated with mutations in CAV3 gene. The disorder is characterized by progressive muscle weakness primarily affecting the shoulders and hips muscles [125]. Caveolin-3 encoded by CAV3 is a member of the caveolin integral membrane proteins and a key structural protein of caveolar membrane in muscle cells [126]. Up to date ten LGMD-1C-disease causing variants of CAV3 have been identified according to the Human Genome Mutation Database (HGMD) (https://www.hgmd.cf.ac.uk/ac/index.php). Amongst the most studied variants are P104L, ΔTFT/63–65 (deletion of amino acids 63 to 65) and A45T. Minetti, Sotgia, Lisanti, and colleagues were the first to report the disease causing variants P104L, ΔTFT/63–65 in LGMD-1C patients [127]. In vitro characterization and immunofluorescence staining experiments have shown that the two variants fail to reach the plasma membrane. Instead, they are retained at the Golgi complex level and subsequently degraded through proteasomal degradation, along with partial degradation of the WT protein [128]. These findings suggest a dominant-negative effect of these two variants on the WT protein, leading to its entrapment in mixed WT/mutant oligomers, which ultimately results in proteasomal degradation, as evidenced by ER-localization. The same research group later demonstrated that treatment with the proteasomal inhibitor (MG-132) significantly enhanced the trafficking of WT protein entrapped at the Golgi complex to the plasma membrane in cells expressing both the WT and mutant variants [34]. Similarly Herrmann and colleagues have demonstrated that disease-causing variant A45T exhibit defective trafficking and also prevent the normal localization of WT caveolin-3 in a dominant-negative manner [129].

Collagen-related mutations in connective tissue disorders

The intricacy of collagen structures and their assembly plays a pivotal role in the diverse phenotypic manifestations observed in individuals with mutant collagen genes. Collagen, being the most abundant protein in the human body and a fundamental component of connective tissues, contributes significantly to the structural integrity of various tissues in the body such as skin, cartilage, and blood vessels [130]. The variability in collagen types and their interactions within heterotypic fibrils adds a layer of complexity to the assembly process. Mutations in collagen genes can disrupt this intricate network, leading to a spectrum of phenotypes broadly known as collagenopathies or connective tissue disorders [131].

A collagen molecule is most characterized by its triple-helical α-domain, which constitutes up to 95% of the molecule in some classes of collagen (reviewed in [132, 133]. Several collagen-related genetic disorders are caused by a dominant inheritance of glycine substitution to a larger amino acid in the triple helical domain of the protein, which structurally affect collagen folding and assembly [131].

Mutations in collagen type VII alpha 1 encoded by the gene COL7A1 cause dystrophic epidermolysis bullosa (DEB, MIM # 131750), a genetic disorder characterized by skin blistering in response to minor trauma or friction [134]. It has long been established that certain disease-causing variants involving glycine substitutions accumulate intracellularly in the endoplasmic reticulum (ER) and fail to undergo proper extracellular secretion [135]. This failure to secrete results in a haploinsufficiency state, contributing to the manifestation of the disease. The same group has later shown that secreted mutant α1(VII) chains exert a dominant-negative effect by interacting with the WT protein forming heterotrimeric triple helix complex, leading to a destabilizing effect on collagen VII structure [131].

Heterozygous mutations in collagen VI genes (COL6A1, COL6A2 and COL6A3) are associated with Bethlem myopathy disorder (BMD, MIM # 158810), which is a milder form of Ullrich congenital muscular dystrophy (UCMD, MIM # 254090) also associated with homozygous mutations in the same genes. Bethlem myopathy is mainly characterized by proximal muscle weakness and joint contracture that progressively affect mobility and flexibility [136]. However, distinction between the two diseases in terms of their mode of inheritance was revised when patients with heterozygous mutations exhibited a severe form of the disease that is typical of UCMD. This finding has given rise to rigorous protein synthesis studies in order to provide an explanation for the consequences of severe mutations in the extremely complex structure of collagen VI [37, 137]. The three alpha chains α1, α2 and α3 that characterize collagen VI fold into triple helical heterotrimeric monomer in the ER, which are then transported to the cell surface via the Golgi apparatus [133]. When these procollagen monomers reach the cell surface, they align in a staggered fashion to form dimers that are bonded via a disulfide bond, then dimers are aligned laterally to form a tetrameric complex in the extracellular matrix (ECM) [138]. It has been demonstrated that large amino acid deletion at the N terminal of the triple helical domain resulted in the secretion of mutant heterotetramers. The tetrameric stoichiometry of collagen VI means that mutations in any of the three alpha chains would result in only 1/16 normal tetramer [37, 137]. This finding unequivocally illustrates the dominant-negative impact exerted by the mutant chain on the overall structural assembly of collagen VI. As a result, the collective effect resembles the complete loss of collagen VI observed in individuals with the homozygous mutation in UCMD. On the other hand, patients carrying heterozygous, in-frame amino acid deletions downstream of the triple-helical domain, which removes cysteines required for dimerization, exhibit a milder form of the disease. This deletion prevents the formation of WT/mutant dimers and consequently reduces the dominant-negative impact on the WT protein [137].

Osteogenesis imperfecta (OI) has served as a classic example for dominant-negative effects of structural proteins [41]. The disease, also known as brittle bone disease, is characterized mainly by bone fragility, short stature, loose joints and other variable skeletal deformities [139]. Classical OI types I to IV are caused by autosomal inherited mutations in COL1A1 or COL1A2 genes that encode α1 and α2 subunits of collagen 1, respectively. Type I collagen is a heterotrimeric complex that consists of two α1 and one α2 chains, synthesized in the ER and assembled via a recognition sequence at the C terminal, along with the formation of disulfide bondings, prior to being transported to the Golgi apparatus [39, 140]. The variability in the phenotype of this disease, coupled with a limited understanding of its underlying mechanisms, has prompted researchers to identify additional collagen-related genes involved in the regulation of collagen metabolism and assembly. Most of these genes have been linked to autosomal recessive pattern of inheritance. Nonetheless, mutations in Collagen1 genes still account for the majority of OI cases [141].

Molecular mechanisms attributed to the manifestation of autosomal dominant OI include decreased transcript due to nonsense mutation, decreased collagen secretion due to ER retention, and disrupted pro-collagen chains assembly and processing [41, 142,143,144]. ER retention of glycine-substitution mutant variants of α1 and α2 subunits of collagen 1 was observed through transmission electron microscopy analysis of OI fibroblasts, revealing the presence of an enlarged ER indicative of ER stress [145]. Apoptotic cellular death was also demonstrated to be triggered despite autophagic activation through the UPR in an attempt to salvage the cells. Severe forms of OI that involve glycine substitution are associated with pathogenic variants that exert a dominant-negative effect, disrupting the assembly of the triple helix and collagen fibril. This disruption results in severe structural damage to the bone matrix [41, 146, 147].

Ehlers-Danlos syndromes (EDS) represent a group of genetically heterogenous conditions that are caused by pathogenic variants in up to 19 genes, mostly encode collagens or collage-related proteins [148]. Classic EDS (cEDS, MIM # 130000) is mainly associated with mutations in COL5A1 or COL5A2 genes encoding α1 and α2 chains of the collagen 5. Patients of this class of EDS present with joint hypermobility, skin hyper-elasticity and a tendency to develop atrophic scars [149]. Despite the limited number of clinically well described cEDS associated with mutations in COL5A2, the majority manifest severe phenotypes and have an impact on the structural integrity of collagen V. These mutations exhibit a dominant-negative effects that disrupt the formation of heterotypic fibrils and the interactions between collagen 5 and other constituents of the extracellular matrix [43, 150].

Pathogenic variants of COL2A1 and COL11A1 genes encoding collagen II α1 and collagen XI α2 chains have been associated with Stickler syndrome type 1 and 2, (ST1, MIM # 108300 and STL2, MIM # 604841), respectively. The α1 chain from COL11A1 combines with the α2 chain from COL11A2 and the α1 chain from COL2A1 to create heterotrimeric type XI collagen [46]. Stickler syndromes are a group of heterogenous connective tissue disorders characterized by distinctive facial feature, ocular abnormalities and joint anomalies. Splice site mutations in COL11A2 are the primary disease-causing mutations reported thus far. However, mutant mRNA does not undergo nonsense-mediated decay (NMD), allowing mutant chains to be expressed and associate with other α chains, leading to the formation of mutant collagen XI trimers in a dominant-negative manner [46, 151]. Conversely, patients with mutations that lead to unexpressed protein due to targeting by the NMD mechanism tend to exhibit a milder form of the disease, and this mechanism is considered haploinsufficiency only [152].

Akawi and colleagues have argued that homozygous misfolding mutations in COL11A are more severe than bi-allelic null mutations as a result of the possible interference of the misfolded COL11A with its other collagen partners, presumably as a result of dominant-negative effects, and hence disrupting the function of the whole complex more severely [153, 154]. On the other hand, pathogenic variants of COL2A1, associated with Stickler syndrome type1, are believed to manifest the disease phenotype through only haploinsufficiency [46]. Several misfolded variants of both types of collagens were reported to be retained in the ER, followed by proteasomal degradation [45]. Nonetheless, to our knowledge, whether ER-retained mutants exert dominant-negative effects by interfering with the WT in heterotrimeric complexes remains largely unexplored.

The preceding discussion highlights how the complexity of collagen structure magnifies the dominant-negative effect of mutant subunits by exacerbating the disruption in the intricate network of collagen assembly. Mutant collagen subunits, with their altered structures, not only compromise the functionality of individual molecules but also introduce a destabilizing influence during fibril formation. These effects ultimately contribute to the diverse phenotypic outcomes observed in collagen-related genetic disorders. It’s important to note that there has been limited research dedicated to investigating the dominant-negative effect exerted by mutant collagen subunits when trapped in the ER, potentially affecting their trafficking. The ER serves as a crucial site for proper folding and post-translational modifications of collagen molecules before they are transported to their functional destinations. The presence of mutant subunits in the ER may disrupt these processes, leading to the accumulation of misfolded or improperly modified collagen. This lack of in-depth exploration into the consequences of such entrapment hinders a comprehensive understanding of how trafficking abnormalities contribute to the overall pathogenesis of collagen-related genetic disorders.

Vascular monogenic disorders

Hereditary haemorrhagic telangiectasia (HHT) is a vascular genetic disorder characterized by vascular dysplasia inherited in an autosomal dominant manner. Its spectrum of phenotypes varies from occasional nasal bleeds to internal organ hemorrhages affecting the gastrointestinal tracts (GI), kidneys, liver, and brain [155].The disease has been classified into four types according to the causative gene: HHT1, HHT2, HHT5 and (JPH) Juvenile polyposis and HHT, associated with mutations in ENG, ACVLR1, GDF2 and SMAD4 genes, respectively [156,157,158,159]. These genes encode proteins that are components of the transforming growth factor beta (TGFβ) signaling pathway, which regulates various cellular processes [109, 160, 161]. Hereditary haemorrhagic telangiectasia type 1 (HHT1, MIM # 187300) is associated with mutations in the gene ENG that encodes endoglin, a dimeric glycoprotein that functions as a co-receptor on the plasma membrane. It is predominantly expressed in vascular endothelial cells of various tissues and organs throughout the body and it is therefore essential for the normal structure of the blood vessels [162]. In earlier work, we have utilized glycosylation profiling assays and immunofluorescence microscopy to demonstrate that several disease-causing missense endoglin variants get trapped in the ER by the machinery of the ERQC mechanism and fail to traffic to the plasma membrane, where they function [13]. Subsequently, we have also demonstrated the implication of ERAD in the degradation of ER-retained missense mutant variants P165L and V105D using HRD1-knockout HEK293 invitro cellular model [163]. Protein elimination leads to a haploinsufficiency state, ultimately contributing to the manifestation of the disease phenotype. Given that endoglin is a homodimeric protein synthesized in the ER, it was logical to explore whether mutant variants expressed by the affected allele would interact with WT endoglin, potentially leading to a dominant-negative effect. Interestingly, our co-immunoprecipitation assays have clearly demonstrated that ER-retained endoglin variants (L32R, V105D, P165L, I271N and C363Y) heterodimerize with WT endoglin in a dominant-negative manner impairing its trafficking to the plasma membrane [47]. This mechanism is likely to exacerbates the disease state, resulting in a scenario where 50% of the protein is lost due to the loss of one allele leading to haploinsufficiency, coupled with a possible additional ~ 25% loss attributed to the dominant-negative effects exerted by the mutant ER-retained protein on its WT counterpart. Our findings were consistent with two prior studies that briefly illustrated the formation of mixed dimers of endoglin WT and mutant variants [164, 165].

Hereditary haemorrhagic telangiectasia type 2 (HHT2, MIM # 600376) is associated with mutations in ACVRL1 gene, encoding activin receptor-like kinase, also denoted as ALK1, a type 1 receptor in the TGFβ signaling pathway [166]. Both HHT1 and HHT2 are presented with similar phenotypes, as both endoglin and ALK1 play a crucial role in endothelial cells differentiation during capillary development leading to vascular malformation phenotypes in both disorders [167]. Similar to endoglin, both our research and other studies have shown that several missense mutant variants located at the intracellular kinase domain of ALK1 receptor become entrapped in the ER and fail to traffic to the plasma membrane where they normally perform their functional role in TGFβ signaling cascade [16, 164, 168]. Currently, haploinsufficiency has been accepted as the primary disease mechanism [158, 169]. However, the homodimeric nature of the ALK1 receptor raises a strong possibility that some of these ER-retained mutants may form heterodimeric complexes with the WT, thereby hijacking it and impairing its trafficking to the plasma membrane in a dominant-negative manner. Recently, variants of ALK1 that exhibit a normal trafficking to the plasma membrane were also proposed to exert a dominant-negative effect on the WT via forming dysfunctional heterodimers on the plasma membrane, which further impedes the 50% functionality of WT expressed by the unaffected allele [49]. These findings further consolidate the prediction that some ER-retained mutants may exhibit a dominant-negative effect on the WT protein through heterodimerization between the mutant and WT alleles.

Pulmonary arterial hypertension (PAH, # 178600) is another hereditary vascular disease associated with heterozygous mutations in BMPR2 gene, encoding yet another type 2 receptor; bone morphogenetic protein receptor 2 [170]. Considerable research efforts have been carried out to understand the molecular mechanisms that may contribute to the wide spectrum of the disease phenotypes as well as the reduced penetrance rate, (reviewed in [171]. In an effort to understand the mechanisms by which missense mutant variants lose their functionality, we investigated the trafficking of various disease-causing variants spanning all receptor’s domains. Our findings revealed that some variants that harbor mutations at the ligand binding domain are entirely or partially trapped in the ER, ultimately leading to premature degradation through, most likely, the ERAD mechanism [15]. Retention of disease-causing missense variants of BMPR2 and the consequential defective trafficking of the receptor to the plasma membrane, which also has also been reported by others, further consolidates the implication of the ERQC mechanism in the pathogenesis of PAH [50, 172]. Furthermore, evidence of a dominant-negative effect exerted by BMPR2 missense variants on a type 1 receptor has also been reported [50]. Remarkably, there has been no exploration into whether those ER-retained mutants would exert a dominant-negative effect on WT BMPR2 expressed from the unaffected allele. Like endoglin and ALK1, BMPR2 is a homodimeric protein. This characteristic raises the possibility that heterodimerization between WT and dominant-negative ER-retained mutants may occur during the early stages of protein biogenesis in the ER, impairing its trafficking to the plasma membrane.

Loeys-Dietz Syndrome (LDS, MIM # 609192) is a rare genetic disorder inherited in an autosomal dominant manner [173]. It is characterized by multisystemic phenotypic presentations including aortic/arterial aneurysms in addition to craniofacial, osteoarticular, musculoskeletal, and cutaneous malformations [173, 174]. Up to date, mutations in six genes (TGFBR1, TGFBR2, SMAD2, SMAD3, TGFB2, and TGFB3) that encode TGFβ signaling components have been associated with LDS [174, 175]. Functional assays have demonstrated that mutated TGFBR1 interfere with the endogenously expressed WT receptor, reducing its activity in a dominant-negative manner [52]. These findings open the doors for further investigation into the molecular mechanisms that lead to the loss of function of these major components in the signaling pathway. A possible dominant-negative effect exerted by the mutant allele on the WT may also represent logical contributor to an aggravated haploinsufficiency state in LDS. Furthermore, considering that LDS is associated with several mutant variants of components functioning along the same signaling pathway, it is worthwhile to investigate whether some of these mutant components might interfere with the function of multiple components within the signaling pathway, potentially contributing to the observed heterogeneity in LDS phenotypes.

It is crucial to highlight that the majority of TGFβ signaling pathways involve dimeric proteins, whether secretory or membrane-bound. As illustrated in a prior review, through an extensive literature search, we have shown that these proteins are implicated in around 25 monogenic human diseases [109]. However, the disease mechanisms of these conditions remain underexplored in terms of possible implication of ERQC mechanism and also the potential existence of dominant-negative effects. Further investigation into these aspects is warranted to enhance our understanding of the pathogenesis associated with TGFβ signaling-related monogenic diseases.

Long QT syndromes (LQTS) are a group of autosomal inherited arrhythmogenic disorders characterized by abnormal cardiac activity presented by prolonged QT intervals, leading to a type of arrhythmia known as torsades de pointes [176]. Irregularities in the heartbeat have the potential to result in fainting, seizures, or sudden cardiac arrest. LQTS is classified into three primary types based on the causative genes: LQTS1, MIM # 192500, LQST2, MIM # 613688, and LQTS3, MIM # 603830 encoded by the genes (KCNQ1), (KCNH2) and (SCN5A), respectively. These genes encode ion channels essential for cardiac repolarization [177]. Nonetheless, each type has distinct triggers, clinical manifestations, severity and penetrance profile, suggesting variable molecular mechanisms involved, in addition to environmental factors, age and gender [178]. LQTS2 is associated with KCNH2, a gene that encodes the voltage-gated K+ channel α-subunit (Kv11.1), which function as tetrameric complex that consists of four Kv11.1 α-subunit [179].

Ficker and colleagues reported through immunoprecipitation analysis that Kv11.1 disease-causing variants R752W and G601S show defective trafficking, evidenced by their strong association with molecular chaperones Hsp90 and Hsp70 in the ER. Defective trafficking results in ER-retention of misfolded Kv11.1variants, followed by premature degradation through the ERAD mechanism. Conversely, the non-functional G628S variant displayed transient associations with the molecular chaperones before being released to the plasma membrane, similar to WT Kv11.1 [54]. Characterizing the physiological properties of Kv11.1 disease-causing variants harboring the missense mutation (E637K) situated in the pore-S6 loop of the channel, using a Xenopus oocyte heterologous expression system, revealed intriguing findings. Coexpression of WT (WT) and E637K variants resulted in a peak tail current significantly lower than the current peaks anticipated from the WT alone. These findings suggest that this mutation exerts a dominant-negative effect on the WT Kv11.1 channel and highlights the significance of the pore-S6 loop in the channel's function [180, 181]. Therefore, if a dominant-negative effect can occur on the plasma membrane through the formation of the WT/mutant tetrameric complexes, then it follows logically that ER-retained mutants may also form similar tetrameric complexes in the ER, that impede the trafficking of the WT ion channel to the plasma membrane, employing a similar dominant-negative mechanism.

Neurological disorders

The spectrum of molecular mechanisms underlying monogenic neurological disorders with autosomal dominant inheritance is diverse, reflecting the wide array of genes and protein variants involved. Nonetheless, protein misfolding and aggregation are recurrent themes, leading to cellular death and permanent neurological dysfunction. Phenotypic traits can be further aggravated by dominant-negative variants that interfere with the remaining 50% functionality of the WT protein.

Heterozygous mutations in the GABAA receptor gamma2 subunit encoded by the gene GABRG2 have been associated with generalized epilepsy with febrile seizures plus (GEFS + , MIM # 607681) [59, 182]. The GABAA receptor, a pentameric ligand-gated ion channels serving as a receptor for the inhibitory neurotransmitter gamma-aminobutyric acid (GABA), is typically composed of two α subunits, two β subunits, and one γ2 subunit [183]. The molecular mechanisms associated with disease-causing variants were identified to involve haploinsufficiency attributable to the mutant allele, along with the exertion of a dominant-negative effect by the mutated γ2 subunit. Kang and colleagues have demonstrated, using in vitro cellular models, that the pathogenic variant γ2(Q351X) associated with GEFS exhibits defective trafficking to the plasma membrane, leading to proteasomal degradation through ERAD. Additionally, γ2(Q351X) was shown to exert a dominant-negative effect on WT receptors, reducing their assembly, trafficking, and surface expression ( 59). This effect likely arises from the oligomerization of mutant and other WT subunits that form the full pentameric receptor complex, resulting in ER retention of both WT and mutant subunits followed by premature degradation through ERAD. Pulse-chase experiments revealed that coexpression of the γ2 subunit mutation, Q351X, with other WT subunits reduced GABAA function to a level lower than the predicted 50% level for heterozygous carriers. Therefore, the combined mechanism of ER retention and dominant-negative effect exerted by the mutant γ2 subunit on partnering subunits is predicted to be the most likely mechanism for the manifestation of GEFS + associated with this mutation.

A combination of haploinsufficiency and dominant-negative effect mechanisms has also been identified as the underlying mechanism for severe cases of Neurofibromatosis type 1 (NF1, MIM # 162200), a rare genetic disorder characterized by the development of tumors on nerve tissues throughout the body [61]. Neurofibromin, encoded by NF1 gene, is a tumor suppressor and a GTPase activating proteins that regulates RAS signaling cascade in various cell types including neuronal cells [184]. In invitro cellular models, it has been demonstrated that misfolded neurofibromins, encoded by variants in codons 844 to 848, exert a destabilizing effect on the WT protein through protein dimerization in the ER. Heterodimeric complexes are recognized by the ERQC mechanism and marked for degradation via ERAD mechanism. This interaction results in a complete reduction of neurofibromin levels, surpassing the threshold observed in haploinsufficiency alone [185].

In genetic disorders, dominant-negative effect might also occur when a mutated protein disrupts the function of another protein it unexpectedly interacts with, leading to the manifestation of disease symptoms. This phenomenon is best demonstrated by missense variants of voltage-gated potassium channel (Kv3.3) associated with autosomal dominant spinocerebellar ataxias 13 (SCA13, MIM #176264), encoded by the gene KCNC3 [186]. These misfolded variants display impaired trafficking, resulting in entrapment within endosomal vesicles and a failure to reach their designated functional location at the plasma membrane [60]. Intriguingly, they also intracellularly engage with the human epidermal growth factor receptor (EGFR) through an unknown mechanism, implying a pivotal role for EGFR in cerebellum development and, consequently, highlighting its involvement in the pathology of SCA13.

DYT1, also known as early onset torsion dystonia, is an autosomal dominant neurological disease characterized by involuntary muscle contractions (dystonic movements) affecting several parts of the body, resulting in severe disability [187]. The disease typically begins in childhood or adolescence and represents the most common and severe type of dystonias. DYT1 dystonia is caused by a specific mutation in the TOR1A gene, which involves the deletion of three base pairs (GAG), resulting in the loss of a single glutamic acid residue in the torsinA protein at position 302 (ΔE-torsinA) [63]. The torsinA protein is an ER-resident glycoprotein and a member of the AAA + (ATPases Associated with diverse cellular Activities) protein family that play key roles in cellular functions related to protein folding, trafficking, and/or degradation [188]. Oligomerization is a conserved feature of members of the AAA+ family of ATPases, however, unlike WT-torsinA, mutant ΔE-torsinA is mislocalized and forms perinuclear aggregates [63]. Furthermore, through coimmunoprecipitation assays, it has been shown that WT torsinA interacts with the mutant protein, resulting in a dominant-negative effect by sequestering WT protein to the perinuclear region, where they form multimeric protein complexes [64]. The number of remaining functional WT-torsinA multimers would depend on the expression ratio of WT-torsinA: ΔE-torsinA. Interestingly, investigations into the degradation pathways of WT and mutant ΔE torsinA proteins have revealed divergent degradation pathways for each. WT-torsinA was found to degrade through autophagy, while ΔE-torsinA, which exhibited a significantly shorter half-life, was selectively and efficiently degraded via the proteasome through ERAD, rendering it a potential target for interventional rescue [64].

Eye disorders

Retinitis pigmentosa (RP, MIM # 613731) is a genetic degenerative eye disorder that is associated with mutations in numerous genes, which contribute to the heterogeneity of the disease. Mutations in rhodopsin gene (RHO) have been identified as one of the most common causes of autosomal dominant PR [189]. Rhodopsin is a light-sensitive receptor protein that plays a crucial role in the conversion of light signals into electrical signals in rod photoreceptor cells of the retina. RHO mutations have been categorized into seven categories according to their effect on the protein’s structure and function, reviewed in [67]. Rhodopsin has a high ability to form dimeric and higher order oligomers which is believed to play a key role in the photoreceptor function [190]. The most common disease-causing misfolded rhodopsin variants P23H and K296E were found to form ER-retained aggregates followed by proteasomal degradation. But if degradation was not complete, mutant rhodopsin can accumulate in photoreceptor cells, leading to cell degeneration and contributing to the development of the disease phenotypes [66]. In addition, It was demonstrated using fluorescence microscopy and immunoblot analysis that co-expression of variants P23H or G188R, together with the WT rhodopsin has resulted in the entrapment of WT rhodopsin in the ER. Misfolded dimers aggregate with the WT Rhodopsin exerting a dominant-negative effect through hetero-oligomerization [68, 69].

Primary open angle glaucoma (POAG, # 137750), has also been identified as a leading cause for irreversible blindness due to damaged optic nerve [191]. Mutations in the myocilin gene (MYOC) have been associated with the disorder in several families with early onset of visual impairment. Total loss of myocilin in samples of patients harboring heterozygous mutations has been attributed to a combinational mechanism of haploinsufficiency and dominant-negative effects exerted by the entrapped mutant myocilin on the WT expressed from the functional allele [72].

Furthermore, Wolfram syndrome (WS, MIM # 222300) is a multisystemic syndrome that is characterized primarily by diabetes mellitus, optic atrophy which represent a major feature affecting nearly all reported patients, in addition to sensorineural deafness, neurodegeneration and psychological imbalances [192]. The autosomal dominant form of the disease is caused by heterozygous mutations in the WFS1 gene that encodes wolframin, a transmembrane protein that regulates calcium homeostasis within the ER. In contrast to homozygous mutants, It has been reported by many that some heterozygous misfolded wolframin exhibit a dominant-negative effect on the WT as it aggregates in the ER lumen causing ER stress and cellular toxicity [75, 193, 194].

Serpinopathies

Serpins are a large group of serine protease inhibitors that play a key role in regulating proteases activities across various organs [195]. Mutations in these genes result in the aggregation of mutant proteins, inducing cellular dysfunction and giving rise to a spectrum of monogenic disorders collectively termed serpinopathies. Serpins are inherently unstable, and this is mainly due to their mechanism of action, as they alternate between folded and unfolded state in order to perform their inhibitory function. Mutations that lead to unfolded proteins destabilize this fine balance and promote protein aggregate formation [196]. Heterozygous mutations in SERPINC1, SERPINA1and SERPING1 have been associated with autosomal dominant antithrombin deficiency (MIM #107300), Alpha-1-antitrypsin deficiency (A1ATD, MIM # 613490) and hereditary angioedema type 1 (HAE1,MIM # 106100), respectively. Dominant-negative variants of these three genes have been associated with the formation of mutant/WT protein aggregates in the ER, resulting in significant reduced plasma levels of the respective proteins and correlating with severe disease phenotypes [78, 79, 81].

Therapeutic challenges and emerging strategies in dominant-negative disorders

Treating conditions that include dominant-negative effect as a contributing mechanism can be a difficult task for medical professionals, as it presents a significant therapeutic challenge. Simply increasing protein levels is not always an effective strategy because as highlighted earlier, the mutant protein may interfere with the function of its WT counterpart [197]. This combination of the "poisoning" effect and potential ER retention and aggregation, resulting from the ER machinery involvement, requires alternative therapeutic approaches. Strategies designed to address the pathological consequences of some dominant-negative mutations fall into three main categories, utilizing diverse tools and techniques summarized in (Fig. 3).

Fig. 3
figure 3

Therapeutic Strategies for Dominant-negative Disorders. A Genetic modulation directly tackles the faulty gene through diverse techniques by introducing a functional copy, eliminating the mutated allele, combining both approaches, or even editing the mutation itself using advanced tools like CRISPR/Cas9 and base editing techniques B) Post-translational modulation mainly targets correcting the underlying folding or structural deformity through chemical and pharmacological chaperones, or through directly manipulating specific components of the ER quality control machinery. For some diseases, recombinant proteins are administered to compensate for the loss of the WT protein C) Pharmacological bypass therapy uses drugs or different pharmacological compounds to mimic the function of the WT protein or compensate for its absence, even without repairing the gene, to relieve the underlying clinical symptoms by targeting downstream pathways affected by the mutation

Therapeutic interventions aimed at counteracting dominant-negative effects may show promise for disorders characterized by ER-retention dominant-negative mechanisms. However, their efficacy depends on factors such as the nature of the disorder, the specific genetic mutation involved, and the mechanism of action of the intervention.

Genetic modulation

The straightforward approach in genetic modulation that comes to mind is to introduce a functional copy of the defective gene into the diseased cell. Therapeutic approaches aimed at increasing the amount of WT protein in autosomal dominant diseases caused by dominant-negative variants can also be effective for variants exhibiting the combinational mechanism of ER retention and dominant-negative effects. The underlying principle is the same: enhancing the availability of functional WT protein to compensate for the defective mutant protein. By boosting the levels of WT protein, these therapies can mitigate the impact of the dominant-negative mutant, regardless of whether the mutation leads to ER retention or other misfolding issues. This strategy holds promise for improving cellular function and alleviating disease symptoms across a range of genetic disorders with similar pathogenic mechanisms. In RP disorder for example, increasing WT rhodopsin to three-fold its normal level in the eyes of transgenic mice carrying the P23H ER-retained/dominant-negative variant has been shown to protects the retina, suggesting gene therapies that carefully boost rhodopsin levels could alleviate such diseases [198]. Delivering a normal copy of the RHO gene via adeno-associated virus serotype 5 (AAV) vector prevented retinal degeneration in P23H transgenic mice through the increased expression of WT rhodopsin. This aims to increase the WT to mutant protein ratio, potentially out-competing the mutant protein and subsequently restoring some function [199]. Similarly, the introduction of a WT copy of the GABRG2 gene into transgenic mice carrying the dominant-negative Q390X variant (Gabrg2 + /Q390X) associated with Dravet syndrome (epileptic encephalopathy) significantly rescued their underlying seizures [58]. However, mutated proteins with dominant-negative effects may still exert detrimental effects even with increased WT protein expression. Therefore, treating such conditions might necessitates targeting the mutant allele at the DNA or RNA levels [200]. AAV vectors have been employed to selectively disrupt the expression of the dominant-negative allele in the COL1A1 gene through the insertion of a neomycin resistance cassette to the first exon of the gene in mesenchymal stem cells (MSCs) derived from patients presented with OI, successfully demonstrating targeted gene modification in adult human stem cells [201]. Alternatively, various methodologies can be used to silence the mutated allele at the transcription level via antisense oligodeoxyribonucleotides (ODNs), short interfering RNA (siRNA), and hammerhead ribozymes. In this context, highly specific siRNA has been designed to selectively suppress the mutant torsin A protein, the primary causative factor in the most common form of primary generalized dystonia [202]. It efficiently suppressed the mutant torsin A in cells mimicking the heterozygous state without affecting the WT allele.

Given the limitations of each approach individually in effectively bypassing the underlying defect, especially for cells sensitive to haploinsufficiency, their synergistic application emerges as a viable solution. Dotzler SM and colleagues presented a therapeutic solution by incorporating a suppression-and-replacement genetic approach for LQT1 syndrome that utilizes a two-component strategy [203]. The first component is based on the use of a short hairpin RNA (shRNA) to specifically suppress the expression of the patient's endogenous, mutated KCNQ1 gene. Secondly, the introduction of a codon-altered, shRNA-immune copy of KCNQ1, thereby achieving functional replacement of the defective allele. This dual-pronged approach demonstrates promising preclinical efficacy, as evidenced by the successful restoration of normal function in induced pluripotent stem cell-derived cardiomyocytes harboring diverse LQT1-causing KCNQ1 variants. Furthermore, it represents a feasible therapeutic strategy that can effectively address the double whammy of ER-retention and dominant negative effects by specifically silencing the mutant allele, which prevents the production of the defective protein, thereby reducing its detrimental interactions with the WT protein. Additionally, it helps in mitigating issues related to ER retention, as fewer mutant proteins are available to be retained in the ER. Consequently, more WT proteins can function correctly, improving cellular function and potentially ameliorate disease symptoms.

Fueled by the discovery of CRISPR/Cas9 technology, genetic tools have been developed to directly correct the disease-causing variant in different genetic diseases [204]. CRISPR/Cas9-mediated gene correction has been implemented in OI patients’ derived iPSCs which were differentiated to Osteoblast cells with recovered type I collagen levels [205]. Besides that, Huang et al. leveraged cutting-edge base editing technology to precisely repair a disease-causing mutation (FBN1; T7498C) in MFS, demonstrating the potential of this approach for gene therapy in MFS and other genetic disorders [26]. Unlike CRISPR/Cas9 base editing technology, the proposed technology is more precise with minimal risk of unintended mutations as it does not require the generation of a double-strand break to correct the intended nucleotide [206].

Post-translational modulation

Instead of genetically manipulating the affected gene, administration of exogenous WT proteins through protein replacement therapy has shown successful outcomes in a few diseases with dominant-negative pathophysiology. Although not universally applicable to dominant-negative disorders, the clinical success of recombinant intravenous (IV) C1INH formulations in hereditary angioedema patients underscores the therapeutic potential of protein replacement therapy for this class of diseases [207]. It demonstrates its ability to mitigate the pathological protein misfolding and abnormal ER aggregation caused by the underlying heterozygous SERPING1 variations. It's important to recognize that protein replacement therapy, despite its success in some dominant-negative diseases, may not be feasible for all due to technological limitations and individual patient factors.

Interventions that promote proper folding and prevent aggregation of the mutated protein show promise in rescuing the WT protein from ER entrapment. Pharmacological chaperones (Pcs) have been used in this context to specifically bind to the mutated protein promoting its proper folding and stabilization which will subsequently prevent its retention and premature degradation along with its WT counterpart [208]. Therefore, the application of Pcs presents a compelling strategy for targeting the combinational mechanism of dominant negative effects exerted by ER-retained mutant variants. Several studies have demonstrated the potential of retinoid analogs to act as specific PC compounds for the P23H mutation in rhodopsin, which causes RP [209]. These chaperones enhance the folding of the mutant protein and reduce its dominant-negative effect on the processing of the WT form [69]. Similarly, the IN3 PC compound corrects folding errors of several GnRH receptor (GnRHR) mutants; causative of hypogonadotropic hypogonadism, and promotes its correct intracellular trafficking along with its interacting WT subunits [210]. Unlike the targeted approach of PCs, chemical chaperones exhibit a broad-spectrum effect stabilizing various proteins and preventing aggregation in a non-specific manner [211]. For example, 4-phenylbutyrate (4-PBA) is a clinically approved medication for urea cycle disorders that showed its potential to prevent P23H rhodopsin aggregation and reduce the associated ER stress in RP [67]. In addition, protein rescue may also involve targeting specific components of ERAD and the ER machinery. Proteasomal inhibitors such as MG-132, MG-115, lactacystin, or proteasome inhibitor I prevented the premature degradation of ER-tagged caveolin-3 mutants, rescuing their interacting WT forms in a LGMD-1C cellular model [34]. Moreover, targeting the abberent activation of the UPR pathway due to ER stress in cells with accumulated misfolded proteins may offer a potential therapeutic approach in dominant-negative diseases. In a mouse model of RP, knocking out ATF4 in mice expressing the dominant-negative T17M rhodopsin mutation halted retinal degeneration. Blocking ATF4 expression lead to the downregulation of multiple UPR components like pEIF2α, ATF6, and CHOP, ultimately blocking the activation of cell death pathways [212]. Overall, addressing distinct elements within the ER machinery, aiming to mitigate the dominant-negative consequences caused by misfolded proteins and restore the WT from the underlying damage, signifies an innovative and promising frontier where cell biology intersects with medicine.

Pharmacological bypass therapy

Besides, several therapeutic interventions have been utilized to bypass the need to directly manipulate the underlying defect with various agents or pharmacological medications, often referred to as phenotypic correctors, that resemble the downstream effects of the WT protein. In ISS therapy, long-term growth hormone (GH) treatment can increase the height in childhood and adult life of familial and nonfamilial ISS cases including patients carrying heterozygous variants in the NPR2 gene showing dominant-negative effects [213]. Despite the response variability towards GH therapy, several NPR2 cases showed promising responses in height correction, especially with earlier (before puberty) and long-term administration [32, 214]. On the other hand, myoblast cultures derived from patients with UCMD, caused by mutations in COL6A1, COL6A2, or COL6A3 genes, displayed increased cellular apoptosis. Oral treatment with cyclosporine A (an immunosuppressive drug; CsA) for one month significantly reduced apoptosis through the normalization of the mitochondrial membrane potential of the tested muscle cells [215]. The overall conclusion from this pilot study is that long-term CsA treatment influences myofiber regeneration and ameliorates muscle cell performance in treated patients. HAE1, characterized by uncontrolled plasma kallikrein due to C1INH deficiency even with heterozygous carriers, shows enhanced treatment response to drugs inhibiting kallikrein, leading to significant clinical improvement [207].

Future perspectives and conclusions

The dominant-negative effects exerted by mutant proteins on either their WT allele or interacting partners represent a major mechanism underlying various autosomal dominant genetic diseases and may contribute significantly to their wide spectrum of phenotypic clinical manifestations. Furthermore, an additional combined mechanism emerges when ER-retained mutant proteins form mixed complexes with WT counterparts or multi-subunit partners, resulting in the mis-localization and premature degradation of these WT partners. As a consequence, an additional loss of functional protein occurs, further compromising cellular function and exacerbating disease phenotypes. Thus, the dual additive impact of the dominant-negative effects and ERAD-mediated degradation is playing a pivotal role in the complexity of disease pathogenesis in numerous autosomal genetic disorders. Notably and surprisingly, this specific and highly damaging combinatorial mechanism remains relatively understudied and underappreciated in the field. This review represents an initial effort to illuminate and highlight this aspect of research, presenting significant potential for elucidating the factors influencing variant-associated phenotypic variability and detailed disease pathogenesis in numerous conditions. By highlighting these complex interactions, this review aims to promote further exploration and potentially uncover novel avenues for understanding and addressing mechanisms underlying autosomal dominant diseases. Furthermore, understanding these intricate mechanisms may offer insights into potential novel therapeutic strategies aimed at mitigating clinical presentations in these diseases including ameliorating their severity.

Availability of data and materials

All dataset was incorporated in this manuscript.

Abbreviations

4-PBA:

4-Phenylbutyrate

ATF6:

Activating transcription factor 6

BMD:

Bethlem myopathy disorder

CFTR:

Cystic fibrosis transmembrane conductance regulator

CNP:

C-type natriuretic peptide

CNX/CRT:

Calnexin/calreticulin

DEB:

Dystrophic epidermolysis bullosa

ECM:

Extracellular matrix

EDS:

Ehlers-Danlos syndrome

EGFR:

Epidermal growth factor receptor

ER:

Endoplasmic reticulum

ERAD:

ER-associated protein degradation

ERQC:

ER quality control

GABA:

Gamma-aminobutyric acid

GEFS + :

Febrile seizures plus

GH:

Growth hormone

GnRHR:

GnRH receptor

HHT1:

Hereditary haemorrhagic telangiectasia type 1

HHT2:

Hereditary haemorrhagic telangiectasia type 2

Hsp:

Heat shock proteins

IRE1 :

Inositol requiring enzyme 1

ISS:

Idiopathic short stature

IV:

Intravenous

LDS:

Loeys-Dietz Syndrome

LGMD:

Limb-girdle muscular dystrophy

LQTS:

LQT syndromes

MFS:

Marfan syndrome

MSCs:

Mesenchymal stem cells

MYOC:

Myocilin gene

NMD:

Nonsense-mediated decay

NF1:

Neurofibromatosis Type 1

ODNs:

Oligodeoxyribo nucleotides

OI:

Osteogenesis imperfecta

PAH:

Pulmonary arterial hypertension

Pcs:

Pharmacological chaperones

PERK:

Protein kinase RNA like endoplasmic reticulum kinase

POAG:

Primary open angle glaucoma

RHO:

Rhodopsin gene

RP:

Retinitis pigmentosa

shRNA:

Short hairpin RNA

siRNA:

Short interfering RNA

STL1:

Stickler syndrome type1

STL2:

Stickler syndrome type 2

TGFβ:

Transforming growth factor beta

UCMD:

Ullrich congenital muscular dystrophy

UGGT:

UDP-glucose: glycoprotein glucosyltransferase

UPR:

Unfolded protein response

VWD:

Von Willebrand disease

VWF:

Von Willebrand factor

WS:

Wolfram syndrome

WT:

Wild-type

References

  1. Li H,Sun S. Protein aggregation in the ER: calm behind the storm. Cells. 2021;10:3337.

  2. Liu Y, Ye Y. Proteostasis regulation at the endoplasmic reticulum: a new perturbation site for targeted cancer therapy. Cell Res. 2011;21:867–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cheng X, Wang K, Zhao Y. Research progress on post-translational modification of proteins and cardiovascular diseases. Cell Death Discov. 2023;9:275.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rizzolo LJ, Kornfeld R. Post-translational protein modification in the endoplasmic reticulum. Demonstration of fatty acylase and deoxymannojirimycin-sensitive alpha-mannosidase activities. J Biol Chem. 1988;263:9520–5.

    Article  CAS  PubMed  Google Scholar 

  5. Sun Z, Brodsky JL. Protein quality control in the secretory pathway. J Cell Biol. 2019;218:3171–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Duttler S, Pechmann S, Frydman J. Principles of cotranslational ubiquitination and quality control at the ribosome. Mol Cell. 2013;50:379–93.

    Article  CAS  PubMed  Google Scholar 

  7. Chen PY, Qin L, Simons M. TGFβ signaling pathways in human health and disease. Front Mol Biosci. 2023;10:1113061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kim G, Lee J, Ha J, et al. Endoplasmic reticulum stress and its impact on adipogenesis: molecular mechanisms implicated. Nutrients. 2023;15:5082.

  9. Antonarakis SE, Krawczak M, Cooper DN. Disease-causing mutations in the human genome. Eur J Pediatr. 2000;159(Suppl 3):S173-178.

    Article  CAS  PubMed  Google Scholar 

  10. Yue P, Li Z, Moult J. Loss of protein structure stability as a major causative factor in monogenic disease. J Mol Biol. 2005;353:459–73.

    Article  CAS  PubMed  Google Scholar 

  11. Veitia RA, Caburet S, Birchler JA. Mechanisms of Mendelian dominance. Clin Genet. 2018;93:419–28.

    Article  CAS  PubMed  Google Scholar 

  12. Gerasimavicius L, Livesey BJ, Marsh JA. Loss-of-function, gain-of-function and dominant-negative mutations have profoundly different effects on protein structure. Nat Commun. 2022;13:3895.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ali BR, Ben-Rebeh I, John A, et al. Endoplasmic reticulum quality control is involved in the mechanism of endoglin-mediated hereditary haemorrhagic telangiectasia. PLoS One. 2011;6: e26206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ali BR, Silhavy JL, Gleeson MJ, et al. A missense founder mutation in VLDLR is associated with Dysequilibrium Syndrome without quadrupedal locomotion. BMC Med Genet. 2012;13:80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. John A, Kizhakkedath P, Al-Gazali L, et al. Defective cellular trafficking of the bone morphogenetic protein receptor type II by mutations underlying familial pulmonary arterial hypertension. Gene. 2015;561:148–56.

    Article  CAS  PubMed  Google Scholar 

  16. Hume AN, John A, Akawi NA, et al. Retention in the endoplasmic reticulum is the underlying mechanism of some hereditary haemorrhagic telangiectasia type 2 ALK1 missense mutations. Mol Cell Biochem. 2013;373:247–57.

    Article  CAS  PubMed  Google Scholar 

  17. Kizhakkedath P, John A, Al-Sawafi BK, et al. Endoplasmic reticulum quality control of LDLR variants associated with familial hypercholesterolemia. FEBS Open Bio. 2019;9:1994–2005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Badawi S, Varghese DS, Raj A, et al. Unveiling the pathogenic mechanisms of NPR2 missense variants: insights into the genotype-associated severity in acromesomelic dysplasia and short stature. Front Cell Dev Biol. 2023;11:1294748.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hegde RN, Subramanian A, Pothukuchi P, et al. Rare ER protein misfolding-mistrafficking disorders: Therapeutic developments. Tissue Cell. 2017;49:175–85.

    Article  CAS  PubMed  Google Scholar 

  20. Volpi VG, Touvier T, D’Antonio M. Endoplasmic reticulum protein quality control failure in myelin disorders. Front Mol Neurosci. 2016;9:162.

    CAS  PubMed  Google Scholar 

  21. Chen Y, Bellamy WP, Seabra MC, et al. ER-associated protein degradation is a common mechanism underpinning numerous monogenic diseases including Robinow syndrome. Hum Mol Genet. 2005;14:2559–69.

    Article  CAS  PubMed  Google Scholar 

  22. Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev. 2012;92:537–76.

    Article  CAS  PubMed  Google Scholar 

  23. Ferro-Novick S, Reggiori F, Brodsky JL. ER-Phagy, ER homeostasis, and ER quality control: implications for disease. Trends Biochem Sci. 2021;46:630–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Whiteman P, Handford PA. Defective secretion of recombinant fragments of fibrillin-1: implications of protein misfolding for the pathogenesis of Marfan syndrome and related disorders. Hum Mol Genet. 2003;12:727–37.

    Article  CAS  PubMed  Google Scholar 

  25. Dietz HC, McIntosh I, Sakai LY, et al. Four novel FBN1 mutations: significance for mutant transcript level and EGF-like domain calcium binding in the pathogenesis of Marfan syndrome. Genomics. 1993;17:468–75.

    Article  CAS  PubMed  Google Scholar 

  26. Zeng Y, Li J, Li G, et al. Correction of the Marfan syndrome pathogenic FBN1 mutation by base editing in human cells and heterozygous embryos. Mol Ther. 2018;26:2631–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chiu HH. An update of medical care in Marfan syndrome. Tzu Chi Med J. 2022;34:44–8.

    Article  PubMed  Google Scholar 

  28. Amano N, Mukai T, Ito Y, et al. Identification and functional characterization of two novel NPR2 mutations in Japanese patients with short stature. J Clin Endocrinol Metab. 2014;99:E713-718.

    Article  CAS  PubMed  Google Scholar 

  29. Hanley PC, Kanwar HS, Martineau C, et al. Short Stature is Progressive in Patients with Heterozygous NPR2 Mutations. J Clin Endocrinol Metab. 2020;105:3190–202.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Vasques GA, Arnhold IJ, Jorge AA. Role of the natriuretic peptide system in normal growth and growth disorders. Horm Res Paediatr. 2014;82:222–9.

    Article  CAS  PubMed  Google Scholar 

  31. Hwang IT, Mizuno Y, Amano N, et al. Role of NPR2 mutation in idiopathic short stature: Identification of two novel mutations. Mol Genet Genomic Med. 2020;8:e1146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Vasques GA, Amano N, Docko AJ, et al. Heterozygous mutations in natriuretic peptide receptor-B (NPR2) gene as a cause of short stature in patients initially classified as idiopathic short stature. J Clin Endocrinol Metab. 2013;98:E1636-1644.

    Article  CAS  PubMed  Google Scholar 

  33. Collett-Solberg PF, Ambler G, Backeljauw PF, et al. Diagnosis, genetics, and therapy of short stature in children: a growth hormone research society international perspective. Horm Res Paediatr. 2019;92:1–14.

    Article  CAS  PubMed  Google Scholar 

  34. Galbiati F, Volonte D, Minetti C, et al. Limb-girdle muscular dystrophy (LGMD-1C) mutants of caveolin-3 undergo ubiquitination and proteasomal degradation. Treatment with proteasomal inhibitors blocks the dominant negative effect of LGMD-1C mutanta and rescues wild-type caveolin-3. J Biol Chem. 2000;275:37702–11.

    Article  CAS  PubMed  Google Scholar 

  35. Georganopoulou DG, Moisiadis VG, Malik FA, et al. A journey with LGMD: from protein abnormalities to patient impact. Protein J. 2021;40:466–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Carotti M, Scano M, Fancello I, et al. Combined Use of CFTR Correctors in LGMD2D myotubes improves sarcoglycan complex recovery. Int J Mol Sci. 2020;21:1813.

  37. Baker NL, Mörgelin M, Peat R, et al. Dominant collagen VI mutations are a common cause of Ullrich congenital muscular dystrophy. Hum Mol Genet. 2005;14:279–93.

    Article  CAS  PubMed  Google Scholar 

  38. Merlini L, Angelin A, Tiepolo T, et al. Cyclosporin A corrects mitochondrial dysfunction and muscle apoptosis in patients with collagen VI myopathies. Proc Natl Acad Sci U S A. 2008;105:5225–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Besio R, Garibaldi N, Leoni L, et al. Cellular stress due to impairment of collagen prolyl hydroxylation complex is rescued by the chaperone 4-phenylbutyrate. Dis Model Mech. 2019;12:e038521.

  40. Duangchan T, Tawonsawatruk T, Angsanuntsukh C, et al. Amelioration of osteogenesis in iPSC-derived mesenchymal stem cells from osteogenesis imperfecta patients by endoplasmic reticulum stress inhibitor. Life Sci. 2021;278:119628.

    Article  CAS  PubMed  Google Scholar 

  41. Pochampally RR, Horwitz EM, DiGirolamo CM, et al. Correction of a mineralization defect by overexpression of a wild-type cDNA for COL1A1 in marrow stromal cells (MSCs) from a patient with osteogenesis imperfecta: a strategy for rescuing mutations that produce dominant-negative protein defects. Gene Ther. 2005;12:1119–25.

    Article  CAS  PubMed  Google Scholar 

  42. Botor M, Fus-Kujawa A, Uroczynska M, et al. Osteogenesis imperfecta: current and prospective therapies. Biomolecules. 2021;11:1493.

  43. Ritelli M, Dordoni C, Venturini M, et al. Clinical and molecular characterization of 40 patients with classic Ehlers-Danlos syndrome: identification of 18 COL5A1 and 2 COL5A2 novel mutations. Orphanet J Rare Dis. 2013;8:58.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Müller GA, Hansen U, Xu Z, et al. Allele-specific siRNA knockdown as a personalized treatment strategy for vascular Ehlers-Danlos syndrome in human fibroblasts. FASEB J. 2012;26:668–77.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hughes A, Oxford AE, Tawara K, et al. Endoplasmic reticulum stress and unfolded protein response in cartilage pathophysiology; contributing factors to apoptosis and osteoarthritis. Int J Mol Sci. 2017;18:665.

  46. Soh Z, Richards AJ, McNinch A, et al. Dominant Stickler syndrome. Genes (Basel). 2022;13:1089.

  47. Gariballa N, Badawi S, Ali BR. Endoglin mutants retained in the endoplasmic reticulum exacerbate loss of function in hereditary hemorrhagic telangiectasia type 1 (HHT1) by exerting dominant negative effects on the wild type allele. Traffic. 2024;25:e12928.

    Article  CAS  PubMed  Google Scholar 

  48. Viteri-Noël A, González-García A, Patier JL, et al. Hereditary hemorrhagic telangiectasia: genetics, pathophysiology, diagnosis, and management. J Clin Med. 2022;11:5245.

  49. Jain K, McCarley SC, Mukhtar G, et al. Pathogenic variant frequencies in hereditary haemorrhagic telangiectasia support clinical evidence of protection from myocardial infarction. J Clin Med. 2023;13:250.

  50. Sobolewski A, Rudarakanchana N, Upton PD, et al. Failure of bone morphogenetic protein receptor trafficking in pulmonary arterial hypertension: potential for rescue. Hum Mol Genet. 2008;17:3180–90.

    Article  CAS  PubMed  Google Scholar 

  51. Maron BA, Abman SH, Elliott CG, et al. Pulmonary arterial hypertension: diagnosis, treatment, and novel advances. Am J Respir Crit Care Med. 2021;203:1472–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Cardoso S, Robertson SP, Daniel PB. TGFBR1 mutations associated with Loeys-Dietz syndrome are inactivating. J Recept Signal Transduct Res. 2012;32:150–5.

    Article  CAS  PubMed  Google Scholar 

  53. Zhou D, Feng H, Yang Y, et al. hiPSC modeling of lineage-specific smooth muscle cell defects caused by. Circulation. 2021;144:1145–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ficker E, Dennis AT, Wang L, et al. Role of the cytosolic chaperones Hsp70 and Hsp90 in maturation of the cardiac potassium channel HERG. Circ Res. 2003;92:e87-100.

    Article  PubMed  Google Scholar 

  55. Bains S, Zhou W, Dotzler SM, et al. Suppression and replacement gene therapy for. Circ Genom Precis Med. 2022;15:e003719.

    Article  CAS  PubMed  Google Scholar 

  56. Peal DS, Mills RW, Lynch SN, et al. Novel chemical suppressors of long QT syndrome identified by an in vivo functional screen. Circulation. 2011;123:23–30.

    Article  PubMed  Google Scholar 

  57. Anderson CL, Kuzmicki CE, Childs RR, et al. Large-scale mutational analysis of Kv11.1 reveals molecular insights into type 2 long QT syndrome. Nat Commun. 2014;5:5535.

    Article  CAS  PubMed  Google Scholar 

  58. Huang X, Zhou C, Tian M, et al. Overexpressing wild-type γ2 subunits rescued the seizure phenotype in Gabrg2. Epilepsia. 2017;58:1451–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kang JQ, Shen W, Macdonald RL. The GABRG2 mutation, Q351X, associated with generalized epilepsy with febrile seizures plus, has both loss of function and dominant-negative suppression. J Neurosci. 2009;29:2845–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Khare S, Nick JA, Zhang Y, et al. A KCNC3 mutation causes a neurodevelopmental, non-progressive SCA13 subtype associated with dominant negative effects and aberrant EGFR trafficking. PLoS ONE. 2017;12:e0173565.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Young LC, Goldstein de Salazar R, Han SW, et al. Destabilizing NF1 variants act in a dominant negative manner through neurofibromin dimerization. Proc Natl Acad Sci U S A. 2023;120:e2208960120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Walker JA, Upadhyaya M. Emerging therapeutic targets for neurofibromatosis type 1. Expert Opin Ther Targets. 2018;22:419–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Torres GE, Sweeney AL, Beaulieu JM, et al. Effect of torsinA on membrane proteins reveals a loss of function and a dominant-negative phenotype of the dystonia-associated DeltaE-torsinA mutant. Proc Natl Acad Sci U S A. 2004;101:15650–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gordon KL, Gonzalez-Alegre P. Consequences of the DYT1 mutation on torsinA oligomerization and degradation. Neuroscience. 2008;157:588–95.

    Article  CAS  PubMed  Google Scholar 

  65. Wu J, Ren J, Luo H, et al. Generation of patient-specific induced pluripotent stem cell line (CSUi002-A) from a patient with isolated dystonia carrying TOR1A mutation. Stem Cell Res. 2021;53:102277.

    Article  CAS  PubMed  Google Scholar 

  66. Saliba RS, Munro PM, Luthert PJ, et al. The cellular fate of mutant rhodopsin: quality control, degradation and aggresome formation. J Cell Sci. 2002;115:2907–18.

    Article  CAS  PubMed  Google Scholar 

  67. Athanasiou D, Aguila M, Bellingham J, et al. The molecular and cellular basis of rhodopsin retinitis pigmentosa reveals potential strategies for therapy. Prog Retin Eye Res. 2018;62:1–23.

    Article  CAS  PubMed  Google Scholar 

  68. Rajan RS, Kopito RR. Suppression of wild-type rhodopsin maturation by mutants linked to autosomal dominant retinitis pigmentosa. J Biol Chem. 2005;280:1284–91.

    Article  CAS  PubMed  Google Scholar 

  69. Mendes HF, Cheetham ME. Pharmacological manipulation of gain-of-function and dominant-negative mechanisms in rhodopsin retinitis pigmentosa. Hum Mol Genet. 2008;17:3043–54.

    Article  CAS  PubMed  Google Scholar 

  70. Musarella MA, Macdonald IM. Current concepts in the treatment of retinitis pigmentosa. J Ophthalmol. 2011;2011:753547.

    Article  PubMed  Google Scholar 

  71. Gobeil S, Rodrigue MA, Moisan S, et al. Intracellular sequestration of hetero-oligomers formed by wild-type and glaucoma-causing myocilin mutants. Invest Ophthalmol Vis Sci. 2004;45:3560–7.

    Article  PubMed  Google Scholar 

  72. Kuchtey J, Chowdhury UR, Uptegraft CC, et al. A de novo MYOC mutation detected in juvenile open angle glaucoma associated with reduced myocilin protein in aqueous humor. Eur J Med Genet. 2013;56:292–6.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Patil SV, Kaipa BR, Ranshing S, et al. Lentiviral mediated delivery of CRISPR/Cas9 reduces intraocular pressure in a mouse model of myocilin glaucoma. Sci Rep. 2024;14:6958.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jain A, Zode G, Kasetti RB, et al. CRISPR-Cas9-based treatment of myocilin-associated glaucoma. Proc Natl Acad Sci U S A. 2017;114:11199–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Batjargal K, Tajima T, Jimbo EF, et al. Effect of 4-phenylbutyrate and valproate on dominant mutations of WFS1 gene in Wolfram syndrome. J Endocrinol Invest. 2020;43:1317–25.

    Article  CAS  PubMed  Google Scholar 

  76. Urano F. Wolfram syndrome iPS cells: the first human cell model of endoplasmic reticulum disease. Diabetes. 2014;63:844–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rigoli L, Caruso V, Salzano G, et al. Wolfram syndrome 1: from genetics to therapy. Int J Environ Res Public Health. 2022;19:3225.

  78. Bravo-Pérez C, Toderici M, Chambers JE, et al. Full-length antithrombin frameshift variant with aberrant C-terminus causes endoplasmic reticulum retention with a dominant-negative effect. JCI Insight. 2022;7:e161430.

  79. Laffranchi M, Berardelli R, Ronzoni R, et al. Heteropolymerization of α-1-antitrypsin mutants in cell models mimicking heterozygosity. Hum Mol Genet. 2018;27:1785–93.

    Article  CAS  PubMed  Google Scholar 

  80. Zieger M, Borel F, Greer C, et al. Liver-directed. Mol Ther Methods Clin Dev. 2022;25:425–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Haslund D, Ryø LB, Seidelin Majidi S, et al. Dominant-negative SERPING1 variants cause intracellular retention of C1 inhibitor in hereditary angioedema. J Clin Invest. 2019;129:388–405.

    Article  PubMed  Google Scholar 

  82. Qiu T, Chiuchiolo MJ, Whaley AS, et al. Gene therapy for C1 esterase inhibitor deficiency in a murine model of hereditary angioedema. Allergy. 2019;74:1081–9.

    Article  CAS  PubMed  Google Scholar 

  83. Herskowitz I. Functional inactivation of genes by dominant negative mutations. Nature. 1987;329:219–22.

    Article  CAS  PubMed  Google Scholar 

  84. Veitia RA. Dominant negative factors in health and disease. J Pathol. 2009;218:409–18.

    Article  CAS  PubMed  Google Scholar 

  85. Veitia RA. Exploring the molecular etiology of dominant-negative mutations. Plant Cell. 2007;19:3843–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Springer TA. von Willebrand factor, Jedi knight of the bloodstream. Blood. 2014;124:1412–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. de Jong A, Dirven RJ, Boender J, et al. Ex vivo improvement of a von Willebrand disease type 2A phenotype using an allele-specific small-interfering RNA. Thromb Haemost. 2020;120:1569–79.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Ghaleb A, Roa L, Marchenko N. Low-dose but not high-dose γ-irradiation elicits the dominant-negative effect of mutant p53 in vivo. Cancer Lett. 2022;530:128–41.

    Article  CAS  PubMed  Google Scholar 

  89. Franken R, den Hartog AW, Radonic T, et al. Beneficial outcome of losartan therapy depends on type of FBN1 mutation in Marfan syndrome. Circ Cardiovasc Genet. 2015;8:383–8.

    Article  CAS  PubMed  Google Scholar 

  90. Daverkausen-Fischer L, Draga M, Pröls F. Regulation of translation, translocation, and degradation of proteins at the membrane of the endoplasmic reticulum. Int J Mol Sci. 2022;23:5576.

  91. Huang B, Sun M, Hoxie R, et al. The endoplasmic reticulum chaperone BiP is a closure-accelerating cochaperone of Grp94. Proc Natl Acad Sci U S A.  2022;119:e2118793119.

  92. Melo EP, Konno T, Farace I, et al. Stress-induced protein disaggregation in the endoplasmic reticulum catalysed by BiP. Nat Commun. 2022;13:2501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Dudek J, Benedix J, Cappel S, et al. Functions and pathologies of BiP and its interaction partners. Cell Mol Life Sci. 2009;66:1556–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Moreno-Gonzalez I, Soto C. Misfolded protein aggregates: mechanisms, structures and potential for disease transmission. Semin Cell Dev Biol. 2011;22:482–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Liu P, Karim MR, Covelo A, et al. The UPR maintains proteostasis and the viability and function of hippocampal neurons in adult mice. Int J Mol Sci. 2023;24:11542.

  96. Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. 2020;21:421–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Ghemrawi R,Khair M. Endoplasmic reticulum stress and unfolded protein response in neurodegenerative diseases. Int J Mol Sci. 2020;21:6127.

  98. Incalza MA, D’Oria R, Natalicchio A, et al. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol. 2018;100:1–19.

    Article  CAS  PubMed  Google Scholar 

  99. Ajoolabady A, Lebeaupin C, Wu NN, et al. ER stress and inflammation crosstalk in obesity. Med Res Rev. 2023;43:5–30.

    Article  CAS  PubMed  Google Scholar 

  100. Ajoolabady A, Liu S, Klionsky DJ, et al. ER stress in obesity pathogenesis and management. Trends Pharmacol Sci. 2022;43:97–109.

    Article  CAS  PubMed  Google Scholar 

  101. Tang Y, Zhou X, Cao T, et al. Endoplasmic reticulum stress and oxidative stress in inflammatory diseases. DNA Cell Biol. 2022;41:924–34.

    Article  CAS  PubMed  Google Scholar 

  102. Lin Y, Jiang M, Chen W, et al. Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother. 2019;118:109249.

    Article  CAS  PubMed  Google Scholar 

  103. Wan L, Chen Z, Yang J, et al. Identification of endoplasmic reticulum stress-related signature characterizes the tumor microenvironment and predicts prognosis in lung adenocarcinoma. Sci Rep. 2023;13:19462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Riordan JR, Rommens JM, Kerem B, et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science. 1989;245:1066–73.

    Article  CAS  PubMed  Google Scholar 

  105. Ron I, Horowitz M. ER retention and degradation as the molecular basis underlying Gaucher disease heterogeneity. Hum Mol Genet. 2005;14:2387–98.

    Article  CAS  PubMed  Google Scholar 

  106. Pornsukjantra T, Saikachain N, Sutjarit N, et al. An increase in ER stress and unfolded protein response in iPSCs-derived neuronal cells from neuronopathic Gaucher disease patients. Sci Rep. 2024;14:9177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Zhu B, Jiang L, Huang T, et al. ER-associated degradation regulates Alzheimer’s amyloid pathology and memory function by modulating γ-secretase activity. Nat Commun. 2017;8:1472.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Park SJ, Kim Y, Chen YM. Endoplasmic reticulum stress and monogenic kidney diseases in precision nephrology. Pediatr Nephrol. 2019;34:1493–500.

    Article  PubMed  Google Scholar 

  109. Gariballa N, Ali BR. Endoplasmic Reticulum Associated protein Degradation (ERAD) in the pathology of diseases related to TGFβ signaling pathway: future therapeutic perspectives. Front Mol Biosci. 2020;7:575608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Luo H, Jiao Q, Shen C, et al. Unraveling the roles of endoplasmic reticulum-associated degradation in metabolic disorders. Front Endocrinol (Lausanne). 2023;14:1123769.

    Article  PubMed  Google Scholar 

  111. Grasemann H, Ratjen F. Cystic fibrosis. N Engl J Med. 2023;389:1693–707.

    Article  CAS  PubMed  Google Scholar 

  112. De Boeck K. Cystic fibrosis in the year 2020: a disease with a new face. Acta Paediatr. 2020;109:893–9.

    Article  PubMed  Google Scholar 

  113. Trouvé P, Férec C,Génin E. The interplay between the unfolded protein response, inflammation and infection in cystic fibrosis. Cells. 2021;10:2980.

  114. Santos JD, Canato S, Carvalho AS, et al. Folding status is determinant over traffic-competence in defining CFTR interactors in the endoplasmic reticulum. Cells. 2019;8:353.

  115. Bergbower E, Boinot C, Sabirzhanova I, et al. The CFTR-associated ligand arrests the trafficking of the mutant ΔF508 CFTR channel in the ER contributing to cystic fibrosis. Cell Physiol Biochem. 2018;45:639–55.

    Article  CAS  PubMed  Google Scholar 

  116. Kim Chiaw P, Huan LJ, Gagnon S, et al. Functional rescue of DeltaF508-CFTR by peptides designed to mimic sorting motifs. Chem Biol. 2009;16:520–30.

    Article  PubMed  Google Scholar 

  117. Kim Chiaw P, Eckford PD, Bear CE. Insights into the mechanisms underlying CFTR channel activity, the molecular basis for cystic fibrosis and strategies for therapy. Essays Biochem. 2011;50:233–48.

    Article  PubMed  Google Scholar 

  118. Bradley KL, Stokes CA, Marciniak SJ, et al. Role of unfolded proteins in lung disease. Thorax. 2021;76:92–9.

    Article  PubMed  Google Scholar 

  119. Izumi M, Kuruma R, Okamoto R, et al. Substrate recognition of glycoprotein folding sensor UGGT analyzed by site-specifically. J Am Chem Soc. 2017;139:11421–6.

    Article  CAS  PubMed  Google Scholar 

  120. Scheffer J, Hasenjäger S, Taxis C. Degradation of integral membrane proteins modified with the photosensitive degron module requires the cytosolic endoplasmic reticulum-associated degradation pathway. Mol Biol Cell. 2019;30:2558–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Fregno I, Molinari M. Proteasomal and lysosomal clearance of faulty secretory proteins: ER-associated degradation (ERAD) and ER-to-lysosome-associated degradation (ERLAD) pathways. Crit Rev Biochem Mol Biol. 2019;54:153–63.

    Article  CAS  PubMed  Google Scholar 

  122. Marelli S, Micaglio E, Taurino J, et al: Marfan syndrome: enhanced diagnostic tools and follow-up management strategies. Diagnostics (Basel) 2023;13.

  123. Zeyer KA, Reinhardt DP. Engineered mutations in fibrillin-1 leading to Marfan syndrome act at the protein, cellular and organismal levels. Mutat Res Rev Mutat Res. 2015;765:7–18.

    Article  CAS  PubMed  Google Scholar 

  124. Reinhardt DP, Sasaki T, Dzamba BJ, et al. Fibrillin-1 and fibulin-2 interact and are colocalized in some tissues. J Biol Chem. 1996;271:19489–96.

    Article  CAS  PubMed  Google Scholar 

  125. Galbiati F, Razani B, Lisanti MP. Caveolae and caveolin-3 in muscular dystrophy. Trends Mol Med. 2001;7:435–41.

    Article  CAS  PubMed  Google Scholar 

  126. Pradhan BS,Prószyński TJ: A Role for Caveolin-3 in the pathogenesis of muscular dystrophies. Int J Mol Sci 2020;21.

  127. Minetti C, Sotgia F, Bruno C, et al. Mutations in the caveolin-3 gene cause autosomal dominant limb-girdle muscular dystrophy. Nat Genet. 1998;18:365–8.

    Article  CAS  PubMed  Google Scholar 

  128. Galbiati F, Volonte D, Minetti C, et al. Phenotypic behavior of caveolin-3 mutations that cause autosomal dominant limb girdle muscular dystrophy (LGMD-1C). Retention of LGMD-1C caveolin-3 mutants within the golgi complex. J Biol Chem. 1999;274:25632–41.

    Article  CAS  PubMed  Google Scholar 

  129. Herrmann R, Straub V, Blank M, et al. Dissociation of the dystroglycan complex in caveolin-3-deficient limb girdle muscular dystrophy. Hum Mol Genet. 2000;9:2335–40.

    Article  CAS  PubMed  Google Scholar 

  130. Smith K, Rennie MJ. New approaches and recent results concerning human-tissue collagen synthesis. Curr Opin Clin Nutr Metab Care. 2007;10:582–90.

    Article  CAS  PubMed  Google Scholar 

  131. Fritsch A, Spassov S, Elfert S, et al. Dominant-negative effects of COL7A1 mutations can be rescued by controlled overexpression of normal collagen VII. J Biol Chem. 2009;284:30248–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Yamazaki CM, Kadoya Y, Hozumi K, et al. A collagen-mimetic triple helical supramolecule that evokes integrin-dependent cell responses. Biomaterials. 2010;31:1925–34.

    Article  CAS  PubMed  Google Scholar 

  133. Ito S, Nagata K. Quality control of procollagen in cells. Annu Rev Biochem. 2021;90:631–58.

    Article  CAS  PubMed  Google Scholar 

  134. Nyström A, Bruckner-Tuderman L, Kiritsi D. Dystrophic epidermolysis bullosa: secondary disease mechanisms and disease modifiers. Front Genet. 2021;12:737272.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Hammami-Hauasli N, Schumann H, Raghunath M, et al. Some, but not all, glycine substitution mutations in COL7A1 result in intracellular accumulation of collagen VII, loss of anchoring fibrils, and skin blistering. J Biol Chem. 1998;273:19228–34.

    Article  CAS  PubMed  Google Scholar 

  136. Bethlem J, Wijngaarden GK. Benign myopathy, with autosomal dominant inheritance. A report on three pedigrees. Brain. 1976;99:91–100.

    Article  CAS  PubMed  Google Scholar 

  137. Pan TC, Zhang RZ, Sudano DG, et al. New molecular mechanism for Ullrich congenital muscular dystrophy: a heterozygous in-frame deletion in the COL6A1 gene causes a severe phenotype. Am J Hum Genet. 2003;73:355–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Karkavelas G, Kefalides NA, Amenta PS, et al. Comparative ultrastructural localization of collagen types III, IV, VI and laminin in rat uterus and kidney. J Ultrastruct Mol Struct Res. 1988;100:137–55.

    Article  CAS  PubMed  Google Scholar 

  139. Marini JC, Forlino A, Bächinger HP, et al. Osteogenesis imperfecta. Nat Rev Dis Primers. 2017;3:17052.

    Article  PubMed  Google Scholar 

  140. Forlino A, Marini JC. Osteogenesis imperfecta: prospects for molecular therapeutics. Mol Genet Metab. 2000;71:225–32.

    Article  CAS  PubMed  Google Scholar 

  141. Jovanovic M, Guterman-Ram G, Marini JC. Osteogenesis imperfecta: mechanisms and signaling pathways connecting classical and rare OI types. Endocr Rev. 2022;43:61–90.

    Article  PubMed  Google Scholar 

  142. Mirigian LS, Makareeva E, Mertz EL, et al. Osteoblast malfunction caused by cell stress response to procollagen misfolding in α2(I)-G610C mouse model of osteogenesis imperfecta. J Bone Miner Res. 2016;31:1608–16.

    Article  CAS  PubMed  Google Scholar 

  143. Doan ND, Hosseini AS, Bikovtseva AA, et al. Elucidation of proteostasis defects caused by osteogenesis imperfecta mutations in the collagen-α2(I) C-propeptide domain. J Biol Chem. 2020;295:9959–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Ho Duy B, Zhytnik L, Maasalu K, et al. Mutation analysis of the COL1A1 and COL1A2 genes in Vietnamese patients with osteogenesis imperfecta. Hum Genomics. 2016;10:27.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Besio R, Iula G, Garibaldi N, et al. 4-PBA ameliorates cellular homeostasis in fibroblasts from osteogenesis imperfecta patients by enhancing autophagy and stimulating protein secretion. Biochim Biophys Acta Mol Basis Dis. 2018;1864:1642–52.

    Article  CAS  PubMed  Google Scholar 

  146. Feingold KR, Anawalt B, Blackman MR, et al: Endotext. 2000.

  147. Ben Amor IM, Glorieux FH, Rauch F. Genotype-phenotype correlations in autosomal dominant osteogenesis imperfecta. J Osteoporos. 2011;2011: 540178.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Chiarelli N, Ritelli M, Zoppi N, et al: Cellular and molecular mechanisms in the pathogenesis of classical, vascular, and hypermobile Ehlers‒Danlos syndromes. Genes (Basel) 2019;10.

  149. Symoens S, Syx D, Malfait F, et al. Comprehensive molecular analysis demonstrates type V collagen mutations in over 90% of patients with classic EDS and allows to refine diagnostic criteria. Hum Mutat. 2012;33:1485–93.

    Article  CAS  PubMed  Google Scholar 

  150. Malfait F, Coucke P, Symoens S, et al. The molecular basis of classic Ehlers-Danlos syndrome: a comprehensive study of biochemical and molecular findings in 48 unrelated patients. Hum Mutat. 2005;25:28–37.

    Article  CAS  PubMed  Google Scholar 

  151. Acke FR, Malfait F, Vanakker OM, et al. Novel pathogenic COL11A1/COL11A2 variants in Stickler syndrome detected by targeted NGS and exome sequencing. Mol Genet Metab. 2014;113:230–5.

    Article  CAS  PubMed  Google Scholar 

  152. Richards AJ, Fincham GS, McNinch A, et al. Alternative splicing modifies the effect of mutations in COL11A1 and results in recessive type 2 Stickler syndrome with profound hearing loss. J Med Genet. 2013;50:765–71.

    Article  CAS  PubMed  Google Scholar 

  153. Akawi NA, Al-Gazali L, Ali BR. Clinical and molecular analysis of UAE fibrochondrogenesis patients expands the phenotype and reveals two COL11A1 homozygous null mutations. Clin Genet. 2012;82:147–56.

    Article  CAS  PubMed  Google Scholar 

  154. Akawi NA, Ali BR, Al-Gazali L. A response to Dr. Alzahrani’s letter to the editor regarding the mechanism underlying fibrochondrogenesis. Gene. 2013;528:367–8.

    Article  CAS  PubMed  Google Scholar 

  155. Faughnan ME, Mager JJ, Hetts SW, et al. Second international guidelines for the diagnosis and management of hereditary hemorrhagic telangiectasia. Ann Intern Med. 2020;173:989–1001.

    Article  PubMed  Google Scholar 

  156. Goldman LA, Cutrone EC, Kotenko SV, et al. Modifications of vectors pEF-BOS, pcDNA1 and pcDNA3 result in improved convenience and expression. Biotechniques. 1996;21:1013–5.

    Article  CAS  PubMed  Google Scholar 

  157. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8:345–51.

    Article  CAS  PubMed  Google Scholar 

  158. Ruiz-Llorente L, Gallardo-Vara E, Rossi E, et al. Endoglin and alk1 as therapeutic targets for hereditary hemorrhagic telangiectasia. Expert Opin Ther Targets. 2017;21:933–47.

    Article  CAS  PubMed  Google Scholar 

  159. Gallione CJ, Repetto GM, Legius E, et al. A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet. 2004;363:852–9.

    Article  CAS  PubMed  Google Scholar 

  160. Caja L, Dituri F, Mancarella S, et al: TGF-β and the tissue microenvironment: relevance in fibrosis and cancer. Int J Mol Sci 2018;19.

  161. Goumans MJ,Ten Dijke P: TGF-β Signaling in control of cardiovascular function. Cold Spring Harb Perspect Biol 2018;10.

  162. Castonguay R, Werner ED, Matthews RG, et al. Soluble endoglin specifically binds bone morphogenetic proteins 9 and 10 via its orphan domain, inhibits blood vessel formation, and suppresses tumor growth. J Biol Chem. 2011;286:30034–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Gariballa N, Kizhakkedath P, Akawi N, et al. Endoglin Wild Type and Variants Associated With Hereditary Hemorrhagic Telangiectasia Type 1 Undergo Distinct Cellular Degradation Pathways. Front Mol Biosci. 2022;9: 828199.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Mallet C, Lamribet K, Giraud S, et al. Functional analysis of endoglin mutations from hereditary hemorrhagic telangiectasia type 1 patients reveals different mechanisms for endoglin loss of function. Hum Mol Genet. 2015;24:1142–54.

    Article  CAS  PubMed  Google Scholar 

  165. Förg T, Hafner M, Lux A. Investigation of endoglin wild-type and missense mutant protein heterodimerisation using fluorescence microscopy based IF BiFC and FRET analyses. PLoS One. 2014;9:e102998.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13:189–95.

    Article  CAS  PubMed  Google Scholar 

  167. Vorselaars VMM, Hosman AE, Westermann CJJ, et al: Pulmonary arterial hypertension and hereditary haemorrhagic telangiectasia. Int J Mol Sci 2018;19.

  168. Ricard N, Bidart M, Mallet C, et al. Functional analysis of the BMP9 response of ALK1 mutants from HHT2 patients: a diagnostic tool for novel ACVRL1 mutations. Blood. 2010;116:1604–12.

    Article  CAS  PubMed  Google Scholar 

  169. Lenato GM, Guanti G. Hereditary Haemorrhagic Telangiectasia (HHT): genetic and molecular aspects. Curr Pharm Des. 2006;12:1173–93.

    Article  CAS  PubMed  Google Scholar 

  170. Morrell NW, Aldred MA, Chung WK, et al: Genetics and genomics of pulmonary arterial hypertension. Eur Respir J 2019;53.

  171. Dunmore BJ, Jones RJ, Toshner MR, et al. Approaches to treat pulmonary arterial hypertension by targeting BMPR2: from cell membrane to nucleus. Cardiovasc Res. 2021;117:2309–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Frump AL, Lowery JW, Hamid R, et al. Abnormal trafficking of endogenously expressed BMPR2 mutant allelic products in patients with heritable pulmonary arterial hypertension. PLoS One. 2013;8:e80319.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Loeys BL, Chen J, Neptune ER, et al. A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in TGFBR1 or TGFBR2. Nat Genet. 2005;37:275–81.

    Article  CAS  PubMed  Google Scholar 

  174. Camerota L, Ritelli M, Wischmeijer A, et al: Genotypic categorization of Loeys-Dietz syndrome based on 24 novel families and literature data. Genes (Basel) 2019;10.

  175. Schepers D, Tortora G, Morisaki H, et al. A mutation update on the LDS-associated genes TGFB2/3 and SMAD2/3. Hum Mutat. 2018;39:621–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Crotti L, Celano G, Dagradi F, et al. Congenital long QT syndrome. Orphanet J Rare Dis. 2008;3:18.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Smith JL, Anderson CL, Burgess DE, et al. Molecular pathogenesis of long QT syndrome type 2. J Arrhythm. 2016;32:373–80.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Schwartz PJ, Ackerman MJ, George AL, et al. Impact of genetics on the clinical management of channelopathies. J Am Coll Cardiol. 2013;62:169–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Curran ME, Splawski I, Timothy KW, et al. A molecular basis for cardiac arrhythmia: HERG mutations cause long QT syndrome. Cell. 1995;80:795–803.

    Article  CAS  PubMed  Google Scholar 

  180. Cox KO, Wang BX. Long QT syndrome type 2: mechanism-based therapies. Future Cardiol. 2021;17:1453–63.

    Article  CAS  PubMed  Google Scholar 

  181. Hayashi K, Shimizu M, Ino H, et al. Characterization of a novel missense mutation E637K in the pore-S6 loop of HERG in a patient with long QT syndrome. Cardiovasc Res. 2002;54:67–76.

    Article  CAS  PubMed  Google Scholar 

  182. Bianchi MT, Song L, Zhang H, et al. Two different mechanisms of disinhibition produced by GABAA receptor mutations linked to epilepsy in humans. J Neurosci. 2002;22:5321–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Zhu S, Sridhar A, Teng J, et al. Structural and dynamic mechanisms of GABA. Nat Commun. 2022;13:4582.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Sherekar M, Han SW, Ghirlando R, et al. Biochemical and structural analyses reveal that the tumor suppressor neurofibromin (NF1) forms a high-affinity dimer. J Biol Chem. 2020;295:1105–19.

    Article  PubMed  Google Scholar 

  185. Buscarini E, Botella LM, Geisthoff U, et al. Safety of thalidomide and bevacizumab in patients with hereditary hemorrhagic telangiectasia. Orphanet J Rare Dis. 2019;14:28.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Waters MF, Fee D, Figueroa KP, et al. An autosomal dominant ataxia maps to 19q13: Allelic heterogeneity of SCA13 or novel locus? Neurology. 2005;65:1111–3.

    Article  CAS  PubMed  Google Scholar 

  187. Klein C, Breakefield XO, Ozelius LJ. Genetics of primary dystonia. Semin Neurol. 1999;19:271–80.

    Article  CAS  PubMed  Google Scholar 

  188. Hewett J, Gonzalez-Agosti C, Slater D, et al. Mutant torsinA, responsible for early-onset torsion dystonia, forms membrane inclusions in cultured neural cells. Hum Mol Genet. 2000;9:1403–13.

    Article  CAS  PubMed  Google Scholar 

  189. Broadgate S, Yu J, Downes SM, et al. Unravelling the genetics of inherited retinal dystrophies: past, present and future. Prog Retin Eye Res. 2017;59:53–96.

    Article  CAS  PubMed  Google Scholar 

  190. Jastrzebska B, Chen Y, Orban T, et al. Disruption of rhodopsin dimerization with synthetic peptides targeting an interaction interface. J Biol Chem. 2015;290:25728–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90:262–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Morikawa S, Tanabe K, Kaneko N, et al. Comprehensive overview of disease models for Wolfram syndrome: toward effective treatments. Mamm Genome. 2024;35:1–12.

    Article  PubMed  Google Scholar 

  193. Morikawa S, Tajima T, Nakamura A, et al. A novel heterozygous mutation of the WFS1 gene leading to constitutive endoplasmic reticulum stress is the cause of Wolfram syndrome. Pediatr Diabetes. 2017;18:934–41.

    Article  CAS  PubMed  Google Scholar 

  194. De Franco E, Flanagan SE, Yagi T, et al. Dominant ER stress-inducing. Diabetes. 2017;66:2044–53.

    PubMed  PubMed Central  Google Scholar 

  195. Irving JA, Pike RN, Lesk AM, et al. Phylogeny of the serpin superfamily: implications of patterns of amino acid conservation for structure and function. Genome Res. 2000;10:1845–64.

    Article  CAS  PubMed  Google Scholar 

  196. Law RH, Zhang Q, McGowan S, et al. An overview of the serpin superfamily. Genome Biol. 2006;7:216.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Carvill GL, Matheny T, Hesselberth J, et al. Haploinsufficiency, dominant negative, and gain-of-function mechanisms in epilepsy: matching therapeutic approach to the pathophysiology. Neurotherapeutics. 2021;18:1500–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Price BA, Sandoval IM, Chan F, et al. Rhodopsin gene expression determines rod outer segment size and rod cell resistance to a dominant-negative neurodegeneration mutant. PLoS One. 2012;7:e49889.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Mao H, James T, Schwein A, et al. AAV delivery of wild-type rhodopsin preserves retinal function in a mouse model of autosomal dominant retinitis pigmentosa. Hum Gene Ther. 2011;22:567–75.

    Article  CAS  PubMed  Google Scholar 

  200. Garofalo S, Quarto R. Knocking out the bad allele. Gene Ther. 2004;11:1301–2.

    Article  CAS  PubMed  Google Scholar 

  201. Chamberlain JR, Deyle DR, Schwarze U, et al. Gene targeting of mutant COL1A2 alleles in mesenchymal stem cells from individuals with osteogenesis imperfecta. Mol Ther. 2008;16:187–93.

    Article  CAS  PubMed  Google Scholar 

  202. Gonzalez-Alegre P, Miller VM, Davidson BL, et al. Toward therapy for DYT1 dystonia: allele-specific silencing of mutant TorsinA. Ann Neurol. 2003;53:781–7.

    Article  CAS  PubMed  Google Scholar 

  203. Dotzler SM, Kim CSJ, Gendron WAC, et al. Suppression-Replacement. Circulation. 2021;143:1411–25.

    Article  CAS  PubMed  Google Scholar 

  204. Cring MR, Sheffield VC. Gene therapy and gene correction: targets, progress, and challenges for treating human diseases. Gene Ther. 2022;29:3–12.

    Article  CAS  PubMed  Google Scholar 

  205. Jung H, Rim YA, Park N, et al: Restoration of Osteogenesis by CRISPR/Cas9 Genome Editing of the Mutated. J Clin Med 2021; 10.

  206. Rees HA, Liu DR. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. 2018;19:770–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Anderson J, Maina N. Reviewing clinical considerations and guideline recommendations of C1 inhibitor prophylaxis for hereditary angioedema. Clin Transl Allergy. 2022;12:e12092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Mohamed FE, Al-Gazali L, Al-Jasmi F, et al. Pharmaceutical chaperones and proteostasis regulators in the therapy of lysosomal storage disorders: current perspective and future promises. Front Pharmacol. 2017;8:448.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Noorwez SM, Malhotra R, McDowell JH, et al. Retinoids assist the cellular folding of the autosomal dominant retinitis pigmentosa opsin mutant P23H. J Biol Chem. 2004;279:16278–84.

    Article  CAS  PubMed  Google Scholar 

  210. Brothers SP, Cornea A, Janovick JA, et al. Human loss-of-function gonadotropin-releasing hormone receptor mutants retain wild-type receptors in the endoplasmic reticulum: molecular basis of the dominant-negative effect. Mol Endocrinol. 2004;18:1787–97.

    Article  CAS  PubMed  Google Scholar 

  211. Cortez L, Sim V. The therapeutic potential of chemical chaperones in protein folding diseases. Prion. 2014;8:197–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Wu KY, Kulbay M, Toameh D, et al: Retinitis pigmentosa: novel therapeutic targets and drug development. Pharmaceutics 2023;15.

  213. Ranke MB. Treatment of children and adolescents with idiopathic short stature. Nat Rev Endocrinol. 2013;9:325–34.

    Article  CAS  PubMed  Google Scholar 

  214. Stavber L, Gaia MJ, Hovnik T, et al: Heterozygous. Genes (Basel) 2022;13.

  215. Merlini L, Sabatelli P, Armaroli A, et al. Cyclosporine A in Ullrich congenital muscular dystrophy: long-term results. Oxid Med Cell Longev. 2011;2011:139194.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by ASPIRE, the technology program management pillar of Abu Dhabi’s Advanced Technology Research Council (ATRC), via individual grant AARE19-086 and the ASPIRE Precision Medicine Research Institute grant VRI-20–10.

Author information

Authors and Affiliations

Authors

Contributions

BRA conceptualized the review, supervised the overall direction and edited all version of the manuscript. NG, FM and SB reviewed the literature, collected the data and wrote the various drafts of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bassam R. Ali.

Ethics declarations

Ethics approval and consent to participate

The manuscript doesn’t include data or description of human patients or animal experiemnts and therefore does not require ethical committee approval at this institution.

Consent for publication

Not applicable.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gariballa, N., Mohamed, F., Badawi, S. et al. The double whammy of ER-retention and dominant-negative effects in numerous autosomal dominant diseases: significance in disease mechanisms and therapy. J Biomed Sci 31, 64 (2024). https://doi.org/10.1186/s12929-024-01054-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-024-01054-1

Keywords