Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Dec 1.
Published in final edited form as: Semin Fetal Neonatal Med. 2023 Nov 22;28(6):101500. doi: 10.1016/j.siny.2023.101500

Towards personalized therapies for genetic disorders of surfactant dysfunction

Maureen Peers de Nieuwburgh 1, Jennifer A Wambach 2, Matthias Griese 3, Olivier Danhaive 1,4
PMCID: PMC10753445  NIHMSID: NIHMS1950447  PMID: 38036307

Abstract

Genetic disorders of surfactant dysfunction are a rare cause of chronic, progressive or refractory respiratory failure in term and preterm infants. This review explores genetic mechanisms underpinning surfactant dysfunction, highlighting specific surfactant-associated genes including SFTPB, SFTPC, ABCA3, and NKX2.1. Pathogenic variants in these genes contribute to a range of clinical presentations and courses, from neonatal hypoxemic respiratory failure to childhood interstitial lung disease and even adult-onset pulmonary fibrosis. This review emphasizes the importance of early recognition, thorough phenotype assessment, and assessment of variant functionality as essential prerequisites for treatments including lung transplantation. We explore emerging treatment options, including personalized pharmacological approaches and gene therapy strategies. In conclusion, this comprehensive review offers valuable insights into the pathogenic mechanisms of genetic disorders of surfactant dysfunction, genetic fundamentals, available and emerging therapeutic options, and underscores the need for further research to develop personalized therapies for affected infants and children.

Keywords: Surfactant, genetic deficiency, gene therapy, lung transplant, neonates, infants

Introduction

Respiratory distress syndrome (RDS) is the consequence of surfactant insufficiency, resulting in increased alveolar surface tension and airspace collapse. In premature infants, RDS primarily arises due to alveolar epithelium immaturity and insufficient surfactant production1. However, regardless of gestational age, chronic, progressive, or refractory RDS may result from genetic diseases of surfactant dysfunction24. Pathogenic variants in several genes, pivotal for maintaining surfactant homeostasis and function, have been identified as rare yet significant contributors to a wide spectrum of chronic lung diseases, ranging from lethal hypoxemic respiratory failure (HRF) in term and preterm neonates to childhood interstitial lung disease (chILD) and even adult-onset pulmonary fibrosis. Despite the current lack of effective therapeutic options, several pre-clinical studies are delineating potential strategies for personalized pharmacologic or genetic treatments. Early recognition of the phenotype, identification of the molecular etiology, and variant functional characterization are important steps to facilitate targeted interventions.

In this review, we aim to provide clinicians with an overview of the current knowledge on disease mechanisms, state-of-the-art management, and future therapies targeting molecular defects.

Surfactant structure, function, and metabolism

Pulmonary surfactant, composed of phospholipids (80%), neutral lipids (10%), and proteins (10%)5, lowers surface tension in the lung preventing atelectasis at end-expiration6, and acts as an innate immunity mechanism7. Pulmonary surfactant is assembled and stored within the lamellar bodies of alveolar type 2 pneumocytes (AEC2)8. Surfactant is secreted from the apical pole as tubular myelin9, then spreads at the air-liquid interface of the alveolar epithelium, forming a dynamic monolayer expanding and contracting over repeated breathing cycles, optimizing surface tension to prevent alveolar collapse and/or overdistension8. Intra-alveolar surfactant homeostasis is maintained by net contributions from synthesis, secretion, uptake, and catabolism by AEC2s and alveolar macrophages5, 10. Figure 1 summarizes the surfactant pathway from gene expression to surfactant assembly, secretion, and degradation.

Figure 1:

Figure 1:

Genetic disorders of surfactant dysfunction: current and emerging treatments

Surfactant proteins B and C (SP-B, SP-C), encoded by the genes SFTPB and SFTPC, are extremely hydrophobic proteins and contribute to the surface tension lowering qualities of surfactant. SP-B is critical for surfactant packaging, adsorption, and surface tension-lowering properties11, 12. SP-C plays a less critical role at birth, contributing to surface film formation and stability, especially at alveolar near-collapse13, 14.

In addition to surfactant proteins, surfactant lipids are critical for surface activity and stability15. ATP-Binding cassette transporter A3 (ABCA3) is a lamellar body transmembrane phospholipid transporter essential for organelle biogenesis, surfactant protein-phospholipid assembly, and alveolar homeostasis16. Thyroid transcription factor 1 (TTF–1), encoded by NKX2.1, plays a central role in lung morphogenesis, epithelial differentiation, and up-regulation of most surfactant genes (SFTPA, SFTPB, SFTPC, SFTPD, ABCA3), in addition to development of the thyroid gland and central nervous system. Finally, surfactant clearance by alveolar macrophages is also a critical step in alveolar homeostasis, for which Granulocyte macrophage-colony stimulating factor (GM-CSF) signaling plays a central role17.

Genetic diseases of the surfactant system

1). Surfactant protein B deficiency

SP-B deficiency is an autosomal recessive disease and occurs ~1 per million births18. Infants typically present with neonatal HRF and die within the first months of life without lung transplantation4, 19. Surfactant replacement has only transient effects on oxygenation and ventilation and does not result in prolonged survival20. Most affected infants have recessively inherited biallelic null variants of SFTPB leading to complete or near complete loss of mature SP-B expression4, 21. The frameshift variant p.Pro133Glnfs*95 [c.361delinsGAA;“121ins2”] is identified in over 70% of affected infants21. This variant introduces a premature stop codon resulting in undetectable levels of SP-B in lung tissue and alveolar fluid, as well as proSP-C misprocessing22. In rare instances, prolonged survival beyond the first months or years has been associated with hypomorphic missense variants resulting in some residual SP-B expression and function23, 24.

2). Surfactant protein C-associated lung disease

SFTPC pathogenic variants are associated with a wide spectrum of clinical presentations, ranging from lethal hypoxemic respiratory failure in neonates25 to, most typically, chILD and adult-onset pulmonary fibrosis26, 27. SP-C -associated lung disease is caused by monoallelic usually missense variants, about half of which are dominantly inherited with variable penetrance, and half arise de novo. Surfactant dysfunction results from dominant-negative or toxic gain-of-function mechanisms. The distal C-terminus sequence of the apoprotein contains a crucial BRICHOS domain which functions as a chaperone to prevent aggregation of the highly hydrophobic mature peptide during post-transcriptional processes. BRICHOS variants lead to misfolding, activation of the unfolded protein response, and cell death25, 28. Pathogenic variants in the apoprotein linker sequence, including the most frequent p.Ile73Thr, cause abnormal intracytoplasmic trafficking and accumulation of unprocessed pro-peptide fragments in the plasma membrane and endosomes25, 29. Missense variants affecting the mature peptide domain, as well as splicing variants and large copy number variants are only rarely reported30.

3). ATP-Binding cassette A3 deficiency

ABCA3 deficiency is the most frequent among the genetic diseases of surfactant dysfunction, typically presenting as neonatal HRF similar to SP-B deficiency, or less commonly as chILD frequently evolving toward end-stage lung disease in the first months or years of life3133. ABCA3 deficiency is an autosomal recessive disease and results from biallelic variants of ABCA3. Over 300 disease-associated variants including nonsense, frameshift, missense, splice site, and small insertion/deletion (‘indel’) variants have been reported to date3436. ABCA3 missense variants may affect ABCA3 trafficking to the lamellar body membrane (class I) or may disrupt ATP hydrolysis and lipid transport (class II). As most affected patients are compound heterozygous, and ABCA3 variants are often private, disease severity is difficult to predict and will depend on the combined residual function resulting from the two variant alleles34, 35.

Single ABCA3 variants, may result in reversible RDS among term and late preterm infants3739. Some infants have been identified to carry only a single ABCA3 variant, but express the full ABCA3 deficiency phenotype, suggesting non-coding variants of the opposite allele, which may not be detected with exon-based sequencing, may alter gene expression. Uniparental disomy has been occasionally described as a potential disease mechanism40, with a low risk of recurrence in future pregnancies. The recurrent deep intronic variant IVS25–98T, resulting in aberrant splicing, was identified in several affected infants in whom only one heterozygous coding variant had been found41. Large intragenic deletions spanning entire exons undetectable by Sanger sequencing have also been reported36. In addition, heterozygous variants in ABCA3 may modify disease presentation or severity among individuals with SFTPC variants42.

4). Thyroid transcription factor 1 associated lung disease

TTF-1 is a small protein expressed in the lung, thyroid, basal ganglia, and hypothalamus, and plays multiple roles in organ formation and function43. TTF-1 -associated lung disease combines features of surfactant dysfunction and impaired alveolarization44. Monoallelic NKX2.1 variants may be associated with the brain-lung-thyroid syndrome, a complex phenotype that includes various combinations and degrees of hypothyroidism, benign hereditary chorea, acinar dysplasia, neonatal HRF, and chILD45, 46. Single nucleotide, small indels and large copy number variants, dominantly inherited or arising de novo, have been reported. Penetrance varies widely, and individuals with the same variant may display varying degrees of neurologic abnormalities, thyroid disease, and/or pulmonary disease, thus making it challenging to predict their individual clinical progression.

5). Other surfactant-related genetic diseases

Rare variants in additional genes have been described in infants with clinical phenotypes of surfactant dysfunction. Variants of the GM-CSF receptors subunits A (CSF2RA)47 and B (CSF2RB)48, 49 manifest as pulmonary alveolar proteinosis (PAP), albeit presenting in late infancy or childhood. Methionyl-tRNA synthetase-1 (MARS1) variants are another ultrarare cause of early-onset PAP associated with liver disease, most frequently identified among individuals from Reunion island50. A RAB5B variant disrupting SP-B and -C intracytoplasmic processing was recently identified in an infant with fatal ILD, neurodevelopmental delay, and dysmorphic features51. Filamin A (FLNA) variants cause a severe form of chILD that may present in the neonatal period, typically associated with pulmonary hypertension, dysmorphic features, and periventricular nodular heterotopia52. Variants in SFTPA1 and SFTPA2 are associated with adult-onset ILD and lung cancer53. These diseases have distinctive features, but their prevalence and potential role in infant and childhood surfactant dysfunction disorders have not been thoroughly explored.

Figure 2 shows radiographic phenotypes of several genetic diseases of surfactant dysfunction.

Figure 2. Clinical evolution of surfactant dysfunction disease.

Figure 2.

A. Chest radiograph in a term infant on day 14 of life with ABCA3 deficiency showing mild diffuse hazy granular airspace opacification resembling the findings of respiratory distress syndrome of prematurity. – B and C. Same infant’s chest radiograph at 2 (B) and 4 (C) months, showing progressive airspace coarsening and septal thickening. – D. Chest CT of a 4-month-old infant with ABCA3 deficiency showing ground-glass opacity, patchy cystic areas, and peribronchial and interlobar thickening. – E. Chest CT of a 6-month-old infant with an SFTPC variant with mild chronic lung disease showing mild interstitial thickening and small subpleural and lobular cysts in clusters. – F. Chest CT of an infant with NKX2.1 variant at 1 month of life, showing diffuse ground glass attenuation and mild septal thickening.

Treatment approaches for genetic disorder of surfactant dysfunction

Table 1 summarizes the genetic mechanisms and histopathology phenotypes associated with pathogenic variants in SFTPB, SFTPC, ABCA3, and NKX2.1 as well as potential treatment options. Infants with SP-B or ABCA3 deficiency due to biallelic loss-of-function variants present with neonatal respiratory failure that is usually progressive despite pharmacological interventions19, 21, 34, 35. The clinical courses of infants and children with pathogenic variants in SFTPC and NKX2–1 as well as missense variants in ABCA3 are more variable and difficult to predict. Lower respiratory tract infections and ILD exacerbations may trigger long-term deteriorations of the clinical course54.

Table 1.

Summary of surfactant deficiencies

Surfactant protein B Surfactant protein C ATP-binding cassette A3 Thyroid transcription factor 1
Estimated incidence 1/1,000,000 Unknown 1/90,000 births Unknown
Gene SFTPB SFTPC ABCA3 NKX2.1
Genomic position 2p11.2 8p21.3 16p13.3 14q13
Heritability Autosomal recessive with complete penetrance

Mostly inherited
Autosomal dominant with variable penetrance (~50%)

Inherited or de novo (~50%)
Autosomal recessive

Mostly inherited
• Autosomal dominant with variable penetrance

• Inherited or de novo
Molecular genetics >40 SNVs: most frequently null variant p.Pro133Glnfs*95 (“121ins2”) (~70%) >60 SNVs:

LINKER propeptide domain; most frequent variant: p.Ile73Thr (25%)

BRICHOS propeptide domain

Rarely, mature peptide Rare large CNV
>300 SNVs, mostly private:

Null variants

Most common pathogenic variant: p.Glu292Val

Rare large CNV
>80 variants:

SNVs: null and hypomorphic variants mostly in the DNA-binding homeodomain

CNV (~15%)
Disease mechanisms Loss of SP-B expression Disrupted intracellular trafficking (LINKER variants)

Protein misfolding, endoplasmic reticulum stress (BRICHOS variants)
Loss of ABCA3 expression

Disrupted phospholipid transport Disrupted intracellular trafficking
Decreased expression of surfactant-related genes (SFTPA, SFTPB, SFTPC, ABCA3) Altered lung development (branching and differentiation)
Clinical presentations Neonatal hypoxemic respiratory failure (most)

Childhood chronic lung disease (very rare)
Neonatal hypoxemic respiratory failure (~10%)

Infantile (~40%) and childhood ILD

Recurrent respiratory infections

Adult idiopathic pulmonary fibrosis
Neonatal hypoxemic respiratory failure

Infantile ILD
Neonatal hypoxemic respiratory failure

Childhood ILD

Hypothyroidism

Benign familial chorea, hypotonia

Lung, thyroid, and blood cancers
Current therapies Early transplant, neonatal or infancy Hydroxychloroquine

Azithromycin

Steroids

Infant/childhood

transplant
Steroids

Hydroxychloroquine

Azithromycin

Infant/childhood

transplant
Steroids

Hydroxychloroquine

Azithromycin

1). Nonspecific pharmacological approaches

Medical therapies for genetic diseases of surfactant dysfunction remain limited and nonspecific35, 55. Affected infants and children primarily receive supportive care, including oxygen supplementation, chronic ventilation, and nutritional supplementation. Current pharmacologic treatments are used without distinction based on the underlying molecular mechanism and include systemic glucocorticoids, hydroxychloroquine, and azithromycin. Exogenous surfactant has been used in infants with neonatal HRF and may bring some transient improvement in oxygenation and ventilation. These empiric treatments can be given alone or in combination, and response varies with the patient’s age, the presence of inflammatory or fibrotic lung disease, and the molecular diagnosis55. Monitoring treatment response is crucial and involves iterative assessment of respiratory function, activity, growth, and development at baseline and during follow up56.

In a recent European expert opinion group Delphi review, the consensus was that infants with chILD who are ventilated or at risk of requiring ventilation show a response to high-dose glucocorticoid “pulse therapy” (10 mg/kg methylprednisolone once daily for 3 days) within 12 to 48 hours, reaching full effect within a week57. In contrast, hydroxychloroquine may take 2–4 weeks to demonstrate noticeable effects. Non-ventilated children typically may show slower clinical responses, requiring up to 4 weeks for glucocorticoids, 12 weeks for hydroxychloroquine or azithromycin in order to assess treatment effects. However, the available literature is hampered by limited data and significant publication bias, especially regarding azithromycin. Clinical practices and protocols rely heavily on small case series and expert opinions, calling for a nuanced interpretation of findings58, 59. Within these limitations, studies have suggested some efficacy in subsets of patients with SP-C-related disease55 and, more limitedly, with ABCA3 deficiency60. Recently, a first double-blinded, randomized, placebo-controlled trial of hydroxychloroquine in chILD that included, among others, 15 children with a genetic disease of surfactant dysfunction did not show significant oxygen level improvement with treatment59. However, this study was limited by a relatively small sample size and lacked lung function testing in children under 6 years old, warranting caution in interpreting the potential benefits of hydroxychloroquine in this group. Altogether, these treatments, albeit based on anecdotal evidence, are applied empirically, alone or combined, in seriously or chronically sick children by various clinicians, and remain the mainstay of current pharmacological therapy for chILD.

2). Prevention of lung fibrosis

Surviving children and adolescents with genetic surfactant dysfunction diseases may develop pulmonary fibrosis and chronic, progressive respiratory failure. While pirfenidone and nintedanib are approved for adults with fibrosing interstitial lung disease (ILD), their use in children is not established61. A recent randomized trial of nintedanib for 24 weeks in 39 patients (age 6–17 years) with fibrosing chILD, including 12 patients with genetic surfactant dysfunction diseases, yielded favorable safety results, but efficacy assessment was not the primary focus61.

3). Lung transplant

Lung transplantation has been performed since the 1990s for progressive respiratory failure among infants and children with genetic surfactant dysfunction diseases. Evaluation for lung transplantation relies strongly on genetic testing, and advances in next-generation sequencing (gene panels, exome, or whole genome), allowing fast precision diagnosis, has reduced the need for lung biopsy62. Genetic results must be interpreted carefully in collaboration with pediatric pulmonologists and clinical geneticists, as some genetic surfactant dysfunction diseases may not necessarily be lethal without lung transplant (i.e., pathogenic variants in SFTPC and NKX2-1, or missense variants in ABCA3). Parental testing may be required in order to determine whether SFTPB or ABCA3 variants are inherited in trans (both alleles affected) or in cis (single allele affected)34.

Pathogenic or likely pathogenic variants may be identified based on the predicted effects on protein function (e.g., nonsense, frameshift variants) or previous identification in unrelated symptomatic individuals34, 60. For conditions like ABCA3 deficiency, the majority of disease-associated variants are missense variants, often unique to an affected individual or family (“private”), thus are often categorized as variants of uncertain significance (VUS)34, 60. Interpretation of VUS remains a significant challenge to the field of medical genetics. When genetic testing results are inconclusive or incongruent with the patient’s clinical course, specialists may pursue lung biopsy, which carries risk for morbidities including air leak and bleeding.

Parental education is crucial when considering lung transplantation for infants and children, as it comes with significant early and late complications. Despite advances in diagnosis and treatment, the 5-year survival rate for lung transplants remains approximately 60% for infants and 80% for children19, 63. Immediate post-transplant complications may include bleeding, anastomosis, or vascular issues, early graft dysfunction, and infections. Within the first 3 months, complications can involve rejection (T-cell or antibody-mediated) and viral or bacterial infections. Late complications, beyond 3 months, include chronic lung allograft dysfunction (bronchiolitis obliterans syndrome and restrictive allograft syndrome)64, along with hypertension, renal insufficiency, post-transplant lymphoproliferative disorder, and diabetes19. Speech and motor developmental delays and hearing loss are especially common among those transplanted during infancy, likely reflecting the severity of disease at time of transplant19. Outcomes for infants and children with genetic surfactant dysfunction diseases are comparable to those who receive lung transplants for other indications65. Nevertheless, due to the chronic, complex medical and surgical burden of pediatric lung transplantation, approximately half of families ultimately pursue comfort care over transplantation66.

4). Targeted pharmacological therapies

Efforts are underway to develop variant-specific small molecules to treat genetic diseases of surfactant dysfunction, exemplified by the success in cystic fibrosis67, 68. These efforts involve several steps: expressing the gene of interest with specific variants in a cellular model4, establishing relevant functional assessments for these variants, screening libraries of small molecules to correct their functional consequences4, optimizing compounds for safety and efficacy, and conducting in vivo studies and clinical trials to assess drug toxicity, pharmacokinetics, pharmacodynamics, and efficacy.

Hydroxychloroquine, empirically used in chILD, recently showed a efficacy for some ABCA3 variants69. In vitro results showed varying degrees of improvement in ABCA3 proteolytic cleavage, ABCA3-positive vesicle volume, and phospholipid transport for different ABCA3 mutants (E292V, D953H, Q1045R, A1046E), suggesting variant-specific effects69. This variant-specific hydroxychloroquine response demonstrated in vitro was in retrospect corroborated with clinical responses and outcomes in patients carrying these same variants.

ABCA3 shares common structural and functional features with other ABC family transporters, in particular ABCC7, also known as cystic fibrosis transmembrane conductance regulator (CFTR), for which pharmacological therapies specific to the underlying molecular defects have been extensively studied and are now approved for the treatment of patients with cystic fibrosis67, 68. Compounds enhancing channel function are designated as potentiators. Chemical or pharmacological chaperones restoring folding and trafficking, thereby increasing the amount of protein available at the plasma membrane, are designated as correctors. Several ABCA3 Class II (phospholipid transport) mutants transfected in a cell line model were functionally rescued with the CFTR potentiators ivacaftor and genistein70. Bithiazole correctors C13 and C17, and the chemical chaperone trimethylamine N-oxide showed promise in correcting some class I (trafficking) mutants in vitro71. These results point towards the possibility that clinically approved CFTR correctors and potentiators may be repurposed for some individuals with ABCA3 deficiency, depending on genotype.

A high-throughput screening platform identified Cyclosporine A as a potential corrector for specific ABCA3 mutants72. While these studies showed improved proteolytic processing and phosphatidylcholine uptake into ABCA3 positive vesicles, they did not assess actual secretion, composition, and biophysical activity of the surfactant.

In desperate clinical situations, these repurposed drugs with known safety profiles like hydroxychloroquine, cyclosporine, or the CFTR modulators may be considered in an attempt to improve clinical course. However, prospective randomized trials are most desirable and need to be implemented with innovative designs.

5). Gene therapy

There have been major advances in gene addition and genome editing strategies that have been applied to human cell lines and model organisms to characterize and even rescue surfactant deficiency or dysfunction. While these discoveries hold tremendous promise for treatment of genetic surfactant diseases, the approaches will likely be gene-specific due to differences in pathogenic mechanisms, gene size, and associated phenotypes.

These advances have brought hopes of correcting gene variations by replacing or repairing the gene to correct cellular and organ function. Extensive research7376 is focused on developing delivery systems (“vectors”) for DNA, antisense oligonucleotides, peptides, long noncoding RNA (lncRNA), or small interfering RNA (siRNA) to target specific organs and cell types involved in disease. Adenoviruses (AV) and adeno-associated viruses (AAV), are widely studied viral vectors. AV is versatile, efficient, and can carry large transgenes but may trigger inflammation and immune clearance, while AAV is less inflammatory but has limitations in gene size and remains transient due to lack of transfection efficiency. Several lentiviruses and retroviruses can insert DNA into human chromosomes, providing a long-term correction strategy. In addition to viral vectors, nanoparticles can also deliver therapeutic cargoes, improving targeting and transfection, though less efficiently and durably than viral vectors18.

In loss-of-function diseases like SP-B and ABCA3 deficiencies, gene therapy aims to restore native gene expression efficiently and persistently. Gene addition, involving the introduction of a full-length functional coding sequence, is a potential strategy. SFTPB has a relatively short coding sequence which makes it amenable to several types of vectors, including AAV which has been approved for clinical use for other indications73. Intratracheal administration of a lung-tropic AAV vector delivering murine or human SFTPB cDNA into SP-B deficient mice restored surfactant homeostasis, prevented lung injury, and improved lung physiology and survival77. A lentiviral vector expressing SP-B was used to rescue an in vitro model of SP-B deficiency in human alveolar epithelial cells78. In a human lung organoid model derived from patient-specific induced pluripotent stem cells, reversal of the SP-B deficiency phenotype was obtained by gene addition using a lentivirus carrying the wild type SFTPB cDNA79.

For ABCA3, the longer coding sequence may require the use of different vectors allowing larger cargoes. Lentiviral vectors like rSIV.F/HN showed promise in CFTR gene therapy80 and could be applied in the future for ABCA3 gene therapy.

SP-C-associated ILD, a dominantly inherited gain-of-function disease, requires a different approach. Gene knockout/knockdown, RNA interference, and antisense RNA can decrease or silence mutated SFTPC peptide expression. CRISPR/Cas9 nuclease-mediated gene knockout has shown success in mice81. AAV-packaged CRISPR/Cas9-based tools may repair individual missense variants18 offering a personalized approach for surfactant-related gene variants, but require precise diagnosis and functional studies.

Tools to study genetic diseases of surfactant dysfunction

Translational research needs to model various aspects of surfactant-related physiology, pathology and therapy in order to develop personalized therapies in human. Mutagenesis techniques provide rapid and reproducible assays for functional testing of individual variants in commercially available cell lines. Induced pluripotent stem cells from affected infants allow for more accurate modeling of individual disease without the need for genetic manipulation. Finally, animal models represent the ultimate preclinical step for testing targeted therapies.

1). Cell transfection and transduction for in vitro modelling

Human cell lines, including A549, H441, and HEK293 cells derived from pulmonary adenocarcinoma and embryonic kidney, have been used to model genetic diseases of surfactant dysfunction in vitro through techniques such as transfection (stable or transient) or viral transduction.39, 8285. These cell models offer valuable insights into disease mechanisms of surfactant-associated genes and variants, although do not fully recapitulate all AEC2 properties. Notably, they provide efficient, easy-to-maintain, and readily available platforms for studying disease-associated variants in SFTPC and ABCA3. For example, assay systems have been developed to functionally characterize ABCA3 variants, including the expression, co-localization with intracellular organelles, phospholipid uptake, and ABCA3-positive vesicular size and ultrastructure and to assess in vitro responses to pharmacologic therapies69, 71, 72, 82, 8689.

2). Induced Pluripotent Stem Cells and Genome Editing

In addition to immortalized cell lines, induced pluripotent stem cells (iPSCs) derived from affected infants and children with genetic diseases of surfactant dysfunction have been differentiated into AEC2-like cells in vitro90, 91. This approach allows investigators to model disease mechanisms and test potential therapies. For example, iPSCs from individuals with SP-B deficiency or SFTPC variants have been used to create iAEC2s with characteristics closely resembling disease phenotypes90, 91. CRISPR/Cas9 genome editing has also been employed to correct these variants, restoring AEC2 cell function in vitro90. Furthermore, these models have been used to assess the cellular impact in vitro of medications like hydroxychloroquine91.

3). Murine Models

Conditional deletions of SFTPB and ABCA3 in murine models have been used to investigate alveolar injury and regeneration as well as therapeutic interventions including AAV-based gene addition77, 92, 93. Depending on the specific strain, SFTPC gene deletion in mice can lead to inflammation and increased disease susceptibility, particularly with viral respiratory infections94. Recently, murine models expressing BRICHOS or non-BRICHOS SFTPC variants have been developed, resulting in spontaneous inflammation and lung fibrosis95, 96. Mouse models with homozygous deletions of SFTPB and ABCA3 display lethal neonatal respiratory failure shortly after birth, closely resembling the human diseases92, 97, 98. Mice homozygous for the most common pathogenic ABCA3 variant p.Glu292Val show age-dependent inflammation and altered macroautophagy in AEC2s, along with collagen deposition in the alveolar septa99. In utero CRISPR/Cas9 genome editing to inactivate non-BRICHOS p.Ile73Thr mutant alleles improved lung morphology and survival in treated pups81. These mouse models play a crucial role in understanding disease mechanisms and evaluating potential therapies.

4). International registries

Finally, international registries and research networks, such as the European Research Collaboration for Children’s Interstitial Lung Disease (chILD-EU, http://www.childeu.net) and the U.S. National Registry for Childhood Interstitial and Diffuse Lung Disease registry100 play important roles in advancing knowledge, promoting research, improving care, and providing support and resources to clinicians, patients, and families in the quest for therapies.

Conclusion

Genetic surfactant dysfunction diseases are rare, have provided important insights into normal human lung development and function, and are targets for promising therapeutic interventions. Rapid identification of the underlying molecular defect is essential for the diagnosis and is highly relevant for a personalized therapeutic approach. Nonspecific pharmacological therapies currently remain the mainstay for these disorders. Personalized therapies targeting specific variants hold promise, but innovative trial concepts are necessary due to the rarity of cases. Gene therapies tailored to address specific molecular defects may offer the perspective of a cure, but research is still at the preclinical stage. Patients affected with these complex diseases benefit from highly specialized centers offering specific expertise, advanced diagnostic resources in genetics, pathology, and imaging, and multidisciplinary care for acute and chronic management, including lung transplantation.

Practice points:

  • Genetic disorders of surfactant dysfunction are a group of severe or lethal rare diseases with a clinical spectrum spanning from birth to adulthood

  • Early recognition and precision diagnosis are essential for care orientation and treatment

  • Personalized therapies offer new perspectives of treatment and potential cure but are still at the preclinical stage

Research directions:

  • Improve and implement tools for functional characterization of surfactant gene variants

  • Develop and implement vectors and technologies for safe human gene therapies

  • Design and implement innovative clinical trials for targeted pharmacological and genetic therapies

Acknowledgments

The work of Jennifer Wambach was supported by the National Institutes of Health (R01HL149853, U01HL134745 to JAW), St. Louis Children’s Hospital/Washington University School of Medicine Children’s Discovery Institute (JAW)

The work of Matthias Griese. was supported by chILD-EU (FP7, No. 305653), the Bundesministerium für Bildung und Forschung (BMBF), Deutschland, project “HCQ4Surfdefect”, under the frame of E-Rare-3, the ERA-Net for Research on Rare Diseases”, and the ChILD Lung Foundation, Wallasey, UK.

None of the funding sources were implicated in the redaction of this manuscript

Footnotes

Conflict of interest statement

Maureen Peers de Nieuwburgh benefits from the financial support from the Belgian French Community through a FRIA scholarship.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Bibliography

  • 1.Zimmermann L, Janssen D, Tibboel D, Hamvas A, and Carnielli V, Surfactant metabolism in the neonate. Neonatology, 2005. 87(4): p. 296–307. [DOI] [PubMed] [Google Scholar]
  • 2.Wegner DJ, Hertzberg T, Heins HB, et al. , A major deletion in the surfactant protein-B gene causing lethal respiratory distress. Acta Pædiatrica, 2007. 96(4): p. 516–520. [DOI] [PubMed] [Google Scholar]
  • 3.Lawson WE, Polosukhin VV, Stathopoulos GT, et al. , Increased and prolonged pulmonary fibrosis in surfactant protein C-deficient mice following intratracheal bleomycin. The American journal of pathology, 2005. 167(5): p. 1267–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nogee LM, Garnier G, Dietz HC, et al. , A mutation in the surfactant protein B gene responsible for fatal neonatal respiratory disease in multiple kindreds. The Journal of clinical investigation, 1994. 93(4): p. 1860–1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Glasser JR and Mallampalli RK, Surfactant and its role in the pathobiology of pulmonary infection. Microbes and infection, 2012. 14(1): p. 17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Daniels CB and Orgeig S, Pulmonary surfactant: the key to the evolution of air breathing. Physiology, 2003. 18(4): p. 151–157. [DOI] [PubMed] [Google Scholar]
  • 7.Wright JR, Immunoregulatory functions of surfactant proteins. Nature Reviews Immunology, 2005. 5(1): p. 58–68. [DOI] [PubMed] [Google Scholar]
  • 8.Pérez-Gil J, Structure of pulmonary surfactant membranes and films: the role of proteins and lipid–protein interactions. Biochimica et Biophysica acta (BBA)-Biomembranes, 2008. 1778(7–8): p. 1676–1695. [DOI] [PubMed] [Google Scholar]
  • 9.Goerke J, Pulmonary surfactant: functions and molecular composition. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 1998. 1408(2–3): p. 79–89. [DOI] [PubMed] [Google Scholar]
  • 10.Yoshida M, Ikegami M, Reed JA, Chroneos ZC, and Whitsett JA, GM-CSF regulates protein and lipid catabolism by alveolar macrophages. American Journal of Physiology-Lung Cellular and Molecular Physiology, 2001. 280(3): p. L379–L386. [DOI] [PubMed] [Google Scholar]
  • 11.Parra E and Pérez-Gil J, Composition, structure and mechanical properties define performance of pulmonary surfactant membranes and films. Chemistry and physics of lipids, 2015. 185: p. 153–175. [DOI] [PubMed] [Google Scholar]
  • 12.Danhaive O, Chapin C, Horneman H, Cogo PE, and Ballard PL, Surface film formation in vitro by infant and therapeutic surfactants: role of surfactant protein B. Pediatric research, 2015. 77(2): p. 340–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mulugeta S and Beers MF, Surfactant protein C: its unique properties and emerging immunomodulatory role in the lung. Microbes and infection, 2006. 8(8): p. 2317–2323. [DOI] [PubMed] [Google Scholar]
  • 14.Schürch D, Ospina OL, Cruz A, and Pérez-Gil J, Combined and independent action of proteins SP-B and SP-C in the surface behavior and mechanical stability of pulmonary surfactant films. Biophysical journal, 2010. 99(10): p. 3290–3299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Picardi MV, Cruz A, Orellana G, and Pérez-Gil J, Phospholipid packing and hydration in pulmonary surfactant membranes and films as sensed by LAURDAN. Biochimica et Biophysica Acta (BBA)-Biomembranes, 2011. 1808(3): p. 696–705. [DOI] [PubMed] [Google Scholar]
  • 16.Peca D, Cutrera R, Masotti A, Boldrini R, and Danhaive O, ABCA3, a key player in neonatal respiratory transition and genetic disorders of the surfactant system. Biochemical Society Transactions, 2015. 43(5): p. 913–919. [DOI] [PubMed] [Google Scholar]
  • 17.Trapnell BC, Nakata K, Bonella F, et al. , Pulmonary alveolar proteinosis. Nature Reviews Disease Primers, 2019. 5(1): p. 16. [DOI] [PubMed] [Google Scholar]
  • 18.Cooney AL, Wambach JA, Sinn PL, and McCray PB Jr, Gene therapy potential for genetic disorders of surfactant dysfunction. Frontiers in Genome Editing, 2022. 3: p. 785829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Eldridge WB, Zhang Q, Faro A, et al. , Outcomes of Lung Transplantation for Infants and Children with Genetic Disorders of Surfactant Metabolism. J Pediatr, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hamvas A, Cole FS, deMello DE, et al. , Surfactant protein B deficiency: antenatal diagnosis and prospective treatment with surfactant replacement. The Journal of pediatrics, 1994. 125(3): p. 356–361. [DOI] [PubMed] [Google Scholar]
  • 21.Nogee LM, Wert SE, Proffit SA, Hull WM, and Whitsett JA, Allelic heterogeneity in hereditary surfactant protein B (SP-B) deficiency. American journal of respiratory and critical care medicine, 2000. 161(3): p. 973–981. [DOI] [PubMed] [Google Scholar]
  • 22.Nogee LM, de Mello DE, Dehner LP, and Colten HR, Brief report: deficiency of pulmonary surfactant protein B in congenital alveolar proteinosis. N Engl J Med, 1993. 328(6): p. 406–10. [DOI] [PubMed] [Google Scholar]
  • 23.Dunbar AE, Wert SE, Ikegami M, et al. , Prolonged survival in hereditary surfactant protein B (SP-B) deficiency associated with a novel splicing mutation. Pediatric research, 2000. 48(3): p. 275–282. [DOI] [PubMed] [Google Scholar]
  • 24.Hamouda S, Trabelsi I, de Becdelièvre A, and Boussetta K, Difficulties in the treatment of an infant survivor with inherited surfactant protein-B deficiency in Tunisia. Annals of Thoracic Medicine, 2022. 17(2): p. 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Peca D, Boldrini R, Johannson J, et al. , Clinical and ultrastructural spectrum of diffuse lung disease associated with surfactant protein C mutations. European Journal of Human Genetics, 2015. 23(8): p. 1033–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Liptzin DR, McGraw M, Stidham T, Wambach J, and Deterding R, Non-invasive management of infants with SFTPC pathogenic variants. Authorea Preprints, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Thomas AQ, Lane K, Phillips III J, et al. , Heterozygosity for a surfactant protein C gene mutation associated with usual interstitial pneumonitis and cellular nonspecific interstitial pneumonitis in one kindred. American journal of respiratory and critical care medicine, 2002. 165(9): p. 1322–1328. [DOI] [PubMed] [Google Scholar]
  • 28.Mulugeta S, Nguyen V, Russo SJ, Muniswamy M, and Beers MF, A surfactant protein C precursor protein BRICHOS domain mutation causes endoplasmic reticulum stress, proteasome dysfunction, and caspase 3 activation. American journal of respiratory cell and molecular biology, 2005. 32(6): p. 521–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Beers MF, Hawkins A, Maguire JA, et al. , A nonaggregating surfactant protein C mutant is misdirected to early endosomes and disrupts phospholipid recycling. Traffic, 2011. 12(9): p. 1196–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang W-J, Mulugeta S, Russo SJ, and Beers MF, Deletion of exon 4 from human surfactant protein C results in aggresome formation and generation of a dominant negative. Journal of cell science, 2003. 116(4): p. 683–692. [DOI] [PubMed] [Google Scholar]
  • 31.Shulenin S, Nogee LM, Annilo T, et al. , ABCA3 gene mutations in newborns with fatal surfactant deficiency. New England Journal of Medicine, 2004. 350(13): p. 1296–1303. [DOI] [PubMed] [Google Scholar]
  • 32.Bullard JE, Wert SE, Whitsett JA, Dean M, and Nogee LM, ABCA3 mutations associated with pediatric interstitial lung disease. American journal of respiratory and critical care medicine, 2005. 172(8): p. 1026–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Brasch F, Schimanski S, Mühlfeld C, et al. , Alteration of the pulmonary surfactant system in full-term infants with hereditary ABCA3 deficiency. American journal of respiratory and critical care medicine, 2006. 174(5): p. 571–580. [DOI] [PubMed] [Google Scholar]
  • 34.Wambach JA, Casey AM, Fishman MP, et al. , Genotype–phenotype correlations for infants and children with ABCA3 deficiency. American journal of respiratory and critical care medicine, 2014. 189(12): p. 1538–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kroner C, Wittmann T, Reu S, et al. , Lung disease caused by ABCA3 mutations. Thorax, 2017. 72(3): p. 213–220. [DOI] [PubMed] [Google Scholar]
  • 36.Henderson LB, Melton K, Wert S, et al. , Large ABCA3 and SFTPC deletions resulting in lung disease. Annals of the American Thoracic Society, 2013. 10(6): p. 602–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wambach JA, Wegner DJ, Depass K, et al. , Single ABCA3 mutations increase risk for neonatal respiratory distress syndrome. Pediatrics, 2012. 130(6): p. e1575–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Naderi HM, Murray JC, and Dagle JM, Single mutations in ABCA3 increase the risk for neonatal respiratory distress syndrome in late preterm infants (gestational age 34–36 weeks). Am J Med Genet A, 2014. 164A(10): p. 2676–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wittmann T, Frixel S, Höppner S, et al. , Increased risk of interstitial lung disease in children with a single R288K variant of ABCA3. Molecular Medicine, 2016. 22: p. 183–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hamvas A, Nogee LM, Wegner DJ, et al. , Inherited surfactant deficiency caused by uniparental disomy of rare mutations in the surfactant protein-B and ATP binding cassette, subfamily a, member 3 genes. The Journal of pediatrics, 2009. 155(6): p. 854–859. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Agrawal A, Hamvas A, Cole F, et al. , An intronic ABCA3 mutation that is responsible for respiratory disease. Pediatric research, 2012. 71(6): p. 633–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bullard JE and Nogee LM, Heterozygosity for ABCA3 mutations modifies the severity of lung disease associated with a surfactant protein C gene (SFTPC) mutation. Pediatric research, 2007. 62(2): p. 176–179. [DOI] [PubMed] [Google Scholar]
  • 43.Maeda Y, Dave V, and Whitsett JA, Transcriptional control of lung morphogenesis. Physiological reviews, 2007. 87(1): p. 219–244. [DOI] [PubMed] [Google Scholar]
  • 44.Galambos C, Levy H, Cannon CL, et al. , Pulmonary pathology in thyroid transcription factor-1 deficiency syndrome. American journal of respiratory and critical care medicine, 2010. 182(4): p. 549–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hamvas A, Deterding RR, Wert SE, et al. , Heterogeneous pulmonary phenotypes associated with mutations in the thyroid transcription factor gene NKX2–1. Chest, 2013. 144(3): p. 794–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Thorwarth A, Schnittert-Hübener S, Schrumpf P, et al. , Comprehensive genotyping and clinical characterisation reveal 27 novel NKX2–1 mutations and expand the phenotypic spectrum. Journal of medical genetics, 2014. 51(6): p. 375–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Suzuki T, Sakagami T, Rubin BK, et al. , Familial pulmonary alveolar proteinosis caused by mutations in CSF2RA. The Journal of experimental medicine, 2008. 205(12): p. 2703–2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dirksen U, Nishinakamura R, Groneck P, et al. , Human pulmonary alveolar proteinosis associated with a defect in GM-CSF/IL-3/IL-5 receptor common beta chain expression. The Journal of clinical investigation, 1997. 100(9): p. 2211–2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hildebrandt J, Yalcin E, Bresser H-G, et al. , Characterization of CSF2RA mutation related juvenile pulmonary alveolar proteinosis. Orphanet journal of rare diseases, 2014. 9: p. 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hadchouel A, Wieland T, Griese M, et al. , Biallelic mutations of methionyl-tRNA synthetase cause a specific type of pulmonary alveolar proteinosis prevalent on Réunion Island. The American Journal of Human Genetics, 2015. 96(5): p. 826–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Huang H, Pan J, Spielberg DR, et al. , A dominant negative variant of RAB5B disrupts maturation of surfactant protein B and surfactant protein C. Proceedings of the National Academy of Sciences, 2022. 119(6): p. e2105228119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Carlens J, Price T, Bültmann E, et al. , Interstitial lung disease caused by filamin A gene mutations: clinical course of three paediatric cases. 2016, Eur Respiratory Soc. [Google Scholar]
  • 53.Nathan N, Giraud V, Picard C, et al. , Germline SFTPA1 mutation in familial idiopathic interstitial pneumonia and lung cancer. Human molecular genetics, 2016. 25(8): p. 1457–1467. [DOI] [PubMed] [Google Scholar]
  • 54.Seidl E, Schwerk N, Carlens J, et al. , Acute exacerbations in children’s interstitial lung disease. Thorax, 2022. 77(8): p. 799–804. [DOI] [PubMed] [Google Scholar]
  • 55.Kröner C, Reu S, Teusch V, et al. , Genotype alone does not predict the clinical course of SFTPC deficiency in paediatric patients. European Respiratory Journal, 2015. 46(1): p. 197–206. [DOI] [PubMed] [Google Scholar]
  • 56.Li Y, Seidl E, Knoflach K, et al. , ABCA3-related interstitial lung disease beyond infancy. Thorax, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bush A, Cunningham S, De Blic J, et al. , European protocols for the diagnosis and initial treatment of interstitial lung disease in children. Thorax, 2015. 70(11): p. 1078–1084. [DOI] [PubMed] [Google Scholar]
  • 58.Braun S, Ferner M, Kronfeld K, and Griese M, Hydroxychloroquine in children with interstitial (diffuse parenchymal) lung diseases. Pediatric pulmonology, 2015. 50(4): p. 410–419. [DOI] [PubMed] [Google Scholar]
  • 59.Griese M, Kappler M, Stehling F, et al. , Randomized controlled phase 2 trial of hydroxychloroquine in childhood interstitial lung disease. Orphanet Journal of Rare Diseases, 2022. 17(1): p. 289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kröner C, Wittmann T, Reu S, et al. , Lung disease caused by ABCA3 mutations. Thorax, 2017. 72(3): p. 213–220. [DOI] [PubMed] [Google Scholar]
  • 61.Deterding R, Young LR, DeBoer EM, et al. , Nintedanib in children and adolescents with fibrosing interstitial lung diseases. European Respiratory Journal, 2023. 61(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Manickam K, McClain MR, Demmer LA, et al. , Exome and genome sequencing for pediatric patients with congenital anomalies or intellectual disability: an evidence-based clinical guideline of the American College of Medical Genetics and Genomics (ACMG). Genetics in Medicine, 2021. 23(11): p. 2029–2037. [DOI] [PubMed] [Google Scholar]
  • 63.Iablonskii P, Carlens J, Mueller C, et al. , Indications and outcome after lung transplantation in children under 12 years of age: a 16-year single center experience. The Journal of Heart and Lung Transplantation, 2022. 41(2): p. 226–236. [DOI] [PubMed] [Google Scholar]
  • 64.Glanville AR, Verleden GM, Todd JL, et al. , Chronic lung allograft dysfunction: Definition and update of restrictive allograft syndrome-A consensus report from the Pulmonary Council of the ISHLT. J Heart Lung Transplant, 2019. 38(5): p. 483–492. [DOI] [PubMed] [Google Scholar]
  • 65.Hubbard R, Miller R, Tumin D, Tobias JD, and Hayes D Jr., Transplant outcomes for idiopathic pulmonary hypertension in children. J Heart Lung Transplant, 2019. 38(5): p. 580–581. [DOI] [PubMed] [Google Scholar]
  • 66.Faro AH, A., Lung Transplantation for Inherited Disorders of Surfactant Metabolism. Neoreviews, 2008. 9(10): p. e468–e476. [Google Scholar]
  • 67.Ramsey BW, Davies J, McElvaney NG, et al. , A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. New England Journal of Medicine, 2011. 365(18): p. 1663–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Griese M, Costa S, Linnemann RW, et al. , Safety and efficacy of elexacaftor/tezacaftor/ivacaftor for 24 weeks or longer in people with cystic fibrosis and one or more F508del alleles: interim results of an open-label phase 3 clinical trial. American journal of respiratory and critical care medicine, 2021. 203(3): p. 381–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yang X, Forstner M, Rapp CK, et al. , ABCA3 Deficiency—Variant-Specific Response to Hydroxychloroquine. International Journal of Molecular Sciences, 2023. 24(9): p. 8179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kinting S, Li Y, Forstner M, et al. , Potentiation of ABCA3 lipid transport function by ivacaftor and genistein. Journal of cellular and molecular medicine, 2019. 23(8): p. 5225–5234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kinting S, Höppner S, Schindlbeck U, et al. , Functional rescue of misfolding ABCA3 mutations by small molecular correctors. Human molecular genetics, 2018. 27(6): p. 943–953. [DOI] [PubMed] [Google Scholar]
  • 72.Forstner M, Lin S, Yang X, et al. , High-content screening identifies cyclosporin A as a novel ABCA3-specific molecular corrector. American Journal of Respiratory Cell and Molecular Biology, 2022. 66(4): p. 382–390. [DOI] [PubMed] [Google Scholar]
  • 73.Wang D, Tai PW, and Gao G, Adeno-associated virus vector as a platform for gene therapy delivery. Nature reviews Drug discovery, 2019. 18(5): p. 358–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Maguire AM, Simonelli F, Pierce EA, et al. , Safety and efficacy of gene transfer for Leber’s congenital amaurosis. New England Journal of Medicine, 2008. 358(21): p. 2240–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mendell JR, Al-Zaidy S, Shell R, et al. , Single-dose gene-replacement therapy for spinal muscular atrophy. New England Journal of Medicine, 2017. 377(18): p. 1713–1722. [DOI] [PubMed] [Google Scholar]
  • 76.Kohn DB, Booth C, Shaw KL, et al. , Autologous ex vivo lentiviral gene therapy for adenosine deaminase deficiency. New England Journal of Medicine, 2021. 384(21): p. 2002–2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kang MH, van Lieshout LP, Xu L, et al. , A lung tropic AAV vector improves survival in a mouse model of surfactant B deficiency. Nature communications, 2020. 11(1): p. 3929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Munis AM, Hyde SC, and Gill DR, A human surfactant B deficiency air-liquid interface cell culture model suitable for gene therapy applications. Molecular Therapy-Methods & Clinical Development, 2021. 20: p. 237–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Leibel SL, Winquist A, Tseu I, et al. , Reversal of surfactant protein B deficiency in patient specific human induced pluripotent stem cell derived lung organoids by gene therapy. Scientific reports, 2019. 9(1): p. 13450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Alton EW, Beekman JM, Boyd AC, et al. , Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax, 2017. 72(2): p. 137–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Alapati D, Zacharias WJ, Hartman HA, et al. , In utero gene editing for monogenic lung disease. Science translational medicine, 2019. 11(488): p. eaav8375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wambach JA, Yang P, Wegner DJ, et al. , Functional genomics of ABCA3 variants. American journal of respiratory cell and molecular biology, 2020. 63(4): p. 436–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Matsumura Y, Ban N, Ueda K, and Inagaki N, Characterization and classification of ATP-binding cassette transporter ABCA3 mutants in fatal surfactant deficiency. Journal of Biological Chemistry, 2006. 281(45): p. 34503–34514. [DOI] [PubMed] [Google Scholar]
  • 84.Matsumura Y, Ban N, and Inagaki N, Aberrant catalytic cycle and impaired lipid transport into intracellular vesicles in ABCA3 mutants associated with nonfatal pediatric interstitial lung disease. American Journal of Physiology-Lung Cellular and Molecular Physiology, 2008. 295(4): p. L698–L707. [DOI] [PubMed] [Google Scholar]
  • 85.Beers MF and Mulugeta S, The biology of the ABCA3 lipid transporter in lung health and disease. Cell and tissue research, 2017. 367: p. 481–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Schindlbeck U, Wittmann T, Höppner S, et al. , ABCA3 missense mutations causing surfactant dysfunction disorders have distinct cellular phenotypes. Human mutation, 2018. 39(6): p. 841–850. [DOI] [PubMed] [Google Scholar]
  • 87.Li Y, Kinting S, Hoeppner S, et al. , Metabolic labelling of choline phospholipids probes ABCA3 transport in lamellar bodies. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids, 2019. 1864(12): p. 158516. [DOI] [PubMed] [Google Scholar]
  • 88.Yang X, Rapp CK, Li Y, Forstner M, and Griese M, Quantifying Functional Impairment of ABCA3 Variants Associated with Interstitial Lung Disease. International Journal of Molecular Sciences, 2023. 24(8): p. 7554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wambach JA, Yang P, Wegner DJ, et al. , Functional Characterization of ATP-Binding Cassette Transporter A3 Mutations from Infants with Respiratory Distress Syndrome. Am J Respir Cell Mol Biol, 2016. 55(5): p. 716–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Jacob A, Morley M, Hawkins F, et al. , Differentiation of human pluripotent stem cells into functional lung alveolar epithelial cells. Cell stem cell, 2017. 21(4): p. 472–488. e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Alysandratos K, Russo S, Petcherski A, et al. , Patient-specific iPSCs carrying anSFTPCmutation reveal the intrinsic alveolar epithelial dysfunction at the inception of interstitial lung disease. 2020. [DOI] [PMC free article] [PubMed]
  • 92.Rindler TN, Stockman CA, Filuta AL, et al. , Alveolar injury and regeneration following deletion of ABCA3. JCI insight, 2017. 2(24). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Melton KR, Nesslein LL, Ikegami M, et al. , SP-B deficiency causes respiratory failure in adult mice. American Journal of Physiology-Lung Cellular and Molecular Physiology, 2003. 285(3): p. L543–L549. [DOI] [PubMed] [Google Scholar]
  • 94.Glasser SW, Senft AP, Maxfield MD, et al. , Genetic replacement of surfactant protein-C reduces respiratory syncytial virus induced lung injury. Respiratory research, 2013. 14: p. 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Nureki S-I, Tomer Y, Venosa A, et al. , Expression of mutant Sftpc in murine alveolar epithelia drives spontaneous lung fibrosis. The Journal of clinical investigation, 2018. 128(9): p. 4008–4024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Katzen J, Wagner BD, Venosa A, et al. , An SFTPC BRICHOS mutant links epithelial ER stress and spontaneous lung fibrosis. JCI insight, 2019. 4(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Clark JC, Wert SE, Bachurski CJ, et al. , Targeted disruption of the surfactant protein B gene disrupts surfactant homeostasis, causing respiratory failure in newborn mice. Proceedings of the National Academy of Sciences, 1995. 92(17): p. 7794–7798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ban N, Matsumura Y, Sakai H, et al. , ABCA3 as a lipid transporter in pulmonary surfactant biogenesis. Journal of Biological Chemistry, 2007. 282(13): p. 9628–9634. [DOI] [PubMed] [Google Scholar]
  • 99.Tomer Y, Wambach J, Knudsen L, et al. , The common ABCA3E292V variant disrupts AT2 cell quality control and increases susceptibility to lung injury and aberrant remodeling. American Journal of Physiology-Lung Cellular and Molecular Physiology, 2021. 321(2): p. L291–L307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nevel RJ, Deutsch GH, Craven D, et al. , The US national registry for childhood interstitial and diffuse lung disease: Report of study design and initial enrollment cohort. Pediatric Pulmonology, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES