Abstract Mucopolysaccharidosis (MPS) encompasses a heterogeneous group of lysosomal storage diseases resulting from mutations in genes encoding lysosomal enzymes responsible for the degradation of mucopolysaccharides, also known as glycosaminoglycans (GAGs). Current therapeutic strategies for MPS include hematopoietic stem cell transplantation (HSCT), enzyme replacement therapy (ERT), and symptomatic therapy. This study investigated dynamic changes in MPS type II (MPS-II) through genomic and single-cell sequencing in a patient undergoing ERT. Analysis of peripheral blood mononuclear cells (PBMCs) from one MPS-II patient of 10 year old at different disease stages through scRNA-seq identified various immune cell types, including natural killer (NK) cells, NKT cells, CD4 + and CD8 + T cells, CD14 + and CD16 + monocytes, and B cells. Monocytes and macrophages were significantly reduced during the severe stage of MPS-II but increased during the recovery stage following ERT. Notably, monocyte subtype mono3 was exclusively expressed in the severe stage, while mono1_2, a subtype of mono1, was absent during the severe stage and exhibited distinct biological functions. These findings suggest that monocytes and macrophages play critical roles in the pathogenesis of MPS-II and in the response to ERT. Pseudotime, Gene Ontology, and cell-communication analyses revealed unique functions for the different cellular subtypes. Notably, key molecules mediating cellular interactions during ERT in MPS-II included CXCR3, PF4, APP, and C5AR1 in macrophages, RPS19 in T cells, HLA-DPB1 in B cells, ADRB2 in NK cells, and IL1B, C5AR1, RPS19, and TNFSF13B in monocytes. Overall, integrative analysis delineated the expression dynamics of various cell types and identified mutations in MPS-II, providing a comprehensive atlas of transcriptional programs, cellular characterizations, and genomic variation profiles in MPS-II. This dataset, along with advanced integrative analysis, represents a valuable resource for the discovery of drug targets and the improvement of therapeutic strategies for MPS-II. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-97330-7. Keywords: Type II mucopolysaccharidosis, Enzyme replacement therapy, Single-cell RNA sequencing, T-cell receptor sequencing, Whole-genome sequencing Subject terms: Endocrine system and metabolic diseases, Disease genetics Introduction Mucopolysaccharidosis (MPS) comprises a heterogeneous group of lysosomal storage diseases caused by mutations in genes encoding lysosomal enzymes responsible for the degradation of mucopolysaccharides, also termed glycosaminoglycans (GAGs)^[36]1. There are seven recognized types of MPS, which include a total of 13 subtypes, each defined by specific gene mutations that result in the dysfunction of corresponding enzymes responsible for GAG degradation^[37]2. MPS types can be categorized into somatic and neuronopathic forms. Type I MPS is caused by the deficiency of a-l-iduronidase, whose deficiency results in accumulation of GAGs^[38]3. Type III MPS (Sanfilippo syndrome) involves multiple enzyme deficiencies involving heparan-N-sulfatase (IIIA), a-N-acetylglucosaminidase (IIIB), a-glucosaminide acetyltransferase (IIIC), and N-acetylglucosamine-6-sulfatase (IIID), causing accumulation of HS. MPS IIIA and IIIB are associated with mutations in the genes SGSH and NAGLU, respectively^[39]4. Type IV MPS (Morquio syndrome) is of two types: type IVA which occurs due to the deficiency of N-acetylgalactosamine-6-sulfatas and Type IVB which occurs due to the deficiency of b-galactosidase enzyme^[40]3. Type VI MPS (Maroteaux Lamy) occurs due to the accumulation of DS and chondroitin-4-sulfate (C4S) as a result of deficiency of the enzyme N-acetylgalactosamine-4-sulfatase involving gene ARSB^[41]3. Partial degradation and accumulation of C4S, chondroitin-6-sulfate (C6S), DS, and HS as a result of b-galactosidase deficiency due mutation in the gene GUSB leads to Type VII MPS (Sly syndrome)^[42]3. Type IX MPS (Natowicz syndrome) is the rarest of all types and occurs due to the deficiency of hyaluronidase enzyme resulting in accumulation of hyaluronan^[43]3. MPS type II (MPS-II), also known as Hunter syndrome, is the only MPS with X-linked inheritance, caused by a deficiency in the enzyme iduronate-2-sulfatase (IDS), leading to the progressive accumulation of dermatan sulfate (DS) and heparan sulfate (HS)^[44]5. Current therapeutic strategies for MPS include hematopoietic stem cell transplantation (HSCT), enzyme replacement therapy (ERT), and symptomatic therapy^[45]6,[46]7. Several clinical trials of gene therapy for MPS-II were conducted, including retroviruses with genome editing systems such as zinc finger nucleases or CRISPR/Cas9^[47]8. Clinical trials of adeno-associated viruses were also conducted for MPS-II patients, which could improve central nervous system function of patients^[48]9,[49]10. ERT can reduce the accumulation of GAGs in cells, alleviating hepatosplenomegaly and improving cardiopulmonary function and joint mobility, but conventional ERT exhibits limited efficacy in improving cognitive and central nervous system functions due to the blood-brain barrier (BBB). New types of ERT using monoclonal antibody (MAb) that functions as a vehicle to bypass the BBB are under clinical trials for MPS-II^[50]11. Another method to penetrate the BBB is to change the injection route, via IT or intracerebroventricular; however, IT ERT for MPS-II did not demonstrate therapeutic efficacy and have been discontinued^[51]12. Although early HSCT can significantly improve neurological symptoms, the risk of graft-versus-host disease and long-term use of multiple immunosuppressants limit its applicability. HSCT can stabilize or prevent hydrocephalus and prevent the deterioration of psychomotor functions, and also has been associated with improvements in cognitive function and CNS manifestation^[52]13,[53]14. Previous meta-analysis has demonstrated that ERT in MPS-II reduces urinary GAG content and exhibits a dose-response relationship, while several animal model studies have reported IDS deficiency and increased GAG levels in the context of MPS-II^[54]15–[55]17. However, these studies failed to capture molecular heterogeneity, necessitating the observation and measurement of ERT effects at the transcriptome level. Single-cell RNA sequencing (scRNA-seq) enables the identification of cell subpopulations and their distinct functions by exploring the transcriptomic profiles of individual cells. This technology has emerged as a powerful tool for elucidating pathophysiological changes in diseases^[56]18 and profiling gene expression at the single-cell level^[57]19. Recently, scRNA-seq has revolutionized and expanded our ability to investigate critical questions in metabolic diseases, such as type 1 diabetes, adipogenesis, osteoclast function, and pancreatic beta-cell differentiation^[58]20–[59]22. Previous studies explored transcriptomic changes in MPS through RNA sequencing^[60]23. However, no research has focused on MPS-II to measure how cells change during treatment at the single-cell level. In this study, whole-genome sequencing (WGS) was performed on an MPS-II patient, along with scRNA-seq, T-cell receptor sequencing (TCR-seq), and B-cell receptor sequencing (BCR-seq) at two different ERT stages. This comprehensive characterization of cell dynamics was conducted using peripheral blood mononuclear cells (PBMCs). The WGS results identified 16 IDS germline mutations in the MPS-II patient, with only one frame shift deletion mutation remaining after filtering. The scRNA-seq results revealed that monocytes and macrophages almost disappeared in severe MPS-II and increased during the recovery stage following treatment. This suggests that monocytes and macrophages play crucial roles in MPS-II pathogenesis and response to ERT. Our genomic and single-cell transcriptomic analyses provide an atlas of transcriptional programs, cellular characterizations, and genomic variation profiles in MPS-II, representing a valuable resource for MPS-II drug target discovery and therapeutic strategy improvement. Methods Patient material, ethics statement, and consent for publication MPS was diagnosed based on clinical manifestations, iduronate-2-sulfatase deficiency, and IDS gene mutations. The MPS patient received four treatments with iduronate-2-sulfatase at a dosage of 0.5 mg/kg. PBMCs were collected after the first and second treatment stages. Clinical data for the patient are provided in Table [61]1. All procedures involving human subjects and tissues were approved by the Ethics Committee of the Children’s Hospital of Chongqing Medical University (NO.202364), with signed informed consent obtained from the patient’s parents. Table 1. Clinical information of MPS II patient. Basic Information Sex Male Onset age 2 years old Diagnosis age 10 years old 2 months Diagnosis date January 2022 Clinical manifestation Coarse faces: macrocephaly, prominent forehead, thick hair, blunt nasal tip, and thick lips Dysostosis multiplex: claw hands, joint stiffness/contracture/ enlargement, short stature Nervous system: intelligence Regression, behavioral abnormalities, disturbed sleep, epilepsy, IQ/DQ assessment was not completed, FMFM98points (98/183, 66.42%), GMFM203points (203/260, 78.00%) Ear-nose-throat and mouth: airway obstruction, tonsil/adenoid hypertrophy, snoring, recurrent respiratory infections Cardiovascular system: the aortic valves regurgitation, ventricular hypertrophy and heart failure Digestive system: abdominal distension, evident hepatosplenomegaly UGAGs (DMB) + IDS(DBS) 0.00(>1.500 nmol/h/mL) WGS IDS (c.1033delT) Clinical classification Severe ERT start time April 7th, 2022 ERT start time April 28th, 2022 MPS1: before ERT MPS2: after ERT Assessment age 10-years-old 2months 10-years-old 6months Clinical manifestation Respiratory infection 2–3/month 1/month Seizures Up to 7 times a day Absent Sleep quality Unable to sleep on back, snore, Occasional apnea, sleep light Able to sleep on back for 3–4 h 6MWT Walk 6 m alone, easy to fall Walk 20 m alone 3MSC Unable to climb stair alone Able to climb 5 stairs alone Volume of liver and spleen Liver was 5 cm below the costal margin, spleen was 3 cm below the costal margin Liver was 4 cm below the costal margin, below spleen was 3 cm the costal margin Height 116.5 cm 119.5 cm [62]Open in a new tab FMFM : Fine Motor Function Measure. GMFM : Gross Motor Function Measure. UGAGs: Urinary glycosaminoglycans. DMB: Simethyl methylene blue. IDS: Iduronate-2-sulfatase. DBS: Dried blood spot. WGS: Whole Genome Sequencing. ERT: Enzyme replacement therapy (idursulfase-β, Hunterase^®). MWT: Minute walking test. MSC: Minute stair climbing experiment. Isolation of PBMCs, single-cell library construction, and VDJ-sequencing Ficoll-Paque PREMIUM was used to isolate PBMCs from 3 mL of EDTA-treated blood from the patient following the manufacturer’s manual. For scRNA-seq data, patient cell suspensions were pooled in equal volumes and diluted to a concentration of 1 × 10^6 cells/mL. There was a slight donor bias in the number of cells from each sample in the mixture. The mixed suspension was then loaded onto the 10× Genomics platform to create a barcoded cDNA library for individual cells. The individual libraries were pooled and sequenced using the Illumina HiSeq 2500 platform. Cryopreserved samples were thawed at 37 ℃ and washed twice in ice-cold 1 × phosphate-buffered saline (PBS). ScRNA-seq, TCR-seq, and BCR-seq were performed on CD138 + and CD138 − bone marrow mononuclear cells (BMMCs) using the Chromium Next GEM Single-cell 5’ Reagent Kit v1.1 and V(D)J Reagent Kit v1.1 according to the protocols provided by the manufacturer (14 000 cells/channel). Generated gene expression libraries were paired-end sequenced using the NovaSeq 6000 S2 platform and generated V(D)J libraries were paired-end sequenced using the NextSeq500 platform. ScRNA-seq Freshly prepared PBMC suspensions were processed immediately using a 10× Chromium 3’ v3 Kit (10× Genomics, Pleasanton, CA, USA), in accordance with the manufacturer’s protocols. The libraries were sequenced using the Illumina NovaSeq 6000 platform with paired-ended sequencing and dual indexing. Sequencing involved 26 cycles for Read 1, eight cycles for the i7 index, and 98 cycles for Read 2. On average, each sample generated approximately 709 million reads (standard deviation (SD) of 121 million). Data processing, integration, and cluster annotation Raw BCL files were demultiplexed and mapped to the reference genome GRCh38 using Cell Ranger Single-cell (v1.1.0). Doublets were removed through DoubletFinder (v2.0.3). The filtered count matrix was converted to a Seurat object using Seurat (v4.3.0). Cells with more than 3 000 genes, fewer than 200 genes, or more than 15% mitochondrial counts were excluded. Data normalization for each sample was performed using the NormalizeData method, and the top 3 000 highly variable genes were selected for subsequent dimensionality reduction. Batch effects among samples were corrected using the integration method in Seurat. Principal component analysis (PCA) and uniform manifold approximation and projection (UMAP) were used to reduce dimensionality and visualize cell distribution. Cluster-specific genes were identified using the Wilcoxon rank-sum test in the FindAllMarkers function (fold-change ≥ 2 and adjusted p ≤ 0.05). Cell clusters were annotated based on well-recognized marker genes. Cell-cell communication analysis To investigate the molecular interaction networks between different cell types, CellPhoneDB (v3.0.0)^[63]24 was used to infer cell-cell communication from the combined expression of multi-subunit ligand-receptor complexes. Ligand-receptor pairs (P < 0.05) were retained to assess the relationships among different cell clusters. Single-cell trajectory and RNA velocity analyses R package Monocle2^[64]25 was used to order cells in pseudotime along a developmental trajectory. After dimensionality reduction, cells were arranged in pseudotime to reflect their progression through the developmental program, enabling single-cell trajectory analysis of cell subtypes using Monocle2. Velocyto R package (v0.6)^[65]26 was used to evaluate cell lineage dynamics and RNA velocity. Spliced and unspliced count matrices were constructed using the Velocyto pipeline based on “Cell Ranger” output files. Loom files for each sample were created and merged, retaining the top 3 000 most variable genes, with a minimum of 100 spliced counts. After data normalization and scaling, cell clusters were embedded with velocity streams based on the ratio of spliced to unspliced counts and projected onto a UMAP plot for visualization. Pathway and gene ontology (GO) enrichment analysis Gene set enrichment analysis (GSEA) was performed using ClusterProfiler (v4.6.2)^[66]27. Differentially expressed genes (DEGs) (adjusted P < 0.05 and |Log2fold-change (FC) > 0.25) from each PBMC subpopulation were used for Kyoto Encyclopedia of Genes and Genomes (KEGG)^[67]28 and GO enrichment analysis. Pathways and biological processes with P < 0.05 were considered significantly enriched. TCR/BCR-seq and copy number variation (CNV) analyses VDJ profiled_contig_annotations files were loaded into R, combined for all patients (separately for TCR-seq and BCR-seq), and mapped to the Seurat object using the scRepertoire package (v1.8.0). CNV features of cell clusters were examined using CopyKat to distinguish aneuploid cells in each sample with default parameters^[68]29. WGS and mutation calling Genomic DNA was extracted from frozen blood plasma samples, with library preparation details provided in the Supplementary Materials and Methods. The DNA libraries were sequenced using the Illumina HiSeq platform, generating 150-bp paired-end reads. The raw sequencing data were mapped to human reference genome hg38 (GATK Resource Bundle) using BWA-MEM (v.0.7.17)^[69]30. Following GATK best practices, the .bam file was sorted and indexed, with polymerase chain reaction (PCR) duplicates marked and removed and base quality recalibrated. Germline mutations were detected using GATK HaplotypeCaller^[70]31, Strelka^[71]32, and VarScan^[72]33, retaining only those mutations called by at least two algorithms. Bam-readcount^[73]34 was used to quantify the number of reference and alternative alleles, filtering variants with fewer than five alternative allele counts or an alternative allele frequency below 20%. Mutations were also filtered for rare variants with an allele frequency below 0.05% in the 1000 Genomes and Genome Aggregation Database (gnomAD). Results Single-cell transcriptome and genome landscape of PBMCs in an MPS-II patient during ERT The MPS-II-diagnosed patient exhibited significant clinical features, including typical facial characteristics such as alar hypertrophy, enlarged tongue, thick lips, and clawed hands. Imaging studies showed a J-shaped sella on X-ray and an enlarged supratentorial region on MRI (Fig. [74]1A). Other clinical manifestations were provided in Table [75]1. To investigate the composition and diversity of PBMCs at different stages of clinical treatment, scRNA-seq, TCR-seq, and BCR-seq were performed on PBMCs from an MPS-II patient at two ERT stages (MPS II (a): severe stage before treatment; MPS II (b): recovery stage after treatment), and related clinical performance was summarized in Table [76]1. Combined with a public scRNA-seq dataset of PBMCs from three healthy children^[77]35, a total of 56 479 cells were obtained with an average of 2 000 genes per cell after quality control. Cell clusters were visualized using UMAP and annotated into nine cell types, including B cells, CD14 + and CD16 + monocytes, CD4 + and CD8 + T cells, macrophages, NK cells, NKT cells, and erythrocytes (FigS. [78]1B and C, S1-A). Analysis of cell type counts and composition in each sample indicated a marked reduction of monocytes and a limited presence of macrophages in MPS II (a) compared to MPS II (b) and normal controls (FigS. [79]1B, S2-B). The recovery of these cells in MPS II (b) suggested an improvement in phagocytic function. Additionally, a reduction in T cells and B cells in MPS II (b) was observed, potentially indicating their depletion following immunoreaction. NK and NKT cells remained steady between the control group and two treatment stages. Fig. 1. [80]Fig. 1 [81]Open in a new tab Clinical manifestations and single-cell transcriptional profile of mucopolysaccharidosis II (MPS-II) patient. (A) Clinical pictures of the patient. (a) claw hands of the patient; (b) X-ray image shows the j-shape sella; (c) MRI image shows enlarged supratentorial ventricle, deepened cerebral sulci, and scattered small cystic with long T1 and long T2 signals. (B) Uniform manifold approximation and projection (UMAP) plot of integrated data splited by group (Health: normal sample; MPS II (a): sample from first stage treatment; MPS II (b): sample from second stage treatment), colored by cell type. (C) Cell types identified by known marker genes. (D) Heatmap showing top 10 differential expressed genes in each group. (E) Location of germline mutation of IDS, HPSE, HPSE2. (F) Feature plot showing IDS gene expression level in three groups. (G) Heatmap illustrating the expression level of genes involved in pathways related with IDS in three groups. DEGs were compared among the control, MPS II (a), and MPS II (b) (Figs. [82]1D, S1-C, S1- D, S1-E, Table S-2). Results showed that MPS II (a) exhibited higher expression of TNFAIP3, TUBB2A, UBE2S, and TRABD2A but lower expression of TYROBP, TRBC1, and TSPO compared to the control and MPS II (b) (Fig. [83]1D). TYROBP, TRBC1, and ZNF385A were up-regulated in MPS II (b) and the control group, but down-regulated in MPS II (a). TYROBP is known to influence immune system dysregulation, bone and skeletal abnormalities, and neuroinflammation^[84]36. Activation and inactivation of these genes may be associated with ERT. GO and KEGG^[85]28 enrichment analyses of DEGs among the healthy control, MPS II (a), and MPS II (b) groups were conducted using clusterProfiler^[86]26 (Figure [87]S1D, S1-E, Table S-3, S-4). ARSK was up-regulated in MPS II (a) but down-regulated in the control and MPS II (b). Genes up-regulated in MPS II (a) were enriched in immune-related pathways, such as immune cell differentiation and proliferation, Wnt signaling pathway, and JAK-STAT signaling pathway (Fig. [88]S1D). In contrast, genes up-regulated in MPS II (b) were predominantly enriched in inflammatory response, phagocytosis, and immune cell activation (Fig. [89]S1E). Based on WGS data of the MPS-II PBMCs, several MPS-II-related germline mutations were detected, including IDS, HPSE, and HPSE2 (Fig. [90]1E, Table S-[91]1). IDS is a key gene associated with MPS-II, involved in calcium binding, protein binding, sulfuric ester hydrolase activity, hydrolase activity, and heparan sulfate metabolism^[92]37. The expression of IDS was high in the early stage (MPS II (a)) but decreased in the later stage (MPS II (b)), suggesting that ERT suppresses its expression (Fig. [93]1F). Similarly, HPSE and HPSE2, which are also involved in GAG metabolism and heparan sulfate biosynthesis, show altered expression in MPS-II according to GeneCards ([94]www.genecards.org). The expression levels of IDS-related genes among the three groups showed that S100A6, CLPX, and GUSB were up-regulated in the control and MPS II (b) groups but down-regulated in MPS II (a) (Fig. [95]1G). Our findings suggest that monocytes and macrophages could play crucial roles in the pathogenesis of our MPS-II patient and response to ERT. Furthermore, genomic results indicated that IDS mutations accompanied by HPSE and HPSE2 were identified in our MPS-II patient. Monocytes play important roles in response to ERT in MPS-II Monocytes, which nearly disappeared in the severe stage (MPS II (a)) but demonstrated recovery following ERT, were re-clustered into four subclusters (Figs. [96]2A, S2-A). Most CD14 + CD16 + monocytes, typically classified in the intermediate subset, fell within classical (mono1) and nonclassical (mono2) monocyte subclusters, with some cells forming a rare subcluster (mono3) associated with the cell cycle, differentiation, and trafficking, as well as a subcluster (mono4) expressing cytotoxic genes involved in NK and T cell activation^[97]38. The average expression of typical marker genes for monocyte subtypes is illustrated in Fig. [98]2B. In MPS II (b), monocytes showed up-regulated expression of SYNE2, TMCC3, VAV3, TBC1D9, and SSBP2 but down-regulated expression of THAP2, TNF, TGFB1, and SEC61G (Figure [99]S2E; Table S5). Mono1 (including mono1_1 and mono1_2), which mediates proinflammatory reactions such as leukocyte recruitment, cytokine and chemokine production, and leukocyte adhesion and migration, was decreased in MPS II (b) (Figure [100]S2C). Notably, mono1_2 was primarily found in the control samples and was absent in MPS II (b) (Fig. [101]2A). The mono2 subcluster, which mediates antigen clearance through antigen-antibody complex binding, the mono3 subcluster, which plays a critical role in the proliferation, differentiation, and survivorship of neutrophilic granulocytes, and the mono4 subcluster, which eliminates intracellular pathogens, were all increased in MPS II (b) (Figure [102]S2C). These dynamic changes in monocyte subtypes could be related with disease development. Fig. 2. [103]Fig. 2 [104]Open in a new tab Single-cell transcriptional profile of monocytes. (A) Umap plot of monocyte subtypes split by group. (B) Dotplot showing marker genes of monocyte subtypes (mono1: classical monocyte; mono2: nonclassical monocyte; mono3: Mono3; mono4: Mono4). (C) KEGG^[105]28 enriched terms of genes upregulated mono3. (D) KEGG^[106]28 enriched terms of genes upregulated mono1_2. (E–F) Visualization of single-cell clusters of monocyte using TooManyCells, which was split by samples and cell subtypes. Cells begin at the central node and are recursively divided based on transcriptional differences. Branch width corresponds to cell number. (G) Ridge plot of the distribution of monocyte subtype along pesudotime(top), heatmap of differential expressed genes along pesudotime trajectory(bottom). Umap plot at right showing the trajectory of monocyte subtypes analyzed by monocle. Next, pathway enrichment analysis was conducted for mono3 and mono1_2 due to their specific distribution between the healthy group and MPS II (b). Genes up-regulated in mono3 were mainly enriched in neuron-related disease pathways, fat-related pathways, and immune-related pathways (Fig. [107]2C; Table S6). Genes up-regulated in mono1_2 were enriched in neuro-related pathways such as neuroactive ligand-receptor interaction and neurodegeneration pathways, as well as immune-related pathways such as the IL-17 signaling pathway and cytokine-cytokine receptor interaction (Fig. [108]2D, Table S7), which are known to function in MPS-II^[109]6. Genes specifically expressed in MPS II (b) were involved in fat metabolism, neural diseases and pathways, and immune-related pathways (Figure [110]S2D, Table S8), potentially elucidating the amelioration of neuropsychiatric symptoms in the patient. Other studies have demonstrated the elevation of lipid peroxidation and DNA damage in MPS-II patients and the reduction of lipid peroxidation after ERT^[111]39, consistent with our data on fat metabolism pathways. The monocyte subclusters analyzed using TooManyCells^[112]40 clearly classified MPS II (b) as a separate clade, with mono3 unique to MPS II (b) and exhibiting aneuploid features (Fig. [113]2A and -E, S2-B). Pseudotime^[114]25 and RNA velocity^[115]26 analysis revealed a trajectory derived from mono1 and mono2 to mono3 and mono4 (Figs. [116]2F and [117]3-D). Dynamic gene expression along the monocyte pseudotime trajectory revealed distinct patterns. TCF7L2, GNAQ, and SLC8A1 were highly expressed at the starting and middle points of the trajectory, corresponding to the mono1 subcluster (Fig. [118]2G). Conversely, HES4, WARS, and RHOC were predominantly expressed at the beginning of the trajectory, while ADM, NLRP3, and CCL20 were highly expressed at the endpoint, associated with the mono3 and mono4 subclusters (Fig. [119]2G). Fig. 3. [120]Fig. 3 [121]Open in a new tab Single-cell transcriptional profile of macrophages. (A) Umap plot of cell subtypes of macrophages split by group. (B) KEGG^[122]28 terms of macrophage in each group. Gene ratio was indicated by the circle size and color indicated the adjusted pvalue. (C) Ridgeplot of the distribution of macrophage subtypes along pesudotime(top), heatmap of differential expressed genes along pesudotime trajectory(bottom). (D) RNA velocity analysis delineated dynamic changes in cell fate, projected onto a UMAP plot. The right illustrates re-clustered result of monocyte and macrophages, the left is RNA velocity plot of monocyte. Arrowheads indicated the predicted direction of cell development, with arrow size denoting the strength of predicted directionality. (E) Visualization of monocyte and macrophage subtypes trajectory using TooManyCells. (F) Dot plot of the predicted interactions between macrophage subtypes. CellPhoneDB^[123]24 was employed to explore cellular interactions among the monocyte subtypes. Results revealed that interactions between mono1_1 and mono2 were stronger in MPS II (b) compared to the control (Figure [124]S2I). Cellular interactions between monocyte subsets primarily involved CD74, TNFRSF1B, and C5AR1 (Figure [125]S2H). Mono1_2 mainly interacted with other monocyte subtypes through CD74 and its corresponding protein receptors, such as APP, COPA, and MIF (Figure [126]S2G). CD74 encodes a protein associated with class II major histocompatibility complex (MHC-II) and serves as a cell surface receptor for the cytokine macrophage migration inhibitory factor (MIF), which activates T and B cells when bound to the CD74 protein. This protein also suppresses the production of amyloid beta when interacting with amyloid precursor protein (APP)^[127]41. Results also showed that mono2 and mono3 interacted strongly via C5AR1-RPS19 pairs, where RPS19 acts as an immunosuppressive factor inducing the production of immunosuppressive cytokines, including transforming growth factor-β (TGF-β) in tumors^[128]42. Reducing RPS19 or blocking the C5AR1-RPS19 interaction could decrease RPS19-mediated immunosuppression^[129]42. Collectively, these results suggest that monocytes may restore immune function during ERT of MPS-II patients. Mono3, unique in MPS II (b) and found at the terminus of the cellular trajectory, may act as a pivotal therapeutic component functioning through the related pathways we discovered. Furthermore, the deficiency of mono1_2 may be a significant factor in MPS-II, as it is challenging to reactivate through therapeutic intervention. Overall, our results revealed restoration of monocytes in the recovery stage of a MPS II patient (MPS II (b)). Those monocytes were involved in inflammation, immune dysregulation, and tissue remodeling and repair process. Those results could provide potential therapeutic targets for this disease. Macrophages play an important role in MPS-II treatment Four subclusters of macrophages were identified via re-clustering analysis. Similar to monocytes, macrophages were almost absent in MPS II (a) (Figs. [130]3A, S3-A). Macrophages can be divided into two subgroups, M1 and M2, based on their localization and environment^[131]43. M1 macrophages are induced by lipopolysaccharide (LPS) and interferon (IFN)-γ and secrete proinflammatory cytokines, while M2 macrophages are involved in tissue repair and immune tolerance, with both able to transform between each other^[132]44. Our results showed that YWHAE, STX7, MYADM, and PLAC8 were up-regulated in the control group and MPS II (b) but down-regulated in MPS II (a). Conversely, PLEK, RNF, NINJ1, and PCNT were up-regulated in MPS II (a) but down-regulated in the control group and MPS II (b) (Figure [133]S3D, Table S-9). Dynamic expression of these genes indicated their response to ERT. Pathway analysis of macrophages indicated that MPS II (a) was involved in fatty acid metabolism and biosynthesis and the PPAE and NF-κB signaling pathways, while MPS II (b) and the control group were enriched in the PI3K-Akt signaling pathway, which was lacking in MPS II (a) (Fig. [134]3B, Table S-10). Trajectory analysis of M1 and M2 macrophages revealed that CITED2, IL32, and MAL were highly expressed in M2, while CASC15, MYL9, and THBS1 were up-regulated in M1 (Fig. [135]3C and -D, S3-C). Among macrophage subtypes, interactions of M2-M2 and M2-M1_2 increased significantly in MPS II (b) compared to MPS II (a) and the healthy controls, while interactions between M2 and M1_2 were weaker in MPS II (a) than in MPS II (b) and the healthy controls (Figure [136]S3E). These cellular interactions of macrophage subtypes revealed that M1_2 and the M2 were strongly connected through PF4-CXCR3 pairs, a connection not observed in the control or MPS II (a) (Fig. [137]3F). CD74 and its corresponding receptors, including MIF, COPA, and APP, were strongly associated with M2 and other macrophage subtypes in the control and MPS II (b) groups, while these associations were absent in MPS II (a) (Fig. [138]3F). Overall, these findings suggest that macrophages may play a role in the pathology of MPS-II, which could be considered as a potential therapeutic target to mitigate disease progression and improve patient outcomes. Both monocytes and macrophages exhibited phagocytic function, with similar distributions in the MPS-II patient. To investigate the relationship between macrophages and monocytes, joint trajectory analysis was performed. RNA velocity analysis of the two cells indicated potential trans-differentiation from mono1 to mono3 (Fig. [139]3D). TooManyCells analysis^[140]40 also showed that the macrophages were significantly distinct from the monocytes, while mono3 and mono2 were separated from the distinct mono1 branch (Fig. [141]3E). Collectively, the deficiencies of macrophages and monocytes appear to be a crucial factor in MPS-II. These two cell types may function through biological pathways deficient in MPS II (a) but restored in MPS II (b) and the control group. Additionally, ligand-receptor pairs interacting between cells may play a potential role in aiding the recovery of MPS-II. Characterization of T cell diversity and clonal expansion in MPS-II A total of 39 454 T cells were obtained from the scRNA-seq data, annotated into 14 subtypes and one cluster of NKT cells based on canonical markers (Figs. [142]4A and B, S4-A, S4-B). The identified subtypes included CD4 + Th22, CD4 + memory T cells (CD4 Tm), CD4 + regulatory T cells (CD4 Treg), four clusters of CD4 + naïve T cells (CD4 Tn), CD8 + Tc17, CD8 + central memory T cells (CD8 Tcm), three clusters of CD8 + effector memory T cells (CD8 Tem), and two clusters of CD8 + cells (CD8 Tn). After ERT, the proportions of two naïve T cell subclusters (CD4 Tn_1, CD8 Tn_1) rebounded. To further explore expression changes in these two naïve T cell subtypes, we identified DEGs in these cells among the three groups (Figure S4-C, S4-D). In the CD4 Tn_1 subtype, TUBB, UCP2, WARS, and TYROBP were down-regulated in MPS II (a) but up-regulated in MPS II (b) and the control, while ZFP36, TUBB4B, YWHAE, and YES1 were up-regulated in MPS II (a) but down-regulated in MPS II (b) and the control (Figure S4-C). These genes may play a role in disease development. In the CD8 Tn_1 subtype, TUBA1B, ZNF331, and TRBV were up-regulated in MPS II (a) but down-regulated in MPS II (b) and the control group, while ZYX, VDAC1, and TRBV5 were down-regulated in MPS II (a) but up-regulated in MPS II (b) and the control group (Figure S4-D). The top 15 DEGs among the three groups are shown in Figure S4-E (Table S11). Pathway enrichment analysis of T cells revealed that MPS II (a) was enriched in neurological disorders, while MPS II (b) was enriched with neuroactive-related pathways (Fig. [143]4C, Table S12). These results may help explain the improvements in clinical neurological manifestations observed in the patient. Fig. 4. [144]Fig. 4 [145]Open in a new tab Single-cell transcriptional profile of T cells and NK cells. (A) UMAP plot of cell subtypes of T cell. (B) Barplot of the percentageof T cell subtypes in each sample. (C) Bubble plot showing gene ontology (GO) terms of each group. Gene ratio was indicated by the circle size and color indicated the adjusted pvalue. (D–E) Ridgeplot shows the distribution of subtypes of CD4 + T and CD8 + T cell along pesudotime(top), heatmap of differential expressed genes along pesudotime trajectory(bottom). (F) RNA velocity plot of T cell. (G) Umap plot of the distribution of TCR clonotype split by group. (H) Alluvial plot illustrating amino acid sequence VDJC genes clonotype changes of TCR between samples. To further delineate the transition states associated with T cells, the trajectories of the CD4 + and CD8 + T cell subtypes were analyzed using Monocle2^[146]25 (Figure S5-G, S5-F). CD4 + and CD8 + naïve T cells were positioned at the starting point of the trajectory path, whereas CD4 Th22, CD4 memory T cells, and two clusters of CD8_Tem were at the terminus. The transcriptional changes related to the transitional states of CD4 and CD8 T cells were then investigated. The CD4 + and CD8 + T cells were categorized into three phases (Fig. [147]4D and -E). In CD4 + T cells, phase 1 was characterized by two clusters of CD4 naïve T cells with up-regulated ITM2A, CHI3L2, SOX4, and LRRN3 expression. Phase 2 was associated with the up-regulation of KLRF1, FGR, TYPOBP, and FCRL3, predominantly featuring CD4 Tm. Phase 3 consisted mostly of CD4 Th22 cells with up-regulated FAM45A, MT2A, TNFRSF4, and FOXP3 expression (Fig. [148]4D). For CD8 + T cells, phase 1 showed up-regulation of BCL3, NR4A1, and PLK2, dominated by CD8 naïve T cells. The second trajectory phase exhibited high expression of MYC, SOX4, AIF1, and AQP3 (Fig. [149]4E). RNA velocity analysis indicated directional flows from CD4 Treg to CD4 Tm cells, and from CD8 Tcm to CD8 Tem cells (Fig. [150]4F). Cellular communication analysis of different T cell subtypes revealed enhanced interactions between CD4 Tn_3 and CD4 Th22 at the MPS II (a) stage, which were weaker in the control group and MPS II (b). Conversely, interactions between CD8 Tm_1 and CD8 Tc17 increased in MPS II (b) compared to the healthy control but decreased in MPS II (a) (Figure S4-H, S4-I). To determine whether the T-cell repertoire is heterogeneous between the two treatment stages, scRNA-seq was combined with TCR-seq data, yielding 30 358 T cells (Fig. [151]4G). Expanded T cell clones were predominantly found in the CD8 Tem_3 and Tem_1 subclusters (Figure S4-J). The top 14 genes in the MPS II (a) clonotypes comprised most of those in MPS II (b) (Fig. [152]4H). Notably, 75% of CD8 Tem_3 cells were substantially expanded (containing large clonotypes), indicating high clonality (Figure S4-J). T cell clonal expansion mainly occurred in CD8 Tem cells, with significant expansion of three clonotypes after ERT, including RAV1-2.TRAC_TRBV20-1.None.TRBJ2-2.TRBC2T, TRAV14/DV4.TRAJ34.TRAC_TRBV19.None.TRBJ1-1.TRBC1, and TRAV29-/DV5. TRAJ17.TRAC_TRBV2.None.TRBJ1-1-2.TRBC1 (Fig. [153]4G, H, S4-J). Clonal diversity of TCR-seq data demonstrated that the two treatment stages of the MPS-II patient were significantly distinct (Figure S4-K). Overall, these findings suggest that T cells play a vital role in antigen recognition, T cell activation, and adaptive immune response in our MPS II patient. Characterization of B cell diversity in MPS-II Re-clustering of the B cell compartment identified six subclusters with recognized cell markers, with the B1/2/5 subclusters showing marked declines in MPS-II (Fig. [154]5A, S5-A, S5-B, S5-C). Three naïve B cell subtypes were clustered together, with one subtype enriched at the MPS II (a) stage and decreased in the control and MPS II (b) stages, showing up-regulation of PIK3AP1, LYST, and GNG5 (Figure S5-D). Further pathway enrichment analysis indicated that MPS-II was enriched in ubiquitin-mediated proteolysis, the PI3K-Akt signaling pathway, and neurodegeneration-related pathways, while MPS II (a) was enriched in nervous system disease pathways (Fig. [155]5B, Table S13). Trajectory analysis showed that naïve B-B1 cells were positioned at the beginning of the trajectory path with up-regulation of GLIPR1, FAM111B, and GBP4, whereas naïve B-B2/3 cells were located at the terminus with high expression of LAIR1, SH3BP2, FCRLA, and PLD4 (Fig. [156]5C, S5-F). Pseudotime analysis indicated a flow from naïve B cells to memory B cells and plasmablasts via scVelo^[157]24 (Fig. [158]5D). Ligand-receptor interaction analysis revealed that classical memory B cells (B4) strongly interacted with naïve B cell cluster two (B2) in the control group and MPS II (b) but showed weaker interactions in MPS II (a) (Figure S5-E). In MPS II (b), classical memory B cells displayed stronger interactions with naïve B cell cluster three (B3) than in the other two groups, as well as more frequent interactions between naïve B cell subclusters. Further exploration of ligand-receptor pairs engaged in interactions between B cell subtypes (Fig. [159]5E) showed that cellular interactions were primarily through CD74 and its corresponding proteins (Fig. [160]5E). Fig. 5. [161]Fig. 5 [162]Open in a new tab Single-cell transcriptional profile of B cells. (A) Umap plot of cell subtypes of B cell split by groups. (B) Bubble plot showing KEGG^[163]28 terms of each group. Gene ratio was indicated by the circle size and color indicated the adjusted pvalue. (C) Ridgeplot of the distribution of B cell subtype (top) and heatmap of differential expressed genes (bottom) along pesudotime trajectory. (D) RNA velocity plot of B cell. (E) Dot plot of the predicted interactions between B cell subtypes in different group. Interactions are indicated by the color. (F) Umap plot illustrating the distribution of BCR clonotype. (G) Umap plot of the distribution of BCR clonotype split by group. (H) Alluvial plot illustrating amino acid sequence VDJC genes clonotype changes of BCR between samples. To evaluate the heterogeneity of the B cell repertoire between the two treatment stages, scRNA-seq was combined with BCR-seq data, yielding 6 972 B cells. Expanded B cell clones were predominantly found in naïve B cells and B cells from MPS II (b) (Fig. [164]5G and H). Both MPS-II stages exhibited clonal expansion, with MPS II (b) showing less intense expansion at the end of the treatment stage (Fig. [165]5F and G). Only two clonotypes were found in both treatment stages, which decreased at the end of the treatment stage, while each stage contained various unique clonotypes (Fig. [166]5H). The B cell clonotype IGH.185.IGHV1-2IGLC: LD.2.IGLV2-23 shrank following ERT, while NA_NA_IGLC: LD.1.IGKV3-15 was only present in MPS II (a). Diversity analysis showed that B cells in MPS II (b) exhibited a higher diversity index score compared to the other groups (Figure S4-K). These clonotypes likely play a crucial role in the ability of B-cells to recognize and respond to specific antigens, thereby contributing to the adaptive immune response. ERT influences metabolism and activates the ligand-receptor component for cell-cell communication in MPS-II Cell-cell interactions mediated by ligand-receptor pairs expressed in different cell types in the three groups were estimated using CellPhoneDB (Fig. [167]6A). The number of interactions between different cell types was similar between the control group and MPS II (b), both of which were stronger than those in MPS II (a). Notably, the interactions between NK, NKT, and CD14 + monocytes increased significantly in MPS II (b) compared to the control group. In contrast, the cellular interactions between macrophages and other PBMCs were not particularly close and remained unchanged across the three groups. Fig. 6. [168]Fig. 6 [169]Open in a new tab Cellular interactions between PBMC cell types. (A) Number of cell-cell interactions between each cell types in each group. (B) KEGG pathway dotplot of health control, MPS1, and MPS2. (C–E). Dot plot of the predicted interactions between each cell types in different group. Interactions are indicated by the color. (C) ligan-receptor pairs between cells except monocyte and macrophage; (D) ligan-receptor pairs between monocyte and other cell types; (E) ligan-receptor pairs between macrophage and other cell types. (F) Predicated cell transformation and cellular interactions through potential ligand-receptor pairs. Monocytes and macrophages were specifically scarce in MPS II (a), indicating a potential mechanism involving interactions between these cells and others that impact therapeutic responses during ERT in MPS-II. To investigate this, we first explored the differences in enriched pathways between different treatment stages. Notably, the Wnt, prolactin, and adipocytokine signaling pathways were enriched in MPS II (a) but absent in the other stages (Fig. [170]6B). Genes related to the TNF and JAK-STAT signaling pathways were more prevalent in MPS II (a) compared to MPS II (b) (Fig. [171]6B), consistent with the increased plasma and serum levels of TNF-α cytokines in MPS-II patients^[172]45, demonstrating the effectiveness of ERT. Dysregulation of the JAK-STAT signaling pathway is associated with various cancers and autoimmune diseases^[173]46. Additionally, the FoxO signaling pathway, which is important in metabolism, cellular proliferation, stress resistance, and apoptosis, was enriched in MPS II (a) but absent in the control group and MPS II (b) (Fig. [174]6B). Neural-related pathways, such as the sphingolipid signaling pathway, serotonergic signaling pathway, neuroactive ligand-receptor interaction, neurotrophin signaling pathway, glutamatergic synapse, and GABAergic synapse, were enriched in MPS II (b) (Fig. [175]6B), consistent with the improved neurological symptoms observed in the patient. To further explore the cellular interactions between monocytes and macrophages with other cells, receptor-ligand interactions between different cell types were predicted (Fig. [176]6C, D and E). The interaction of NK cells with other immune cells was primarily mediated by HLA-E and their corresponding protein receptors, which were enhanced in MPS II (b) but absent in MPS II (a) and the control group (Fig. [177]6C). CD74 and its corresponding receptors, mediating interactions between B cells and T cells, exhibited higher expression in MPS II (b) compared to MPS II (a) and the control group. Interactions between monocytes and other cell types were mainly mediated by ILB1, ANXA1, LGALS9, and their corresponding protein receptors, which are associated with anti-inflammatory activity (Fig. [178]6D). The IL1B-ADRB2 and C5AR1-RPS19 pairs were significantly enriched in monocyte and other cell types. Animal studies have shown that activation of ADRB2 can attenuate monocyte activation and proinflammatory and pro-fibrotic responses^[179]47. C5AR1 is an upstream signal in the Toll-like pathway, which can activate downstream inflammatory cytokines^[180]48, such as TNFRA family members (TNFAIP6, TNFRSF1B, and TNFSF10) and IL family members (IL1B, IL7R, and IL18R1)^[181]34,[182]35. Macrophages were primarily associated with B cells through CD74 across all three groups and with NK cells through PPBP in MPS II (a) and MPS II (b) (Fig. [183]6E). In conclusion, the transformation of mono3/4 from mono1/2 and their interactions with NK cells through the IL1B-ADRB2 pair and B cells through the TNFSF13B-HLA-DPB1 pair were revealed in our results. Although only one subject remains controversial, our results also could propose a possible atlas of cellular interaction network during ERT in MPS II (Fig. [184]6F). Discussion In our study, we explored the transcriptional and genomic changes of MPS-II patients during ERT. Our findings revealed up-regulation of TNFAIP3 in MPS II (a), the early stage of ERT, and down-regulation of this gene in the end stage of ERT. The protein encoded by TNFAIP3 inhibits NF-κB activation and TNF-mediated apoptosis^[185]49. Accumulation of the GAG heparan sulfate in conditions such as MPS-I and MPS-II may trigger the production and release of proinflammatory cytokines like TNF-α^[186]50,[187]51. Elevated TNF-α levels were overserved in MPS VII and related with severe somatic pain^[188]52. Prior research has also demonstrated that anti-TNF-α is effective in human studies of MPS I and MPS II, and in mouse models of MPS VI and MPS VII^[189]53,[190]54. The transcriptional changes in TNFAIP3 during different ERT stages may be associated with our patient’s improvement. Genes up-regulated in MPS II (b) and the control group but down-regulated in MPS II (a), such as TSPO, TKT, WARS, and TYMP, may also play a role in the therapeutic response to ERT. MPS-II results from a deficiency of iduronate-2-sulfatase encoded by the IDS gene. Our research showed that genes involved in pathways related to IDS, such as CLPX, GUSB, and DSC3, exhibited changes at different treatment stages, suggesting potential as therapeutic targets for MPS. Furthermore, our findings indicated that monocytes and macrophages were notably absent or scarce in MPS II (a) (the severe stage), likely due to their migration from peripheral blood to tissues with accumulated “GAGs” to eliminate these deposits. This suggests that these cell types may represent therapeutic targets during ERT. The mono3 subcluster was unique to MPS II (b), located at the terminal point of the trajectory from mono1 and mono2, suggesting that the transition of monocytes is closely associated with disease recovery. Genes up-regulated in mono3 were primarily enriched in neuron-related pathways, including neurodegeneration and Alzheimer’s disease, immune-related pathways, including T cell differentiation and antigen processing, and fatty-related pathways, which may be related to the improvements observed in the neurological and immune systems of the patient. CD4 naïve T cells were reduced in MPS II (a) and MPS II (b) compared to the healthy controls, possibly due to the activation of naïve T cells by metabolic dysfunction of “GAGs” and infection in the patient, or developmental disability of thymus T cells caused by the disease itself. Targeted treatment of inflammation to attenuate neurodegeneration may represent a novel therapeutic direction for patients undergoing ERT, which has limited effects on the nervous system. One subtype of mono1 (mono1_2) was scarce in MPS II (b), showing enrichment in genes involved in the Toll-like receptor signaling pathway, TNF signaling pathway, and IL-17 signaling pathway. T cells in MPS II (b) also showed enrichment in genes involved in the Toll-like receptor and TNF signaling pathways. Microtubulation of phagosomes and lysosomes induced by Toll-like receptor 4 (TLR4) signal transduction plays an important role in antigen presentation^[191]50. The accumulation of GAGs can overactivate TLR4, leading to the up-regulation of TNF-α and IL-1β, which are significantly down-regulated upon TLR4 inhibition, indicating that the TLR4-mediated inflammatory response may play a role in MPS-IIIA disease^[192]52,[193]55,[194]56. Elevated TNF-α levels are associated with severe somatic pain and impaired physical function in patients with MPS-I, -II, and -VI^[195]57. IL-1 can enhance inflammatory responses and neurodegenerative diseases^[196]58. GAGs bind to TLR4 receptors and activate the TLR4 pathway, leading to the release of cytokines such as TNF-α and IL-1β. Inactivation of the TLR4 pathway in MPS-VII mice lacking TLR4 alleviates clinical and pathological features, while anti-TNF-α drug administration in MPS-VI rats reduces inflammatory cytokine levels and improves joint pathology^[197]59. Furthermore, treatment with pentosan polysulfate (PPS), an anti-TNF-α medication, can improve joint motion range in adult patients with MPS-II^[198]60. In conclusion, our findings suggest that the TLR4/TNF-α signaling pathway may be implicated in MPS-related osteoarthropathy, and the TLR4/IL-1 signaling pathway may play a role in neurodegeneration. Interactions between monocytes and other cell types increased in MPS II (b), particularly with NK and NKT cells, primarily mediated through IL1B-ADRB2 pairs. Interactions between B cells and monocytes were predominantly mediated by HLA-DPB1-TNFSF13B pairs, which also increased in MPS II (b), while monocytes and T cells connected through C5AR1-RPS19 pairs. Interactions between monocytes and macrophages were closer than with other cells, mainly mediated through ADORA2A-NAMPT pairs that only existed in MPS II (b). The mono3 subtype of monocytes, which was unique to MPS II (b), could transform from other monocyte subtypes (mono1 and mono2). The connection with mono2 through C5AR1-RPS19 pairs may play a role in the function of ERT, contributing to prognosis. However, our studies have certain limitations, included small sample size, lacking of long-term observation, and incomplete clinical data of biomarkers (GAGs, pro-inflammatory factors, etc.) from different specimens (urine, blood, and CSF). Considering the broad heterogeneity within MPS, more samples of different types of MPS should be brought into our future studies. Conclusion Our study presents a comprehensive transcriptional and genomic atlas of MPS-II, utilizing multi-omics approaches and scRNA-seq to investigate dynamic changes during ERT treatment. Our findings highlight the crucial roles of monocytes and macrophages in ERT, with their restoration at the end stage suggesting involvement in pathways such as NOD-like receptor and PPAR signaling, and interactions with NK cells, T cells, and B cells to improve prognosis. However, the lack of patient samples and animal models limits further exploration of the detailed molecular mechanisms underlying MPS-II pathology. Future research should focus on enrolling more patients and developing animal models to enhance our understanding of MPS-II. Electronic supplementary material Below is the link to the electronic supplementary material. [199]Supplementary Material 1^ (1.6MB, pdf) [200]Supplementary Material 2^ (60.3KB, pdf) [201]Supplementary Material 3^ (112.6KB, xls) Acknowledgements