Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Jun;15(6):1431-1446.
doi: 10.1038/s41385-022-00572-1. Epub 2022 Oct 27.

Hermansky-Pudlak syndrome type 1 causes impaired anti-microbial immunity and inflammation due to dysregulated immunometabolism

Affiliations

Hermansky-Pudlak syndrome type 1 causes impaired anti-microbial immunity and inflammation due to dysregulated immunometabolism

Athena Cavounidis et al. Mucosal Immunol. 2022 Jun.

Abstract

Hermansky-Pudlak syndrome (HPS) types 1 and 4 are caused by defective vesicle trafficking. The mechanism for Crohn's disease-like inflammation, lung fibrosis, and macrophage lipid accumulation in these patients remains enigmatic. The aim of this study is to understand the cellular basis of inflammation in HPS-1. We performed mass cytometry, proteomic and transcriptomic analyses to investigate peripheral blood cells and serum of HPS-1 patients. Using spatial transcriptomics, granuloma-associated signatures in the tissue of an HPS-1 patient with granulomatous colitis were dissected. In vitro studies were conducted to investigate anti-microbial responses of HPS-1 patient macrophages and cell lines. Monocytes of HPS-1 patients exhibit an inflammatory phenotype associated with dysregulated TNF, IL-1α, OSM in serum, and monocyte-derived macrophages. Inflammatory macrophages accumulate in the intestine and granuloma-associated macrophages in HPS-1 show transcriptional signatures suggestive of a lipid storage and metabolic defect. We show that HPS1 deficiency leads to an altered metabolic program and Rab32-dependent amplified mTOR signaling, facilitated by the accumulation of mTOR on lysosomes. This pathogenic mechanism translates into aberrant bacterial clearance, which can be rescued with mTORC1 inhibition. Rab32-mediated mTOR signaling acts as an immuno-metabolic checkpoint, adding to the evidence that defective bioenergetics can drive hampered anti-microbial activity and contribute to inflammation.

PubMed Disclaimer

Conflict of interest statement

H.H.U. received research support or consultancy fees from UCB Pharma, Eli Lilly, Boehringer Ingelheim, Pfizer, Celgene, and AbbVie. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR, or the Department of Health. A.C. and S.P. are employees of GSK and MC is an employee of SenTcell L.t.d. D.A. is an employee of Novartis Pharma AG.

Figures

Fig. 1
Fig. 1. Mass cytometry of HPS-1 PBMCs identifies distinct inflammatory monocyte populations.
a FlowSOM analysis of CyTOF data defines 100 cell clusters (individual nodes) organized into 13 metaclusters (node number and color) for 12 controls and 8 HPS-1 patients. We denoted clusters with surface markers that resembled particular cell types. b Specific metacluster 11 and metacluster 5 cell clusters are significantly associated with HPS-1 patients. Metacluster 11.1 was defined by CXCR3, CD64++, CD62L++, metacluster 11.2 by Beta7++, CD64++, CD62L++ and 11.3 by CD16. c Principal component analysis of FlowSOM clusters separates HPS-1 patients and healthy controls. d viSNE analysis of CD14+ monocytes defines distinct populations of monocytes in patients with HPS-1 highly expressing CD62L and CD64. e Histogram of marker expression in patients with HPS-1 or Healthy Controls. f Heatmap with hierarchical clustering of inflammatory proteins of O-link inflammation panel for 50 controls, 5 HPS-1, 2 HPS-1 PF, and 4 HPS-1 IBD patients. g Dot plots of IL-1α and TNF protein expression. h Principal component analysis of the O-link inflammatory panel. For CyTOF analysis, populations are compared with an unpaired t-test with Bonferroni-Dunn correction for multiple comparisons. Data are mean+/− SEM. *P < 0.05 **P < 0.01 ***<0.001. All P values are adjusted. For O-link analysis, the Benjamini, Krieger, and Yekutieli multiple nonparametric t-test was performed, ***adjusted p < 0.001, ****adjusted p < 0.0001.
Fig. 2
Fig. 2. Spatial transcriptomics uncovers granuloma-associated signatures in the intestine of an HPS-1 patient.
a HPS-1 gastrointestinal tissue used for spatial transcriptomics and immunofluorescently labelled for DNA, CD3, and CD68 staining. b Principal component analysis reveals cellular populations and anatomical regions as main drivers of variation. c Cell deconvolution of spatial transcriptomic profiles, mapped against a selection of cellular profiles provided by human adult gut scRNAseq atlas from the Human Cell Atlas project. d Heatmap of differentially expressed genes (p < 0.001) across the granuloma core, granuloma periphery and mucosa for CD68+CD3− CD45+cells. e HPS1 and HPS4 expression in different immune cell types (n = 4–16). Immune cells from peripheral blood were FACS sorted. Monocytes and monocyte-derived macrophages were isolated using CD14 beads and macrophage differentiation involved M-CSF treatment of monocytes for 5 days. Dot plot, mean and SEM are provided.**p < 0.01, ****p < 0.0001; Statistical significance was determined using a Tukey multiple comparisons test on log10-transformed data.
Fig. 3
Fig. 3. HPS1 deficiency leads to a dysregulated metabolic program.
a Differential expression of HPS-1 and control macrophages. Known IBD-related cytokines are bolded. B Weighted Gene Correlation Network Analysis reveals a module highly associated with the HPS-1 disease trait; ClueGO was performed on the top 200 hub genes of this module. c Heatmap of the genes contributing to the LDL particle receptor catabolic signature. d Gene set enrichment analysis using hallmark pathways reveals a reduction in fatty acid metabolism. e Gene set enrichment analysis highlights decreased oxidative phosphorylation. f Sanger sequencing of HPS1 knockout HAP1 cells and control as a comparison. g Oxygen consumption rate of control and HPS1 knockout cells. Error bars in SEM. 12 replicates from one experiment, representative of two independent experiments. h MitoSOX staining on control and HPS1 knockout cells (n = 6). i Serum-starved HAP1 cells were treated with 2.5 μg/mL LDL-BODIPY for 3 h (n = 7). j HAP1 cells were stimulated with cholesterol for 2 h and stained with BODIPY 493/503 (n = 5). *p < 0.05, **p < 0.01, ****p < 0.0001; Ratio paired t-test on mean fluorescence intensity. Unpaired t-test on OCR log-transformed data. Padj = adjusted p-value; NES normalized enrichment score, R & A rotenone and antimycin A.
Fig. 4
Fig. 4. Rab32-mediated enhanced mTORC1 signaling in HPS1 deficiency.
a Cholesterol stimulation was performed for 2 h prior to staining with BODIPY 493/503 in patient monocyte-derived macrophages (n = 5). Representative flow cytometry histogram plots. b Schematic of lysosomal immunoprecipitation and confocal microscopy of TMEM192-3xHA and LysoTracker (LT) in HAP1 cells. Lysosomal immunoprecipitation in cells approach was assessed through LAMP2 and CTSC Western blot. c LysoIP proteomics (n = 3) and Western blotting of lysosomal immunoprecipitation samples for mTOR, LAMP2 and TMEM192-3xHA tag (n = 4). d HAP1 cells were serum-starved overnight and treated with 50 μM rapamycin for 3 h prior to pS6 staining (n = 4). e Quantification of Western blot of control and HPS-1 patient macrophage samples for pS6 and GAPDH (n = 3 for healthy controls and HPS-1 patients). Quantification was performed using ImageJ. Western blot image used for quantification. f HAP1 cells were treated with 25 μg/mL LDL for 1 h and then stained for pS6 (n = 3). g LDL uptake in HAP1 cells using LDL-BODIPY for 1 h at 25 μg/mL were pre-treated with 50 μM rapamycin for 3 h (n = 4). h HAP1 cells were transfected with siRNA against Rab32 or a control pool and probed for S6 phosphorylation (n = 5). Statistics: Unpaired parametric test on log10-transformed mean fluorescence intensity data for a. Paired t-test on ratio data for c. Ratio paired t-test on mean fluorescence intensity (d, f, g). An unpaired t-test on ratio data was performed for the pS6/GAPDH quantification.
Fig. 5
Fig. 5. mTORC1 inhibition and fatty acids rescue HPS1 bacterial clearance defects.
a HAP1 cells were treated with 10 nM bafilomycin A1 for 2 h and the FlowCellect autophagy kit was used (n = 5). b The gentamicin protection assay was performed in HAP1 cells using GFP-Salmonella Typhimurium (n = 8), Adherent Invasive E. coli (n = 3) or Staphylococcus aureus (n = 4). c Gentamicin protection assay of healthy donor and HPS-1 patient-derived macrophages (n = 10). Representative agar plate of gentamicin protection assay. d Quantification of number of bacteria in each cell using confocal microscopy in control and HPS-1 macrophages (7 controls, 6 HPS-1 patients). The average bacteria per cell in each donor was used for statistical tests, where we performed an unpaired t-test. Representative confocal images; scale bar 5 μm. White asterisk denotes DAPI + GFP- bacteria; yellow asterisk DAPI + GFPbright bacteria; purple asterisk DAPI + GFPdim bacteria. e Differential expression of HPS1 patient and control macrophages following Salmonella Typhimurium infection (n = 4). f HAP1 cells were stimulated with 50 μM cholesterol for 2 h prior to the gentamicin protection assay (n = 5). g HAP1 cells were treated with free fatty acids (FFA) for 2 h prior to a gentamicin protection assay (n = 5). h HAP1 cells were treated with 50 μM rapamycin for 3 h followed by the gentamicin protection assay (n = 4, Salmonella Typhimurium). i Macrophages were treated with 50 μM rapamycin for 2 h prior to Salmonella Typhimurium infection and the gentamicin protection assay (n = 6); each dot represents one patient. Significance was determined using a paired t-test on the autophagy flux values. For gentamicin protection assays, we used a ratio paired t-test for HAP1 cells and an unpaired t-test on log-transformed data for patient macrophages. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Fig. 6
Fig. 6. Summary of intestinal inflammation and molecular mechanisms in HPS1 deficiency.
In health, monocytes from the blood differentiate into macrophages, which interact with regulatory T cells and fibroblasts, promoting homeostasis. On a molecular level, lipid droplet metabolism and lower mTORC1 levels facilitate the degradation of microbes, where Rab32 plays a key role in phagosome maturation. In HPS1 deficiency, activated monocytes are recruited from the blood, differentiating into macrophages that secrete inflammatory mediators such as TNF, IL-1, OSM and PTGS2. On an intracellular level, high levels of mTORC1 activity mediated through Rab32 and deregulated immune metabolism dampen anti-microbial activity. The suppressed degradation of bacteria and increased cytokine production in HPS-1 patients can lead to tissue inflammation involving crypt abscesses, epithelial barrier damage, and fistulizing disease. Figure elements are derived from Servier Medical Art.

References

    1. Dharmasiri S. et al. Human intestinal macrophages are involved in the pathology of both ulcerative colitis and Crohn disease. Inflamm. Bowel Dis.27, 1641–1652 (2021). - PMC - PubMed
    1. Heller S, et al. Intestinal inflammation requires FOXO3 and prostaglandin E2-dependent lipogenesis and elevated lipid droplets. Am. J. Physiol. Gastrointest. Liver Physiol. 2016;310:G844–G854. doi: 10.1152/ajpgi.00407.2015. - DOI - PMC - PubMed
    1. Singh A., Sen P. Lipid droplet: A functionally active organelle in monocyte to macrophage differentiation and its inflammatory properties. Biochim Biophys Acta. Mol. Cell Biol. Lipids1866, 158981 (2021). - PubMed
    1. Gahl WA, et al. Genetic defects and clinical characteristics of patients with a form of oculocutaneous albinism (Hermansky-Pudlak syndrome) N. Engl. J. Med. 1998;338:1258–1264. doi: 10.1056/NEJM199804303381803. - DOI - PubMed
    1. Huizing M, et al. Hermansky-Pudlak syndrome: Mutation update. Hum. Mutat. 2020;41:543–580. doi: 10.1002/humu.23968. - DOI - PMC - PubMed

Publication types

Supplementary concepts

LinkOut - more resources