Graphical abstract graphic file with name fx1.jpg [71]Open in a new tab Highlights * • rs194800 at 16p21 is associated with MAC disease susceptibility and PRKCB expression * • PRKCB exacerbates M. avium infection and promotes intracellular survival of M. avium * • PRKCB inhibits the fusion between lysosomes and mycobacterial phagosomes * • PRKCB inhibitor has therapeutic potential against M. avium infection __________________________________________________________________ Zheng et al. identify that 16p21 is the susceptibility locus for pulmonary MAC disease, and the risk allele of rs194800 promotes PRKCB expression. PRKCB blocks the fusion between lysosomes and mycobacterial phagosomes to promote the intracellular survival of M. avium, thus increasing susceptibility to MAC disease. Introduction Non-tuberculous mycobacterial (NTM) pulmonary disease (NTM-PD) is a severe progressive illness caused by non-tuberculous mycobacteria that is increasing in incidence and prevalence worldwide.[72]^1^,[73]^2^,[74]^3 Similar to the case for tuberculosis, NTM disease requires complicated treatment lasting more than 12 months. Unfortunately, treatment outcomes are unfavorable due to these bacteria responding poorly to multiple anti-mycobacterial drugs. There is thus a need to elucidate the pathogenesis and discover therapeutic strategies to control NTM infection. Non-tuberculous mycobacteria are ubiquitous environmental microorganisms, the human exposure to which is likely to be widespread. However, only a fraction of the population will develop disease related to these bacteria. This suggests that, in addition to microbial pathogenic or environmental factors, host factors also play a crucial role in the development of NTM disease. Previous studies have reported that underlying structural lung diseases are host risk factors for NTM disease.[75]^4^,[76]^5^,[77]^6 Nevertheless, many patients suffering from NTM disease had no known risk factors, implying that host genetic factors are also involved in the pathogenesis of NTM disease. To date, only two genome-wide association studies (GWASs) have been conducted in NTM-PD, one in the Japanese population and the other in the Korean population.[78]^7^,[79]^8 This indicates a pronounced paucity of GWAS in this disease context compared with those in the two other mycobacterial diseases tuberculosis[80]^9^,[81]^10^,[82]^11^,[83]^12^,[84]^13 and leprosy.[85]^14^,[86]^15^,[87]^16^,[88]^17^,[89]^18 Meanwhile, family-based whole-exome sequencing revealed an association between the TTK protein kinase gene (TTK) and susceptibility to NTM-PD.[90]^19^,[91]^20 However, in these published studies, only a few loci with genome-wide significance were identified and no follow-up functional studies on possible causal genes were performed. It is thus valuable to conduct a GWAS for NTM-PD in diverse populations to obtain a deeper understanding of the genetic basis of NTM-PD. In this work, we performed a two-stage GWAS in the Chinese population and discovered a significant susceptibility locus at 16p21 (rs194800, p[meta] = 7.85 × 10^−13) associated with NTM-PD, and the subtyping analysis showed that the 16p21 locus retained genome-wide significance mainly in Mycobacterium avium complex (MAC) patients. The rs194800 variant regulated the expression of the protein kinase C beta (PRKCB) gene and was associated with the severity of NTM-PD. Furthermore, Prkcb-deficient mice were found to be more resistant to M. avium infection. In vitro, Prkcb was shown to enhance the intracellular survival of M. avium by inhibiting the fusion between lysosomes and mycobacterial phagosomes in macrophages. The inhibition of PRKCB by inhibitors impaired M. avium growth in vitro and in vivo. Our findings offer valuable insights into the genetic causes and pathogenesis of MAC disease and provide a promising target for host-directed therapy for MAC disease. Results GWAS To identify loci conferring susceptibility to NTM-PD in the Chinese population, we performed genome-wide analysis of 430 NTM cases and 475 healthy controls (discovery stage, [92]Table S1). Here, 51 variants at chromosome 16p21 reached genome-wide significance (p < 5 × 10^−8, [93]Figures 1A and 1B). We next took forward the most significant independent variants (p < 1 × 10^−5, [94]Table S2) for replication in a sample of 202 NTM cases and 319 healthy controls ([95]Tables S1 and [96]S2). Meta-analysis of the discovery and replication datasets identified one locus with genome-wide significance at chromosome 16p21 indexed by rs194800 (p[meta] = 7.85 × 10^−13; odds ratio [OR] = 2.02, [97]Tables S2 and [98]S3), which is in the intergenic region between CHP2 and PRKCB ([99]Figure 1C). Further fine-mapping of the 16p21 region using the sum of single effects (SuSiE) method identified 22 variants with a posterior inclusion probability (PIP) greater than 0.01 ([100]Table S4).[101]^21 Among these, rs194800 had the highest PIP of 0.045, suggesting its potential role in disease susceptibility. After adjusting for the effects of confounding variables (age, gender, and BMI), rs194800 remained significant in the NTM-PD, MAC, and M. abscessus (MAB) analyses ([102]Table S5). We also identified a suggestive locus (p < 5 × 10^−6) at chromosome 2p24.2 indexed by rs2881207 (p[meta] = 3.70 × 10^−6; OR = 0.34), which is located in the downstream region of the RDH14 gene ([103]Figure S1A). No evidence for heterogeneity across stages (p[het] > 0.05) was observed for either rs194800 or rs2881207 ([104]Table S2). In addition, the suggestive signals on chromosome 6p21.32 implicated the human leukocyte antigen (HLA) region as a potential risk factor for NTM-PD ([105]Figure S1B). Figure 1. [106]Figure 1 [107]Open in a new tab Plots showing putatively significant loci in the NTM GWAS (A) Manhattan plot for the GWAS conducted on NTM-PD in Han Chinese population. The x axis represents chromosomal position, and the y axis represents the −log10 (p values) of genome-wide SNPs analyzed in the GWAS. The horizontal red line represents the genome-wide significance threshold of p = 5.0 × 10^−8. (B) Quantile-quantile (Q-Q) plot for genome-wide association analysis of NTM-PD in discovery stage. The horizontal axis shows −log10 transformed expected p values, while the vertical axis indicates −log10 transformed observed p values. The blue line represents the expected −log10(p values) under the null hypothesis. (C) Regional plot for the 16p21 susceptibility region. The most significant variant, represented in purple diamond, serves as a lead variant. −log[10]p values are shown for variants for the region 200 kb on either side of the lead variant. The color of each variant represents its pairwise linkage disequilibrium (LD) with the lead SNP, which is quantified by the r^2 values. These r^2 values were determined using the 1000 Genomes ASN data (November 2014). The recombination rate (second y axis) is plotted in light blue color. The genes within the relevant region are annotated and displayed as arrows. See also [108]Figure S1 and [109]Tables S1–S7. To further confirm the role of host genetic factors in different types of NTM infections, we divided the NTM-PD patients into three groups, namely, pulmonary MAC patients, pulmonary Mycobacterium kansasii (MAK) patients, and pulmonary MAB patients, and conducted a subtyping analysis. For 311 MAC cases and 794 healthy controls, the 16p21 locus retained genome-wide significance and the most significant variant was rs2520016 (p = 5.35 × 10^−10, [110]Table S2), followed by rs194800 (p = 1.96 × 10^−9, [111]Table S2). Both rs2520016 and rs194800 showed significant associations in MAB cases (p = 1.72 × 10^−4 and 5.29 × 10^−6, respectively; [112]Table S2), but not in MAK patients (p > 0.05, [113]Table S2), indicating that chromosome 16p21 was primarily a susceptibility locus for MAC disease. Furthermore, we investigated rs109592, rs11646605, and rs849177 identified in reported GWASs on NTM disease in our dataset.[114]^7^,[115]^8 We first analyzed the association of these SNPs in total NTM samples and then conducted separate analyses in the MAC, MAB, and MAK groups. The variants rs109592 and rs11646605 were significantly associated with susceptibility to NTM disease, MAC disease, and MAB disease in our samples, and rs849177 was not replicated in our samples ([116]Table S6). We explored the association of rs194800 and rs2520016 with susceptibility to TB based on our previous GWAS of TB in the Chinese Han population.[117]^11 These single-nucleotide polymorphisms (SNPs) exhibited marginal evidence of associations with TB (p < 0.10, [118]Table S7). Genetic variation is associated with gene expression and clinical characteristics The SNPs rs194800 and rs2520016 are located in the intergenic region between CHP2 and PRKCB ([119]Figure S2A). To investigate the potential biological significance of the two variants, we conducted an expression quantitative trait locus (eQTL) mapping analysis using peripheral blood mononuclear cells (PBMCs) obtained from NTM patients and healthy individuals. For NTM patients, carriers of the rs194800 C allele exhibited higher PRKCB expression than T-allele carriers, whereas no such association was observed in the normal subjects, suggesting that allele-specific expression of PRKCB might be induced by NTM infection ([120]Figure 2A). However, the rs194800 variant had no impact on CHP2 expression ([121]Figure 2B) and rs2520016 had no correlation with PRKCB or CHP2 expression ([122]Figures 2C and 2D). Additionally, we performed eQTL analysis using Open Targets Genetics,[123]^22 GTEx (Genotype-Tissue Expression, v.10),[124]^23 ImmuNexUT (Immune Cell Gene Expression Atlas from the University of Tokyo),[125]^24 and scQTLbase.[126]^25 Open Targets Genetics revealed that rs2520016 and rs194800 were consistently associated with the expression of the PRKCB gene across various biological contexts, including different states of monocytes and macrophages such as naive, lipopolysaccharides (LPS)_stimulated, and interferon gamma (IFNG) exposed ([127]Table S8). ImmuNexUT demonstrated significant eQTL associations between both variants and PRKCB in classical monocytes ([128]Table S9). Similarly, analysis from scQTLbase further supported these findings, showing significant eQTL associations between these SNPs and PRKCB expression in various immune cell types, including CD14^+ monocytes and conventional dendritic cells ([129]Table S9). Moreover, GTEx v.10 analysis suggested that these variants might also influence CHP2 expression in lung tissue ([130]Table S9). Collectively, these analyses underscore the substantial role of these variants in modulating PRKCB expression. Furthermore, we generated a promoter reporter construct containing a 1,000-bp fragment with the SNP rs194800 T or C allele ([131]Figure 2E) and performed a dual-luciferase assay. The C allele exhibited significantly higher promotion of luciferase expression than the T allele in HEK293T cells ([132]Figure 2F). To confirm whether the rs194800-containing region has the features of an enhancer, we have examined the H3K4me1, H3K27ac chromatin modifications, assay for targeting accessible-chromatin with high-throughout sequencing (ATAC-seq) signal, and DNase cluster distribution in the regions surrounding rs194800 with RegulomeDB and the Encyclopedia of DNA Elements (ENCODE, the ENCODE Project Consortium) datasets in University of California, Santa Cruz (UCSC) Genome Browser. The obvious epigenetic signals of H3K4me1 and H3K27ac near the rs194800 locus strongly supported the presence of enhancer activity in the rs194800-containing region ([133]Figure 2G). The high signals of ATAC-seq signal and DNase clusters near rs194800 reinforced the probability of the existence of enhancer activity ([134]Figure 2G). Overall, these findings suggested that the risk allele of rs194800 might promote allele-specific expression of PRKCB. Figure 2. [135]Figure 2 [136]Open in a new tab Association of rs194800 and rs2520016 with PRKCB and CHP2 mRNA expression in PBMCs Genomic DNA and mRNA were isolated from PBMCs from NTM-PD patients (n = 93) and healthy controls (n = 96). CHP2 and PRKCB mRNA expression was measured via quantitative reverse-transcription PCR (RT-PCR) and normalized to GAPDH. (A–D) The association of PRKCB and CHP2 mRNA expression with rs194800 genotype (A and B) and rs2520016 genotype (C and D). Each symbol represents an individual (mean ± SEM). (E) Schematic of the vector constructs. (F) Luciferase reporter plasmids were transfected into HEK293T cells, and relative luciferase activities were assayed by using Renilla luciferase activity for normalization (mean ± SEM). (G) Overview of the H3K27ac and H3K4me1chromatin modifications, ATAC, and DNase cluster distribution in the regions surrounding rs194800 supported from the UCSC Genome Browser. Rs194800 is indicated by the black vertical line. Data are representative of one experiment with at least three independent biological replicates in (A)–(D) and (F). Two-tailed unpaired Student’s t test (A–D, F) was used for statistical analysis. See also [137]Figure S2 and [138]Tables S8 and [139]S9. Next, we examined the association between the rs194800 polymorphism and various clinical indicators in NTM patients ([140]Figure S2B). The patients carrying the rs194800 C allele had lower plasma levels of soluble interleukin-2 receptor (sIL-2R), tumor necrosis factor alpha, and interleukin (IL)-1β than those carrying the T allele ([141]Figures S2C–S2E). The rs194800 C-allele carriers had a higher proportion of lung fields with lesions than those without the C allele ([142]Figure S2F). Meanwhile, the proportion of hemoptysis ([143]Figure S2G) and the proportion of symptom scores greater than 4 or less than 4 ([144]Figure S2H) showed no association with rs194800. Taken together, the rs194800 C allele was associated with the severity of NTM infection. Prkcb-deficient mice are more resistant to M. avium infection Because the 16p21 locus retained genome-wide significance only in MAC disease, we assessed the effect of PRKCB on the M. avium infection. Wild-type (WT) and Prkcb^−/− mice were infected with M. avium for 4 weeks. Subsequently, histopathological impairment and bacterial burden in the lung were examined. The Prkcb^−/− mice exhibited reduced infiltration of immune cells and fewer inflammatory lesions compared with WT mice ([145]Figures 3A and 3B). For colony-forming unit (CFU) assay, Prkcb^−/− mice displayed decreased bacterial burden in their lungs ([146]Figure 3C). Moreover, we detected PRKCB expression in PBMCs and found that PRKCB mRNA expression was significantly elevated in NTM-PD patients compared with that in healthy controls, especially in those with MAC infection ([147]Figure 3D). Furthermore, we analyzed PRKCB protein expression immunohistochemically in sections of lung tissue surgically excised from NTM-PD patients, with adjacent normal tissue serving as a control. PRKCB was expressed prominently in MAC and MAK patients, whereas it was nearly undetectable in MAB patients ([148]Figures 3E–3J). Strong PRKCB staining from a necrotizing granuloma of MAC patients was located at the periphery of granulomatous lesions but not in necrotic areas ([149]Figure 3K), suggesting that PRKCB involved in MAC disease by regulating cellular functions rather than by promoting tissue necrosis. In addition to its high expression in granuloma, PRKCB is also present in small amounts in the epithelium of healthy lung ([150]Figure 3K). Together, Prkcb exacerbates M. avium infection and plays a negative regulatory role in host anti-M. avium immunity. Figure 3. [151]Figure 3 [152]Open in a new tab Prkcb-deficient mice are more resistant to M. avium infection (A) Histopathology of lung sections from WT (n = 6) or Prkcb^−/− (n = 7) mice infected with M. avium by intranasal infection for 4 weeks. ^#1–3 indicate three representative lung sections for each group. The quantitation of inflammatory areas is shown in each section. (B) Quantitation of inflammatory areas in the lungs of mice infected with M. avium as in (A) (mean ± SEM). (C) Bacterial CFU in the lungs of the mice infected with M. avium as in (A) (mean ± SEM). (D) Real-time qPCR detection of PRKCB expression in PBMCs from controls (n = 96) or M. avium complex pulmonary disease (MAC-PD) patients (n = 48), M. abscessus pulmonary disease (MAB-PD) patients (n = 30), and M. kansasii pulmonary disease (MAK-PD) patients (n = 9) (mean ± SEM). (E–G) Immunohistochemical examination of PRKCB expression in granulomas from MAC patients (n = 2) (E), MAK patients (n = 2) (F), MAB patients (n = 2) (G), and adjacent normal tissue control (scale bars, 200 μm [top] and 20 μm [bottom]). One representative field for each group was showed, and positive cells are brown. (H–J) Quantification of PRKCB expression by immunostaining from the MAC group (H), MAK group (I), and MAB group (J) (mean ± SEM). 3 fields per group for quantitation of PRKCB-positive cells. (K) Representative PRKCB staining in a necrotizing granuloma of lung sections from MAC patients. Blue dashed lines delineated the granuloma. Yellow dashed lines delineated area surrounding necrotic zone within the granuloma. The right panels showed PRKCB staining in the granuloma. The middle and bottom left panels showed PRKCB staining in the normal lung. The red arrows indicated positive cells. (scale bars, 2000 μm [left top], 200 μm [left middle], 50 μm [left bottom], 200 μm [right top], 100 μm [right middle], and 50 μm [right bottom]). Images shown are representative of those observed in 2 MAC patients. Data are representative of one experiment with at least three independent biological replicates in (C) and (D). Two-tailed unpaired Student’s t test (B, D, H–J) was used for statistical analysis. Two-sided Mann-Whitney U test (C) was used for statistical analysis. PRKCB promotes intracellular survival of M. avium through inhibiting phagosomal acidification in macrophages To further explore the function of PRKCB during M. avium infection, we conducted transcriptomic analysis on the lung tissue of WT and Prkcb^−/− mice infected with M. avium for 4 weeks by using RNA sequencing (RNA-seq). A number of significantly differentially expressed genes (DEGs) were identified ([153]Figures S3A and S3B), and pathway enrichment analyses on these DEGs revealed that the innate immune system was the most significant pathway ([154]Figure S3C), suggesting that PRKCB might play a crucial role during M. avium infection through regulating innate immunity. Macrophages are the primary host innate immune cells, and their effectiveness in containing or eliminating intracellular non-tuberculous mycobacteria is essential for host anti-NTM immunity.[155]^26 Therefore, we analyzed the effect of PRKCB on the clearance of non-tuberculous mycobacteria through macrophages. Primary murine peritoneal macrophages from WT and Prkcb^−/− mice were infected with M. avium, MAB, and MAK for 3 h and 24 h and then subjected to a CFU assay. The intracellular NTM loads were similar at 3 h post-infection, indicating that PRKCB had no effects on the phagocytosis of non-tuberculous mycobacteria by macrophages. However, the intracellular amounts of M. avium and MAK were dramatically decreased at 24 h post-infection in Prkcb^−/− macrophages ([156]Figures 4A–4F), suggesting PRKCB as a host factor that aided the intracellular survival of NTM. It has been reported that phagosomal acidification is essential for the clearance of non-tuberculous mycobacteria.[157]^27^,[158]^28 We thus next examined whether PRKCB promoted intracellular survival of NTM by inhibiting phagosomal acidification. For this, the macrophages from WT and Prkcb^−/− mice were pretreated with Bafilomycin A1 (BafA1), an inhibitor of vacuolar ATPase (V-ATPase), and the intracellular survival of M. avium was examined. The inhibitors rescued the promoting effect of PRKCB on the intracellular survival of M. avium ([159]Figures 4G and 4H), suggesting that PRKCB promoted intracellular survival of M. avium through inhibiting V-ATPase-mediated phagosomal acidification. By LysoTracker, an acidotrophic fluorescent dye accumulated in acidic organelles, phagosomal acidification was assessed in primary murine peritoneal macrophages from WT and Prkcb^−/− mice infected with M. avium for 6 h. The results showed that Prkcb deficiency promoted phagosomal acidification ([160]Figures 4I and 4J). We treated macrophages with BafA1 and examined phagosomal acidification. BafA1 eliminated the difference in phagosomal acidification between WT and Prkcb^−/− mice ([161]Figures 4I and 4J), suggesting that PRKCB suppresses phagosomal acidification via V-ATPase. In contrast to the results of M. avium infection, PRKCB promoted phagosomal acidification to inhibit intracellular survival of M. tuberculosis through V-ATPase ([162]Figures S4A–S4D), suggesting that there was no shared pathogenesis mechanism mediated by PRKCB between NTM-PD and TB. Figure 4. [163]Figure 4 [164]Open in a new tab PRKCB promotes intracellular survival of NTM through inhibiting phagosomal acidification in macrophages (A–F) Intracellular CFU and bacterial survival in WT or Prkcb^−/− mice peritoneal macrophages infected with M. avium (A and B), M. kansasii (C and D), and M. abscessus (E and F) for the indicated times (MOI = 5) (mean ± SEM). (G and H) Intracellular CFU and bacterial survival in WT or Prkcb^−/− mice peritoneal macrophages treated with BafA1 (100 nM, 2 h) and infected with M. avium (MOI = 5) (mean ± SEM). (I) LysoTracker staining of macrophages from WT and Prkcb^−/− mice treated with BafA1 (100 nM, 2 h) and infected with M. avium for the 6 h (MOI = 5) (scale bar, 5 μm). M. avium were labeled with fluorescein isothiocyanate (FITC), and DAPI staining was used to identify nuclei. (J) Quantitative analysis of fluorescence by ImageJ as in (I) counting at least 200 random cells from each sample (mean ± SEM). Data are representative of one experiment with at least three independent biological replicates in (A)–(J). Two-tailed unpaired Student’s t test (A–H, J) was used for statistical analysis. See also [165]Figures S3 and [166]S4. PRKCB interacts with VPS16 and ATP6V1G To uncover the key effectors of PRKCB inhibition on phagosomal acidification, we performed a label-free proteomic analysis. The differential peptides were primarily enriched in vesicle fusion, protein localization to vacuole or lysosome, and lysosomal or vacuolar transport signaling ([167]Figure 5A). We examined the association of PRKCB with various components of these pathways by co-immunoprecipitation and identified the interaction of PRKCB with vacuolar protein sorting-associated protein 16 homolog (VPS16) ([168]Figure 5B). We further confirmed the interaction by overexpressing PRKCB and VPS16 ([169]Figure 5C). Colocalization of PRKCB with VPS16 was found by immunofluorescence ([170]Figure 5D). VPS16, a common subunit of the class C core endosomal vacuole tethering (CORVET) and homotypic fusion and vacuole protein sorting (HOPS) complex, is reported to function as a tether protein involved in phagosome-lysosome fusion.[171]^29 To examine the association of PRKCB and V-ATPase machinery, the interaction between PRKCB and various subunits of V-ATPase complex was analyzed using immunoprecipitation, and the results revealed that PRKCB interacted with the G subunit ([172]Figures 5E and 5F). The colocalization of PRKCB with ATP6V1G was also identified ([173]Figure 5G). The interaction of PRKCB with VPS16 and ATP6V1G implied that the V-ATPase and class C VPS complexes might be closely associated during phagosome maturation and endosome-lysosome fusion. In order to examine this hypothesis, we detected the interaction of VPS16 with ATP6V1G and found that VPS16 interacted with ATP6V1G ([174]Figures 5H and 5I). The colocalization of VPS16 with ATP6V1G was also observed ([175]Figure 5J), suggesting that VPS16 and ATP6V1G might mediate the interaction between the class C VPS complex and V-ATPase machinery. Figure 5. [176]Figure 5 [177]Open in a new tab PRKCB interacts with VPS16 and ATP6V1G (A) Schematic diagram (upper) showing the proteomic analysis of overexpression of PRKCB or vector in HeLa cells. The top enriched signaling pathways are shown (bottom). (B) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmids encoding FLAG-PRKCB and detected with the indicated antibody. (C) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmid encoding FLAG-PRKCB or hemagglutinin (HA)-VPS16. (D) Immunofluorescence of HeLa cells transfected with plasmids encoding FLAG-PRKCB or HA-VPS16 (scale bar, 5 μm). DAPI staining was used to identify nuclei. (E) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmids encoding FLAG-PRKCB or HA-tagged subunits of V-ATPase. (F) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmids encoding FLAG-PRKCB or HA-V1G. (G) Immunofluorescence of HeLa cells transfected with plasmids encoding FLAG-PRKCB or HA- V1G (scale bar, 10 μm). DAPI staining was used to identify nuclei. (H) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmids encoding HA-VPS16 and detected with the indicated antibody. (I) Immunoprecipitation and immunoblot of lysates of HeLa cells transfected with plasmids encoding FLAG-VPS16 or HA-V1G. (J) Immunofluorescence of HeLa cells transfected with plasmids encoding FLAG-VPS16 or HA-V1G (scale bar, 5 μm). DAPI staining was used to identify nuclei. PRKCB blocks the interaction between Vps-C and V-ATPase complexes Because PRKCB interacted with VPS16 and ATP6V1G, and VPS16 was recruited to ATP6V1G, we asked whether PRKCB binding to VPS16 and ATP6V1G affected the association of VPS16 and ATP6V1G during M. avium infection. To address this question, we examined endogenous interaction using VPS16 antibodies as the bait and detected endogenous ATP6V1G in WT and Prkcb^−/− macrophages infected with M. avium. Endogenous interactions of VPS16 and ATP6V1G could be detected in both the uninfected and infected WT macrophages ([178]Figure 6A), suggesting that interaction between ATP6V1G and the class C VPS regulated endosomal-lysosomal fusion in resting cells and phagosome-lysosome fusion in infected cells. Compared to WT macrophages, the interaction of VPS16 and ATP6V1G was significantly upregulated in Prkcb^−/− macrophages ([179]Figure 6A), suggesting that PRKCB inhibited the interaction of the class C VPS complex and V-ATPase complex. Disruption of the interaction between VPS16 and ATP6V1G by PRKCB could be observed using ATP6V1G antibody as the bait and detected VPS16 ([180]Figure 6B). Next, we detected the colocalization of VPS16, ATPV1G, and M. avium within WT and Prkcb^−/−macrophages. The results showed that VPS16 and ATP6V1G colocalized to the mycobacterial phagosome within macrophages and the colocalization enhanced in Prkcb^−/− macrophages compared to WT macrophages ([181]Figures 6C and 6D), suggesting that PRKCB blocked the fusion of lysosomes and bacilli-containing phagosomes. Furthermore, blocking of Vps16 with small interfering RNA (siRNA) attenuated Prkcb deficiency-mediated enhancement of M. avium-induced phagosomal acidification ([182]Figures 6E–6G), suggesting that PRKCB inhibited phagosomal acidification through VPS16. Knockdown of Vps16 in WT and Prkcb^−/− macrophages diminished the PRKCB-induced increase in the intracellular survival of M. avium in macrophages ([183]Figures 6H and 6I). It has been reported that M. avium could evade macrophages’ killing through secreting effector molecules into the cytoplasm to interfere with phagosomes maturation.[184]^30^,[185]^31 To determine whether M. avium proteins interacted with PRKCB or VPS16 to involve in the fusion of phagosomes and lysosomes, we isolated the M. avium phagosomes and performed a mass spectrometric analysis to find M. avium proteins bound to phagosome according to the methods of Danelishvili and Bermudez ([186]Figures S5A and S5B).[187]^28 Thirteen M. avium proteins were identified in our samples including M. avium proteins of alpha (MAV_1525) and beta (MAV_1527) subunits of ATP synthase reported in the Danelishvili and Bermudez study. We examined the interaction of PRKCB and the two reported M. avium proteins MAV_1525 and MAV_1527 using immunoprecipitation techniques. Additionally, we investigated the interaction of VPS16 and these M. avium proteins. Furthermore, we assessed the interaction of PRKCB or VPS16 and the two other M. avium proteins MAV_4671 and MAV_4996, which have been reported to interact with host proteins.[188]^27^,[189]^28 The results indicated that three out of the four M. avium proteins, namely MAV_1525, MAV_1527, and MAV_4996, were found to interact with VPS16. However, none of the four M. avium proteins showed any interaction with PRKCB ([190]Figures S5C and S5D), suggesting that the VPS16-mediated phagosomal fusion pathway was crucial for the host to eliminate M. avium. The pathway was utilized not only by host molecules like PRKCB but also by M. avium proteins. Together, these results demonstrate that PRKCB inhibits phagosomal acidification through blocking the class C VPS- and V-ATPase complexes-mediated phagosome-lysosome fusion during M. avium infection. Figure 6. [191]Figure 6 [192]Open in a new tab PRKCB blocks the interaction between Vps-C and V-ATPase complexes during infection (A and B) Endogenous interactions of VPS16 with ATP6V1G in WT or Prkcb^−/− mice peritoneal macrophages infected with M. avium for the indicated time and immunoprecipitated with VPS16 antibody (A) or V1G antibody (B). Rabbit immunoglobulin G (IgG) was used as negative control. (C) Localization of M. avium, VPS16, and V1G was detected in WT or Prkcb^−/− mice peritoneal macrophages infected with M. avium for 6 h with immunofluorescence staining (scale bar, 5 μm). DAPI staining was used to identify nuclei. (D) Quantitative analysis of puncta area by ImageJ as in (C) counting at least 200 random cells from each sample (mean ± SEM). (E) The efficiency of siRNA knockdown of vps16. (F) LysoTracker staining of macrophages from WT and Prkcb^−/− mice transfected with control siRNA or siRNA targeting mouse vps16 for 48 h and infected with M. avium for 6 h (MOI = 5) (scale bar, 5 μm). M. avium were labeled with FITC, and DAPI staining was used to identify nuclei. (G) Quantitative analysis of fluorescence by ImageJ as in (F) counting at least 200 random cells from each sample (mean ± SEM). (H and I) Intracellular CFU and bacterial survival in vps16 knockdown primary peritoneal macrophages from WT and Prkcb^−/− mice and infected with M. avium for the indicated times (MOI = 5) (mean ± SEM). Data are representative of one experiment with at least three independent biological replicates in (C)–(I). Two-tailed unpaired Student’s t test (D, E, G, H, and I) was used for statistical analysis. See also [193]Figure S5. PRKCB inhibitor has therapeutic potential against M. avium infection PRKCB inhibitors, particularly ruboxistaurin (LY-333531, RBX), have been the subject of extensive research.[194]^32^,[195]^33^,[196]^34 The reported randomized and multicenter clinical trials have demonstrated that ruboxistaurin can reduce the development and/or progression of diabetic microvascular complications, with phase 3 trials having been completed. Recent study has also indicated that ruboxistaurin is a potential therapeutic option for COVID-19.[197]^35 To explore the therapeutic effects of PRKCB inhibitors on NTM disease, peritoneal macrophages were pretreated with one of two effective PRKCB inhibitors, enzastaurin (ENZ) and ruboxistaurin, followed by infection with M. avium. Both inhibitors were capable of inhibiting the intracellular survival of pathogens in macrophages, with ruboxistaurin exhibiting a superior inhibitory effect ([198]Figures 7A and 7B). Ruboxistaurin exerted its effect by targeting PRKCB in vitro ([199]Figures 7C and 7D). In vivo, mice were intranasally infected with M. avium for 2 weeks and subsequently administered an intragastric solution of ruboxistaurin at a dosage of 1 mg/kg for an additional 4 weeks ([200]Figure 7E). In WT mice, treatment with the inhibitor significantly alleviated pathological damage in the lung tissue ([201]Figures 7F and 7H) and reduced bacterial burden ([202]Figure 7I) compared with the findings in the untreated group, suggesting that PRKCB inhibitor may hold therapeutic potential against M. avium infection. The difference in pathological damage ([203]Figures 7G and 7H) and bacterial burden ([204]Figure 7I) was eliminated in Prkcb^−/− mice treated with ruboxistaurin, suggesting that ruboxistaurin played a role through targeting PRKCB. Figure 7. [205]Figure 7 [206]Open in a new tab PRKCB inhibitor ameliorates M. avium infection (A and B) Intracellular CFU in mice peritoneal macrophages treated with enzastaurin (10 μM, 24 h) or ruboxistaurin (10 μM, 24 h) and infected with M. avium for 24 h (MOI = 5) (mean ± SEM). (C and D) Intracellular CFU and bacterial survival in WT and Prkcb^−/− mice peritoneal macrophages treated with ruboxistaurin (10 μM, 24 h) and infected with M. avium for the indicated times (MOI = 5) (mean ± SEM). (E) Schematic diagram of the procedure to evaluate the effect of PRKCB inhibitor on WT mice (n = 7) and Prkcb^−/− mice (n = 6) intranasally infected with M. avium for 2 weeks and intragastric administrated with ruboxistaurin (1 mg/mL) or vehicle solution for another 4 weeks. (F and G) H&E staining of lung sections of WT mice (F) and Prkcb^−/− mice (G) infected with M. avium as in (E). ^#1–3 indicate three representative lung sections for each group. The quantitation of inflammatory areas is shown in each section. (H) Quantitation of inflammatory areas in the lungs of mice infected with M. avium as in (E) (mean ± SEM). (I) Bacterial CFU in lungs of WT mice and Prkcb^−/− mice as in (E). RBX, ruboxistaurin; ENZ, enzastaurin. Data are representative of one experiment with at least three independent biological replicates in (A)–(D), (H), and (I). Two-tailed unpaired Student’s t test (A–D, H) was used for statistical analysis. Two-sided Mann-Whitney U test (I) was used for statistical analysis. Discussion In this two-stage GWAS of NTM-PD, we identified that the locus rs194800 on chromosome 16p21 was significantly associated with NTM-PD risk, especially for MAC disease. We further demonstrated that the rs194800 C allele was associated with PRKCB expression and the severity of NTM infection. Follow-up functional studies showed that PRKCB exacerbated M. avium infection in mice and promoted the intracellular survival of M. avium in macrophages through inhibiting the fusion of lysosomes and phagosomes. Finally, PRKCB inhibitors made mice more resistant to M. avium infection. This GWAS of NTM-PD attempted to identify the causal gene and mechanism behind the GWAS-related findings. These findings suggest that PRKCB is a strong causal gene involved in the pathogenesis of MAC disease and that treatment with a PRKCB inhibitor may serve as a therapeutic strategy against MAC disease. Non-tuberculous mycobacteria are ubiquitous in the environment. However, despite widespread exposure to them, only a few people develop NTM disease. Candidate gene studies have identified NRAMP1, CFTR, IL-10, and IL-28B as susceptibility genes for NTM-PD.[207]^36^,[208]^37^,[209]^38^,[210]^39 To date, only two GWASs of NTM-PD have been performed. In 2021, Ho Namkoong and colleagues conducted a GWAS in a Japanese population with pulmonary MAC infection and identified chromosome 16p21, specifically rs109592, as having the strongest association with MAC disease susceptibility. They further validated rs109592 in Korean and European populations. Interestingly, in our work, we also found the same susceptibility region on chromosome 16p21 in the Chinese population. Of note, in our study, the strongest association with NTM-PD was rs194800 but not rs109592. We also analyzed rs109592 in our samples and found that it had a significant association with susceptibility to NTM-PD in the Chinese population. Linkage disequilibrium analysis revealed that rs109592 and rs194800 were located within the same linkage equilibrium region (r^2 > 0.80 based on the 1000G European [EUR] and East Asian [ASN] data). Notably, two GWASs conducted in different populations determined that chromosome 16p21 exhibited the strongest association with susceptibility to MAC disease, suggesting that this region may contain causative variants and genes responsible for driving susceptibility to MAC disease. However, further investigation is warranted, including larger sample sizes and international collaborative GWAS across diverse populations. Because clinical manifestations of NTM-PD are similar to TB, we investigated the SNPs responsible for NTM-PD susceptibility in TB patients, and the role of PRKCB in Mtb infection. None of the SNPs showed an association with TB, and PRKCB exerted opposing regulatory effects on the intracellular survival of NTM and Mtb, suggesting that the chromosome 16p21 region might be specific to NTM-PD susceptibility and there is no genetic sharing mechanism or shared pathogenesis mediated by PRKCB between NTM-PD and TB. Within the chromosome 16p21 region, CHP2 and PRKCB are the two closest neighboring genes. In Ho Namkoong’s study, it was emphasized that CHP2 was a strong candidate gene involved in the pathogenesis of pulmonary MAC disease, while PRKCB was considered a potential gene alongside CHP2 based on multi-tissue eQTL analysis. In our study, rs194800 exhibited eQTL effects on PRKCB. Analysis of enhancer activity indicated that the region containing the variant rs194800 had allele-specific enhancer activity, and the risk allele C of rs194800 might correspond to higher enhancer activity as compared to the non-risk allele. However, whether rs194800 is a functional SNP warrants further investigation. Notably, PRKCB has also been identified as a genetic susceptibility locus for primary biliary cholangitis and systemic lupus erythematosus.[211]^40^,[212]^41 Taking together, rs194800 is a probable causative variant for NTM-PD and PRKCB may also be a candidate effector gene contributing to such diseases. PRKCB is a member of the protein kinase C (PRKC) family and functions as a serine/threonine-protein kinase.[213]^42 Increasing evidence suggests that PRKCB regulates B cell polarity and activation and autophagy levels[214]^43^,[215]^44^,[216]^45 and is involved in the pathogenesis of several diseases.[217]^46^,[218]^47^,[219]^48 However, the function of PRKCB in NTM-PD remains unknown. Our study demonstrates that PRKCB involves in the pathogenesis of NTM-PD. PRKCB weakened the host’s ability of clearance of M. avium. The persistent M. avium might induce excessive or prolonged inflammatory responses to cause pathological damage to the lungs. Although it has been long observed that inhibition of phagosome-lysosome fusion and alteration of phagosomal pH may contribute to the intracellular survival of M. avium,[220]^49 the underlying mechanism remains poorly understood. V-ATPase is recruited to the phagosomal membrane during phagosome maturation for luminal acidification and the class C VPS complex, as the membrane-tethering factor is involved in this process.[221]^50^,[222]^51 Our studies demonstrated that VPS16 and subunit G of V-ATPase were key players to mediate phagosome-lysosome fusion during M. avium infection. Vesicle fusion in the endocytic pathway depended on two membrane-tethering protein complexes, HOPS and CORVET. The subunits of VPS11, VPS16, VPS18, and VPS33 associate to form the stable VPS-C core complex for HOPS and CORVET.[223]^52^,[224]^53 VPS16 is required for the fusion of intracellular compartments with lysosomes.[225]^54^,[226]^55^,[227]^56 Our research revealed that PRKCB interacted with VPS16 and V1G, suggesting that this interaction may potentially lead to the specific localization of PRKCB at the phagosome-lysosome interface. This specific localization hindered the fusion of V-ATPase with the phagosome, resulting in inhibition of mycobacterial phagosome acidification. Our findings show a mechanism for explaining how M. avium prevents lysosomal fusion with mycobacterial phagosome in macrophages, enabling its survival within phagosomes. However, whether PRKCB depends on its kinase activity to block the interaction between VPS16 and ATP6V1G needs to be explored further. The limited efficacy of current treatments for NTM disease poses a significant challenge for disease management. One of the main reasons for the inadequate success of NTM disease treatment is the resistance of NTM pathogens to multiple anti-mycobacterial drugs.[228]^57 There is thus an urgent need for novel therapeutic strategies that target the host rather than the NTM pathogens themselves.[229]^26 Ruboxistaurin, a highly specific inhibitor of PRKCB,[230]^58 has been extensively studied in phase 3 trials for diabetic retinopathy and has shown good tolerance in patients.[231]^59 Our study found that ruboxistaurin could inhibit the intracellular survival of non-tuberculous mycobacteria in macrophages and reduced the bacterial burden and pathological impairments in the lung tissue of mice infected with M. avium through specifically targeting PRKCB. These findings suggest that targeting PRKCB could be a promising host-directed therapy against MAC disease. Importantly, ruboxistaurin has been proven to be safe, making it a promising therapeutic strategy for MAC disease. Together, these findings not only support the significance of PRKCB in M. avium infection but also reveal a potential therapeutic approach for managing MAC disease. Identifying the causal genes and mechanisms behind GWAS hits poses various challenges. Building on our GWAS, we performed functional studies to identify rs194800 as a probable causative variant and PRKCB as a candidate effector gene involved in the pathogenesis of MAC disease. These findings enhance our understanding of the genetic basis of MAC disease and contribute to our knowledge of its pathogenesis. Moreover, targeting PRKCB may hold promise as a therapeutic approach for MAC disease. Limitations of the study The present study suggests that the rs194800-containing region has the characteristics of an enhancer through bioinformatics analysis. Additional functional experiments, such as constructing rs194800 knockin cells or mice, could provide further evidence to confirm that rs194800 is a functional SNP. In addition, the mechanism by which PRKCB binding to VPS16 and ATP6V1G affects phagosome-lysosome fusion needs further exploration. The role of PRKCB kinase activity in this process is necessary to evaluate. Further experiments with PRKCB kinase activity mutants, combined with analyses such as phosphoproteome analysis of PRKCB, could provide additional mechanistic insights. Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Wei Sha (Shfksw@tongji.edu.cn). Materials availability Plasmids generated in this study are available from lead contact upon request. Data and code availability * • RNA-seq data have been deposited at BioSample database, and GWAS summary statistics data have been deposited at [232]bio-x.com (gwas.bio-x.cn) and [233]figshare.com and are publicly available as of the date of publication. Accession numbers are listed in the [234]key resources table. * • This paper does not report original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [235]lead contact upon request. Acknowledgments