Abstract
Measles virus (MeV) binds, infects, and kills CD150+ memory T cells, leading to immune amnesia. Whether MeV targets innate, memory-like T cells is unknown. We demonstrate that human peripheral blood and hepatic mucosa-associated invariant T (MAIT) cells and invariant natural killer T cells express surprisingly high levels of CD150, more than other lymphocyte subsets. Furthermore, exposing MAIT cells to MeV results in their efficient infection and rapid apoptosis. This constitutes the first report of direct MAIT cell infection by a viral pathogen. Given MAIT cells’ antimicrobial properties, their elimination by MeV may contribute to measles-induced immunosuppression and heightened vulnerability to unrelated infections.
Keywords: measles, MAIT cells, iNKT cells, immune amnesia, immunosuppression, secondary infections, CD150, cell death, apoptosis, vaccination
Measles is a contagious and potentially deadly, but vaccine-preventable, disease. Measles outbreaks have been alarmingly on the rise. Many deaths from measles are due to secondary infections with unrelated pathogens, a reflection of measles virus (MeV)-induced immunosuppression [1].
MeV employs CD46, nectin 4, and CD150 to enter host cells. CD46 is ubiquitously expressed but not efficiently utilized by wild-type (WT) MeV isolates, and nectin 4 is the epithelial receptor for MeV. Vaccine, laboratory-adapted, and WT strains employ CD150 to infect permissive immune cells, and measles lymphopenia and antigen-presenting cell dysfunctions are linked to CD150 expression. Importantly, by infecting and destroying preexisting CD150+ memory cells, MeV hinders the body’s recollection of past encounters with pathogens, a phenomenon called immune amnesia [2].
Whether MeV targets innate, memory-like T cells, including mucosa-associated invariant T (MAIT) and invariant natural killer T (iNKT) cells, is unknown. As “emergency responders” to infection, MAIT and iNKT cells recognize vitamin B metabolites and glycolipid antigens of microbial origin, respectively [3]. They also respond to inflammatory cues and cytokines such as interleukin-12 (IL-12) and IL-18. Of note, these invariant T cells express surface molecules routinely used as phenotypic markers of conventional T cells (eg, CD3), NK cells (eg, CD56 and CD161), and memory T (TM) cells (eg, CD45RO) [4]. Therefore, they may have been overlooked in previous studies on immune amnesia.
Here, we demonstrate that CD150 is most abundantly expressed by invariant T cells, at levels that surprisingly exceed those of TM cells. Furthermore, MeV efficiently infects MAIT cells and quickly programs them for apoptotic death. These findings may partially explain impaired immunity to unrelated pathogens during and after measles.
METHODS
Ethics Statement
Peripheral blood (PB) was drawn from healthy volunteers, 11 men and 8 women (Supplementary Table 1). Tumor-free hepatic tissue samples were harvested from patients undergoing surgical operations listed in Supplementary Table 2. Our protocols were approved by the Western University Research Ethics Board for Health Sciences Research Involving Human Subjects.
Transcriptomic Analysis of Peripheral Blood Mononuclear Cells
The pbmc_10k_v3 dataset, consisting of single-cell RNA sequencing (scRNA-Seq) data from 11 769 peripheral blood mononuclear cells (PBMCs), was created by 10x Genomics (https://support.10xgenomics.com/single-cell-gene-expression/datasets/3.0.0/pbmc_10k_v3). Preprocessing and analysis were performed using Seurat v3.1.1. Features were retained if detected in ≥5 cells. Cells were retained if they contained ≥500 features, between 2000–20 000 unique molecular identifiers, and <20% mitochondrial transcript reads. Scrublet was used to identify and remove cells with doublet scores >0.1. After log-normalizing the count data and identifying the 3000 most variable genes, expression values were scaled, centered, and used for principal component analysis. The top 30 components were used for cluster detection with the spatial linear model algorithm of Seurat v3.1.1 (resolution = 0.4). The resulting clusters were assigned cell type identities based on known marker genes (Supplementary Figure 1).
PBMC and Hepatic Mononuclear Cell Isolation
PBMCs were isolated using SepMate-50 tubes (STEMCELL Technologies) and density gradient centrifugation at 1200 × g. To obtain nonparenchymal hepatic mononuclear cells (HMNCs), liver samples were homogenized and parenchymal cells were removed by centrifugation at 700 × g in 33.75% Percoll PLUS. Exposure to ammonium-chloride-potassium lysis buffer eliminated erythrocytes, and the remaining HMNCs were washed in phosphate-buffered saline (PBS) containing 2% fetal bovine serum (FBS).
Cytokine Stimulation
PBMCs were suspended in RPMI supplemented with 10% FBS, 2 mM GlutaMAX-I, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate, and 10 mM HEPES. Cells were seeded at a density of 5×105 cells/well of a U-bottom microplate and cultured for 24 hours at 37°C with or without 5 ng/mL each of recombinant human IL-12 (rhIL-12) p70 (PeproTech) and rhIL-18 (MBL International).
MeV Propagation and Infection
WT MeV strains Ichinose-B 323 and Khartoum Sudan were engineered to express enhanced green fluorescent protein (GFP). They are referred to as IC323-eGFP and KS-eGFP, respectively. MeVs were propagated in Vero cells expressing human CD150. PBMCs were seeded at 1×105 cells/well of a microplate containing DMEM with 5% FBS, to which MeVs were added at a multiplicity of infection (MOI) of 1. Cultures were incubated at 37°C for 1, 4, 8, or 24 hours. In several experiments, PBMCs were stimulated with rhIL-12 and rhIL-18 before they were exposed to IC323-eGFP.
Tetramers and Antibodies
Phycoerythrin (PE)-conjugated 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU)-loaded MHC-related protein 1 (MR1) tetramers and allophycocyanin (APC)-conjugated PBS-57-loaded CD1d tetramers were provided by the National Institutes of Health Tetramer Facility. 6-formylpterin-loaded MR1 tetramers and unloaded CD1d tetramers served as staining controls.
Fluorochrome-labeled monoclonal antibodies (mAbs) to CD3ε (clone UCHT1), CD4 (RPA-T4), CD19 (SJ25C1), CD38 (HIT2), CD45RO (UCHL1), CD46 (8E2), CD69 (FN50), CD150/SLAM [A12 (7D4)], CD197/CCR7 (3D12), and γδ TCR (B1.1), a mouse IgG1κ isotype control (P3.6.2.8.1), and an Annexin V Apoptosis Detection Kit APC were purchased from Thermo Fisher.
Cytofluorimetric Analyses
PBMCs and HMNCs were stained for 30 minutes at room temperature with tetramer/mAb cocktails prepared in PBS containing 2% FBS, washed, and interrogated using a FACSCanto II cytometer.
MAIT cells were defined as CD3+MR1 tetramer+ cells, iNKT cells as CD3+CD1d tetramer+ cells, γδ T cells as CD3+γδTCR+ cells, central memory T (TCM) cells as CD3+MR1 tetramer−CD1d tetramer−γδTCR−CD45RO+CCR7+ cells, effector memory T (TEM) cells as CD3+MR1 tetramer−CD1d tetramer−γδ TCR−CD45RO+CCR7− cells, and naive T (TN) cells as CD3+MR1 tetramer−CD1d tetramer−γδTCR−CD45RO−CCR7+ cells. Gates were drawn based on uninfected, unstained, or isotype control-stained conditions as appropriate. Bulk conventional CD4+ T cells and B cells were defined as CD3+CD4+tetramer− and CD3−CD19+ cells, respectively.
To detect MAIT cells in early and late stages of death, PBMCs from MeV-infected and -uninfected cultures were harvested, washed, and stained at 4°C with Fixable Viability Dye eFluor 780. Cells were then washed and surface-stained with a PE-Cy7-conjugated anti-CD3 mAb and PE-labeled, 5-OP-RU-loaded MR1 tetramers. After 30 minutes, cells were washed and stained with Annexin V-APC for 15 minutes at room temperature.
Statistical Analyses
Statistical comparisons were performed using Student t tests, ANOVA, or Wilcoxon signed-rank tests. Kolmogorov-Smirnov tests were conducted to assess the normal distribution of indicated variables (Supplementary Table 3).
RESULTS
MAIT Cells Are the Foremost Expressors of CD150 Among PBMCs
To determine whether MAIT cells express SLAMF1, the gene encoding CD150 that mediates measles-associated immunosuppression, we analyzed a PBMC scRNA-Seq dataset from 10x Genomics.
Unsupervised clustering identified several PBMC subsets based on canonical marker genes (Supplementary Figure 1A). These included CD14+ monocytes, CD16+ monocytes, classical dendritic cells, plasmacytoid pre-DCs (pDCs), NK cells, B cells, and three T cell populations, which all expressed CD3D, CD3E, and TRAC and were further classified into conventional CD4+ T cells (CD4, MAL), conventional CD8+ T cells (CD8A, CD8B), and MAIT cells (NCR3, SLC4A10, RORC, DPP4, ZBTB16) [5] (Figure 1A and Supplementary Figure 1A and 1B). A minor platelet population was also detectable.
Figure 1.
MAIT cells express the highest levels of SLAMF1/CD150 among PBMC subsets. A, Publicly available scRNA-Seq data from 11 769 PBMCs were analyzed. The 9432 cells that passed quality control filters are visualized on a t-SNE projection (left panel) demonstrating clusters corresponding to CD14+ monocytes (n = 2992), CD16+ monocytes (n = 328), cDCs (n = 74), pDCs (n = 68), NK cells (n = 544), B cells (n = 1419), CD4+ T cells (n = 2643), CD8+ T cells (n = 720), MAIT cells (n = 592), and platelets (n = 52). Each dot represents a single cell. The expression of SLAMF1 across indicated clusters, quantified as normalized and scaled count data, is shown in a violin plot (right panel), in which dots represent single cells and widths denote cell densities. B and C, MAIT cells and other peripheral blood T cell subsets were examined for CD150 expression. MAIT and iNKT cells were identified by MR1 tetramer staining (PubMed IDs 24101382 and 24695216) and CD1d tetramer staining (PubMed IDs 10839805 and 10974039), respectively. Open and filled histograms correspond to the staining of PBMCs with anti-CD150 and isotype control, respectively, after gating on CD3+MR1 tetramer+ MAIT cells (B). The frequencies of CD150+ cells and the gMFI of CD150 staining (C) in indicated T cell subsets are summarized using Box-and-Whisker plots, with each symbol representing an individual donor. *, *** and **** denote differences with P < .05, P < .001, and P < .0001, respectively, using matched one-way ANOVA with Dunnett post-hoc analysis. D, PBMCs (n = 4) were left untreated or stimulated with rhIL-12 plus rhIL-18. The frequencies of MAIT cells expressing CD150, CD69, or CD46 were determined 24 hours later by flow cytometry. ** denotes a difference with P < .05 by paired Student t test. Abbreviations: 5-OP-RU, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil; APC, allophycocyanin; cDC, classic dendritic cell; FITC, fluorescein isothiocyanate; gMFI, geometric mean fluorescence intensity; iNKT, invariant natural killer T cell; MAIT, mucosa-associated invariant T cell; MR1, MHC-related protein 1; NK cell, natural killer cell; NS, not significant; PBMC, peripheral blood mononuclear cell; pDC, plasmacytoid pre-dendritic cell; PE, phycoerythrin; rhIL, recombinant human interleukin; scRNA-Seq, single-cell RNA sequencing; TCM, central memory T cell; TEM, effector memory T cell; TN, naive T cell; t-SNE, t-distributed stochastic neighbor embedding.
As expected, all PBMC subsets, but not the accompanying platelets, expressed CD46 (Supplementary Figure 2A and 2B). Strikingly, SLAMF1 expression was not only evident but most pronounced within the MAIT cluster (Figure 1A). Statistically, SLAMF1 was more highly expressed by MAIT cells than by any other cell types (P < 2.2e-16 by Wilcoxon signed-rank tests). Of note, iNKT cells did not form a distinct cluster due likely to their low frequency in human PB.
Cytofluorimetric staining of MAIT cells by 5-OP-RU-loaded MR1 tetramers confirmed their abundance in the circulation (Supplementary Figure 3A) and their surface expression of CD150 (Figure 1B and 1C). Both CD150+ cell frequencies and CD150 expression on a per-cell basis were significantly higher in PB MAIT cells than in bulk CD4+ T cells and B cells (Supplementary Figure 4A and 4B), γδ T, TN, TCM, and TEM cells (Figure 1C and Supplementary Figure 5). This was notable because TCM and TEM cells are considered the main T cell subsets targeted by MeV, resulting in immune amnesia [2, 6]. The magnitude of CD150 expression by MAIT cells could be matched only by that of iNKT cells (Figure 1C and Supplementary Figure 4A and 4B).
MAIT cells are enriched in the human liver [4] (Supplementary Figure 3B). We asked whether hepatic and PB MAIT cells follow the same CD150/CD46 expression pattern. We found all MAIT cells among HMNCs to copiously express CD150 (Supplementary Figure 6A, 6B and 6C). Furthermore, the expression pattern of CD150 on various hepatic T cell subsets was similar to that of PB T cells (Supplementary Figure 6B and 6C).
MAIT Cells Exposed to IL-12 and IL-18 Retain Their CD150/CD46 Expression
Viruses lack the vitamin B biosynthesis machinery, which supplies prototoypic MR1 ligands [3], and fail to stimulate MAIT cells in a TCR-dependent fashion. However, they can trigger MAIT cell activation through cytokines, primarily through IL-18 and IL-12 whose receptors MAIT cells express [7]. We asked whether exposing MAIT cells to rhIL-12 and rhIL-18 alters their expression levels of MeV receptors. While 24-hour stimulation with these cytokines upregulated CD69, an early activation marker, the frequencies of CD150+ and CD46+ MAIT cells remained stable (Figure 1D). This treatment also increased, rather than decreased, the geometric mean fluorescence intensity of staining for these receptors (Supplementary Figure 7). Therefore, MAIT cells should maintain their susceptibility to MeV in a cytokine milieu established through infection with MeV or other viruses, or during coinfections.
MeV Infects MAIT Cells
To explore whether MeV could indeed infect MAIT cells, we incubated PBMCs for 24 hours with IC323-eGFP or KS-eGFP. Exposure to MeV virions resulted in efficient MAIT cell infection (Figure 2A, Supplementary Figure 8, and Supplementary Figure 9A). In these cultures, eGFP+ (infected) MAIT cells were significantly more frequent than any other infected T cell subsets with the sole exception of iNKT cells (Figure 2B and Supplementary Figure 9B). Importantly, the percentage of infected cells was higher among MAIT cells than in TCM and TEM cells, thus closely mirroring the CD150 expression pattern of these populations (Figure 1C). We found MeV infection not to activate MAIT cells because infected cells did not upregulate CD69 or CD38 (Supplementary Figure 10).
Figure 2.
MeV efficiently infects and destroys MAIT cells. A and B, PBMC cultures were left untreated or inoculated with IC323-eGFP (PubMed ID: 18568079) at an MOI of 1. After 24 hours, the percentage of eGFP+ cells in indicated T cell subsets was determined by flow cytometry after setting the appropriate gates using uninfected PBMCs. A representative dot plot displays eGFP+ (grey) and eGFP− (black) cells after gating on CD3+MR1 tetramer+ MAIT cells (A). Summary data depicting the frequencies of infected cells across different T-cell subsets are shown (B). Each symbol represents an individual donor, and error bars represent SEM. **** denotes differences with P < .0001 using one-way ANOVA with Dunnett post-hoc analysis. C and D, PBMCs were left untreated or inoculated with IC323-eGFP. After 1, 4, 8, and 24 hours, cells were stained with anti-CD3, 5-OP-RU-loaded MR1 tetramers, Annexin V, and Fixable Viability Dye. Indicated populations were defined based on their staining with Annexin V and Viability Dye, or lack thereof, after gating on total, eGFP+ (infected) or eGFP− (uninfected) MAIT cells. Quadrant gates in representative contour plots were drawn after identifying MAIT cells among PBMCs not stained with Annexin V or Viability Dye (C). Changes in the frequencies of indicated populations over time are summarized (D). Error bars represent SEM (n = 3). * denotes statistically significant differences with P < .05 (or less) using matched two-way ANOVA with Dunnett post-hoc analysis. Abbreviations: 5-OP-RU, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil; APC, allophycocyanin; eGFP, enhanced green fluorescent protein; IC323, Ichinose-B 323; iNKT, invariant natural killer T cell; MAIT, mucosa-associated invariant T cell; MeV, measles virus; MOI, multiplicity of infection; MR1, MHC-related protein 1; PBMC, peripheral blood mononuclear cell; TCM, central memory T cell; TEM, effector memory T cell; TN, naive T cell.
MeV Infection Kills MAIT Cells
To ascertain whether MeV infection of MAIT cells triggers their destruction, we costained PBMCs recovered from uninfected and IC323-infected cultures with Annexin V and Fixable Viability Dye. After gating on CD3+MR1 tetramer+ events, Annexin V+Viability Dye− and Annexin V+Viability Dye+ cells were defined as early-stage apoptotic and late-stage apoptotic/necrotic MAIT cells, respectively. MAIT cells that did not stain positively with Annexin V and Fixable Viability Dye were considered viable. As early as 1 hour after MeV inoculation, the majority of eGFP+ (infected) MAIT cells were Annexin V+ (Figure 2C and 2D). Moreover, a substantial proportion of this population was in an early stage of the apoptotic process as judged by a lack of staining with Fixable Viability Dye (Figure 2C). In contrast, eGFP− (uninfected) MAIT cells harvested from infected cultures contained smaller Annexin V+Viability Dye− and Annexin V+Viability Dye+ fractions (Figure 2C and 2D). At 4-, 8-, and 24-hour timepoints post-MeV exposure, higher percentages of dying or dead cells were similarly detectable among infected MAIT cells (Figure 2D). MAIT cells from parallel cultures that had not received an MeV inoculum remained fairly viable over time (Figure 2D). Collectively, these results indicate that MeV infection quickly programs MAIT cells for apoptotic death.
DISCUSSION
To date, memory T/B cells have been the focus of investigations on immune amnesia [2, 6]. We now report that compared to TM cells, MAIT cells express higher levels of CD150 and are more permissive to MeV. Importantly, MeV infection of MAIT cells results in their quick demise. This work constitutes the first report of direct MAIT cell infection by a viral pathogen.
MAIT cells are abundant in the respiratory tract, the main point of entry for MeV and many other microbes. They also comprise a major T cell population in the liver [4], which receives gut microbes and their products through the portal vein. Importantly, measles-induced mortality due to immunosuppression is typically caused by secondary infections in respiratory and gastrointestinal tracts [8]. We propose that by depleting MAIT cells, MeV promotes unrelated infections, a hypothesis that warrants further investigation. Investigating invariant T cell homeostasis and recovery after measles will be an exciting line of inquiry. It will also be important to assess local MAIT cell responses to hepatotropic microbes in the face or in the wake of measles.
MAIT cells participate in antiviral defense through cytokine receptor signaling. Measles can change the cytokine landscape in the PB and likely elsewhere [9, 10]. Coinfections with MeV and other viruses also do occur [11]. In this study, exposure to IL-12 and IL-18, potent MAIT-activating cytokines often produced during viral infections [7, 12], did not reduce the expression of CD46/CD150 on MAIT cells (Figure 1D and Supplementary Figure 7), their permissiveness to MeV, or their propensity for MeV-induced apoptosis (Supplementary Figure 11).
Unlike classical HLA molecules, MR1 and CD1d are monomorphic. Therefore, MAIT and iNKT cell ligands work beyond the HLA restriction barrier and may potentially serve as therapeutics in genetically diverse populations. Synthetic vitamin B metabolites (eg, 5-OP-RU) and certain drugs or drug-like molecules, such as diclofenac metabolites, bind MR1 [13, 14]. A CD1d-restricted glycolipid agonist of iNKT cells called α-galactosylceramide has shown promise in preclinical models and clinical trials for viral diseases [15]. Such and similar compounds may be efficacious in targeting the remaining or returning MAIT cells to reverse MeV-induced immunosuppression.
Future studies should evaluate invariant T cell frequencies and functions before and after the experimental MeV infection of nonhuman primates and also in unvaccinated children before and after measles.
Supplementary Data
Supplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.
Notes
Acknowledgments. We thank Mr Gary Sisson for his assistance with MeV propagation; and Drs Anton Skaro, Ken Leslie, and Edward Davies for providing hepatic tissue samples.
Financial support. This work was supported by the Canadian Institutes of Health Research (grant number PJT-156295 to S. M. M. H.). P. T. R. is a recipient of an Alexander Graham Bell Canada Graduate Scholarship (doctoral) from Natural Sciences and Engineering Research Council of Canada.
Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
References
- 1. Griffin DE Measles virus-induced suppression of immune responses. Immunol Rev 2010; 236:176–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. de Vries RD, McQuaid S, van Amerongen G, et al. Measles immune suppression: lessons from the macaque model. PLoS Pathog 2012; 8:e1002885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Chandra S, Kronenberg M. Activation and function of iNKT and MAIT cells. Adv Immunol 2015; 127:145–201. [DOI] [PubMed] [Google Scholar]
- 4. Dusseaux M, Martin E, Serriari N, et al. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood 2011; 117:1250–9. [DOI] [PubMed] [Google Scholar]
- 5. Park D, Kim HG, Kim M, et al. Differences in the molecular signatures of mucosal-associated invariant T cells and conventional T cells. Sci Rep 2019; 9:7094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. de Swart RL, Ludlow M, de Witte L, et al. Predominant infection of CD150+ lymphocytes and dendritic cells during measles virus infection of macaques. PLoS Pathog 2007; 3:e178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Ussher JE, Bilton M, Attwod E, et al. CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner. Eur J Immunol 2014; 44:195–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Shanks GD, Hu Z, Waller M, et al. Measles epidemics of variable lethality in the early 20th century. Am J Epidemiol 2014; 179:413–22. [DOI] [PubMed] [Google Scholar]
- 9. Ten Oever J, Riza A, Sabou M, Cismaru C, Netea MG, Slavcovici A. Characterization of the acute inflammatory response in measles infection. J Pediatric Infect Dis Soc 2014; 3:197–200. [DOI] [PubMed] [Google Scholar]
- 10. Lin WW, Nelson AN, Ryon JJ, Moss WJ, Griffin DE. Plasma cytokines and chemokines in Zambian children with measles: innate responses and association with HIV-1 coinfection and in-hospital mortality. J Infect Dis 2017; 215:830–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hang LK, Do LP, Van TT, et al. Viral co-infections among children with confirmed measles at hospitals in Hanoi, Vietnam, 2014. Asian Pac J Trop Med 2017; 10:171–4. [DOI] [PubMed] [Google Scholar]
- 12. van Wilgenburg B, Scherwitzl I, Hutchinson EC, et al. ; STOP-HCV Consortium MAIT cells are activated during human viral infections. Nat Commun 2016; 7:11653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kjer-Nielsen L, Patel O, Corbett AJ, et al. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature 2012; 491:717–23. [DOI] [PubMed] [Google Scholar]
- 14. Keller AN, Eckle SB, Xu W, et al. Drugs and drug-like molecules can modulate the function of mucosal-associated invariant T cells. Nat Immunol 2017; 18:402–11. [DOI] [PubMed] [Google Scholar]
- 15. van den Heuvel MJ, Garg N, Van Kaer L, Haeryfar SM. NKT cell costimulation: experimental progress and therapeutic promise. Trends Mol Med 2011; 17:65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.