Abstract Fibrosis-associated fibroblasts have been identified across various fibrotic disorders, but not in the context of biomaterials, fibrotic encapsulation, and the foreign body response. In other fibrotic disorders, a fibroblast subpopulation defined by Thy-1 loss is strongly correlated with fibrosis yet we do not know what promotes Thy-1 loss. We have previously shown that Thy-1 is an integrin regulator enabling normal fibroblast mechanosensing, and here, leveraging nonfibrotic microporous annealed particle (MAP) hydrogels versus classical fibrotic bulk hydrogels, we demonstrate that Thy1^−/− mice mount a fibrotic response to MAP gels that includes inflammatory signaling. We found that a distinct and cryptic α–smooth muscle actin–positive Thy-1^− fibroblast population emerges in response to interleuklin-1β (IL-1β) and tumor necrosis factor–α (TNFα). Furthermore, IL-1β/TNFα-induced Thy-1^− fibroblasts consist of two distinct subpopulations that are strongly proinflammatory. These findings illustrate the emergence of a unique proinflammatory, profibrotic fibroblast subpopulation that is central to fibrotic encapsulation of biomaterials. __________________________________________________________________ A regenerative, nonfibrotic biomaterial helped identify inflammatory fibroblast subpopulations central to fibrotic encapsulation. INTRODUCTION Engineered materials have long been used as implants to serve as sensors, drug delivery devices, stents, tissue/joint replacement components, or surgical meshes ([38]1). Implant success can be hindered by the foreign body response, a chronic inflammatory response that leads to fibrotic encapsulation and inhibits implant function ([39]2–[40]5). This fibrotic response is a critical barrier to success for many implants that the biomaterials and regenerative medicine fields are focused on overcoming. Recently, a microporous annealed particle (MAP) hydrogel system was developed, which entails injectable microgel particles that are subsequently annealed together in situ, creating an interconnected porous structure. In various animal models, MAP gels have been shown to regulate innate and acquired immunity, evade fibrosis, and accelerate wound closure and regeneration ([41]6–[42]9). Given the reduced inflammation and fibrosis associated with MAP gel implantation, this biomaterial system has the ability to also serve as model system to study fundamental mechanisms of biomaterial-associated fibrosis. Fibrotic disorders (e.g., cardiovascular fibrosis, idiopathic pulmonary fibrosis, liver cirrhosis, and systemic sclerosis) have been associated with 45% of deaths in the developed world, indicating that the problem of fibrosis is a major health care burden ([43]10). Fibrosis is characterized by chronic inflammation followed by excessive myofibroblast accumulation, aberrant extracellular matrix (ECM) deposition, and aberrant mechanotransduction. The chronic inflammatory component of fibrosis has been well characterized ([44]10–[45]15), and even more so the myofibroblast and ECM components ([46]16–[47]18). However, there is a dearth of studies investigating how chronic inflammation leads to pathogenic myofibroblast and ECM accumulation in fibrosis. Bridging the gap between inflammation and dysfunctional mechanotransduction is a critical piece to better understand key mechanisms behind fibrosis. In addition, understanding how immune-stromal cross-talk can inspire or mitigate fibrosis could help inform better biomaterial design and identify previously unknown avenues for therapeutic approaches for fibrosis ([48]19). In human fibrotic disorders, Thy-1–negative fibroblasts emerge as a key player in progressive tissue remodeling and fibrosis. Thy-1 is a glycosylphosphatidylinositol-anchored membrane protein that is expressed on fibroblasts, neurons, and thymocytes, and we have previously established that it is essential to normal fibroblast mechanosensitivity. Thy-1 directly binds αvβ3 integrin in cis, regulates its tonic/baseline activity, and primes the integrin for efficient signal transduction. This critical regulatory mechanism for Thy-1 prevents aberrant fibroblast contractility, strain stiffening of surrounding matrix, and fibrosis ([49]16, [50]20). Thy-1–negative fibroblasts, even while on soft substrates, display elevated contractility and ECM deposition and are prone to myofibroblastic differentiation in fibrosis in the lung and heart ([51]Fig. 1A) ([52]20–[53]23). Fibrotic disorders and, in some cases, the very fibrotic lesions themselves are characterized by Thy-1–negative fibroblasts ([54]21, [55]24). However, what is unknown is how this subpopulation loses Thy-1 and whether Thy-1–negative fibroblasts contribute to the fibrotic encapsulation that occurs with implanted biomaterials. Fig. 1. Hydrogel fabrication and subcutaneous implantation. [56]Fig. 1. [57]Open in a new tab (A) Schematic describing regulatory mechanism of Thy-1 in fibroblast mechanotransduction. TGFBR, TGF-β receptor. (B) Illustration detailing subcutaneous injection regimen on the dorsal side of mice and a summary of tissue and cellular-level responses to MAP and NP PEG hydrogels. (C) H&E, Masson’s trichrome, and picrosirius red micrographs of MAP and NP hydrogels subcutaneously implanted into WT and Thy-1^−/− mice for 3 weeks; brackets indicate the fibrous capsule. (D) Quantification of fibrotic capsule thickness with multiple measurements across each micrograph, (E) Thy-1–positive fibroblasts, and (F) percent area of picrosirius red staining under polarized light. KO, knockout; ns, not significant. ****P < 0.0001, **P < 0.01, *P < 0.05, determined by two-way ANOVA with Tukey’s multiple comparison test. n = 5 mice per group; two sections per animal were analyzed. Scale bars, 100 μm. With the advent of single-cell transcriptomics, fibroblast heterogeneity is becoming better characterized with each new study ([58]25–[59]31). In addition to the heterogeneity of their cell state, there is also diversity in the origin of fibroblasts and myofibroblasts. Some critical fibroblast populations in dermal repair have been shown to mobilize from the underlying fascia ([60]32), while other studies have demonstrated that adipocytes and adipogenic progenitors are a source for myofibroblasts and are required for skin repair ([61]29, [62]33). As for the variety of cell subpopulations that can emerge from these different origins, the presence of immunologically active fibroblasts in both health and disease is becoming more clear, especially in the cancer field ([63]34–[64]38). However, we do not know whether immunologically active/inflammatory fibroblasts contribute to fibrosis. In this study, by using regenerative and nonfibrotic MAP gels compared to traditional fibrotic bulk nanoporous (NP) hydrogels and implanting them in wild-type (WT) versus Thy1^−/− mice, we demonstrate that, in the absence of Thy-1, MAP gels become as fibrotic as the NP gel. The fibrotic capsule around the NP gel in WT mice displayed increased interleukin-1 receptor I (IL-1RI) expression, α–smooth muscle actin (α-SMA) expression, and nuclear factor κB (NF-κB) p65 phosphorylation when compared to MAP gels. However, among Thy1^−/− mice, both MAP and NP gels displayed elevated IL-1RI expression, α-SMA expression, and NF-κB p65 phosphorylation, indicating that the absence of Thy-1 predisposed biomaterial implants to both inflammation and fibrosis and implicates Thy-1–negative fibroblasts at the center. We demonstrated that, while fibroblasts were cultured on a soft hydrogel substrate conjugated with fibronectin to recapitulate their native biophysical and biochemical microenvironment, interleuklin-1β (IL-1β) and tumor necrosis factor–α (TNFα) were able to promote a Thy-1–negative subpopulation that matched the previously reported phenotype of Thy-1–negative fibroblasts and that some cells were predisposed to Thy-1 loss while other cells were resistant to Thy-1 loss. This noted heterogeneity led us to use single-cell RNA sequencing (scRNA-Seq) to identify the subpopulations present with and without IL-1β and TNFα treatment. This demonstrated that two subpopulations emerged, which were defined by inflammatory cytokines or receptors among their differentially expressed genes, inflammatory pathways up-regulated via Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis, and low transcriptional expression of Thy-1. Together, these findings demonstrate that Thy-1–negative fibroblasts help bridge the gap between inflammation and dysfunctional mechanotransduction via emerging in response to inflammatory cytokine signaling and demonstrating a proinflammatory, profibrotic phenotype that is central to material-associated fibrosis. RESULTS Absence of Thy-1 promotes fibrosis in traditionally regenerative, immunoregulatory biomaterials To determine if expression of the αvβ3 integrin regulator Thy-1 ([65]Fig. 1A) contributes to biomaterial-mediated fibrosis, we specifically asked if Thy-1 loss is sufficient to reverse the nonfibrotic nature of the host response to MAP hydrogels ([66]6). While Thy-1 expression is present among both fibroblasts and lymphocytes, due to the absence of a reliable pan fibroblast marker and a previous work demonstrating that the loss of Thy-1 among lymphocytes has no effect on fibrosis ([67]39), we used Thy1^−/− mice to interrogate Thy-1 loss in biomaterial-mediated fibrosis. We leveraged Thy1^−/− mice and controlled the experiment with a standard fibrogenic NP bulk hydrogel with the same polyethylene glycol (PEG)–based chemistry as the implanted MAP gels ([68]Fig. 1B). As expected, when implanted subcutaneously into WT mice, the NP hydrogel promoted a fibrous capsule that was ~220 μm in thickness after 3 weeks, whereas the MAP hydrogel displayed an almost undetectable fibrous capsule that was 20 μm in thickness and retained its porous structure, as previously described (6). In stark contrast, when implanted into Thy1^−/− mice, both hydrogels mounted a robust fibrous encapsulation with no discernable or statistical difference between NP and MAP hydrogels ([69]Fig. 1, C and D, and fig. S1). Furthermore, we harvested and digested NP and MAP implants to determine the presence of Thy-1–positive fibroblasts at the implant site and there was significantly higher Thy-1–positive fibroblasts collected from MAP gels compared to NP gels ([70]Fig. 1E and fig. S2). Using picrosirius red staining imaged under polarized light, we were able to determine collagen deposition among each group. This captured the collagenous fibrotic capsule present around the NP hydrogel and both gels implanted into Thy1^−/− mice. The most amount of collagen was detected with the MAP gel, which can be attributed to tissue ingrowth into the MAP gel based on the depth of the collagen network within the hydrogel ([71]Fig. 1F). These findings are in keeping with prior evidence that loss of Thy-1 expression leads to a highly activated, ECM-synthetic fibroblast, even in normally quiescence-inducing soft environments. Thy-1–negative fibroblasts display high baseline levels of active αvβ3, are far more adhesive, and display rapid turnover of integrin adhesion clusters, which promotes elevated matrix assembly and contraction, both hallmarks of tissue fibrosis ([72]16, [73]20, [74]23). IL-1RI expression and NF-κB signaling correspond to α-SMA expression and fibrotic encapsulation Given the role of macrophage-derived foreign body giant cells in biomaterial responses, it is critical to understand how local fibroblasts contribute to the foreign body response. More specifically, Thy-1 loss is not predicted to affect inflammatory responses as Thy-1 loss in models of fibrosis is historically tied to mechanotransduction and integrin regulation ([75]16, [76]20, [77]21, [78]40). Although there is strong evidence to suggest that Thy1^−/− fibroblasts are prone to myofibroblastic differentiation and generation of fibrosis, an elevation in inflammation due to a fibroblast defect would be unexpected. This highlights a gap in our understanding of the nuanced relationship between inflammation and fibrosis. Certainly, for over a decade, it has been appreciated that skewing macrophage responses can lead to better tissue remodeling outcomes, and recently, it has been shown that recruitment of interleukin-33 type 2 myeloid cells and activation of adaptive immunity can drive biomaterial-associated regeneration ([79]9). However, these results further corroborate that immune signaling may not be unidirectional, from immune cells to stromal cells, but rather bidirectional ([80]41–[81]45). We further corroborated this within the biomaterial context by using a publicly available scRNA-Seq dataset that identifies immune and stromal cell subpopulations ([82]GSE175890) ([83]46) and used the Connectome package to infer cell-cell signaling based on ligand and receptor expression ([84]47). This analysis revealed that, within the biomaterial microenvironment, macrophages and fibroblasts display bidirectional signaling across multiple pathways (fig. S3). Among the pathways that came out of this analysis, the IL-1β–IL-1R signaling axis emerged between multiple subpopulations of macrophages and fibroblasts. We began unraveling the immune-stromal signaling axis by exploring IL-1R signaling. Chronic IL-1R signaling is elevated in fibrosis across various tissues ([85]48–[86]50). IL-1RI expression was clearly evident within the fibrotic capsule of the NP hydrogel and grossly absent in and around the MAP hydrogel within WT mice. In juxtaposition, IL-1RI expression was present and prominent in the fibrotic capsule of both NP and MAP hydrogels implanted in Thy1^−/− mice ([87]Fig. 2A). IL-1RI expression within fibrotic capsules corresponded with downstream NF-κB signaling, as shown by phospho-p65, and α-SMA expression, a marker of myofibroblast differentiation. Specifically, in NP hydrogels, there was notable coexpression of α-SMA and phosphorylated p65 within the fibrous capsule among WT mice, indicating a potential role for inflammatory signaling in the tissue remodeling phase in addition to the inflammatory phase in response to biomaterials. MAP hydrogels implanted in WT mice demonstrated very little detectable α-SMA expression or p65 phosphorylation at the biomaterial surface or among cells migrating into the implant. These differences are also reflected in regions with less fibrotic encapsulation of the NP hydrogel; there is less α-SMA expression and p65 phosphorylation (fig. S4, A and B). Consistent with our analysis of fibrotic outcomes, NP and MAP hydrogels both display robust α-SMA expression and p65 phosphorylation within their fibrous capsule when implanted in Thy1^−/− mice ([88]Fig. 2, B to D). The data clearly demonstrate that genetic knockout of Thy-1 is sufficient to overcome the regenerative and nonfibrotic nature of MAP hydrogels and illustrate a potential fibroblast cytokine signaling mechanism. Fig. 2. Inflammatory signaling among hydrogels in Thy-1^−/− mice. [89]Fig. 2. [90]Open in a new tab (A) Fluorescence images of MAP and NP hydrogel subcutaneous implants stained for IL-1RI (green) and DAPI (blue). (B) Fluorescence images staining for α-SMA (green), Thy-1 (red), phospho-p65 NF-κB (cyan), and DAPI (blue) with quantification for the sum of (C) α-SMA pixel values and (D) phospho-p65 NF-κB^+ cells per field of view (FOV). ***P < 0.001, **P < 0.01, *P < 0.05, determined with two-way ANOVA with Tukey’s multiple comparison test. n = 5 mice per group; two sections per animal were analyzed. Scale bars, 100 μm. IL-1β and TNFα promote fibroblast Thy-1 loss The evident differences in immune-stromal signaling between NP and MAP hydrogels and the ability of Thy1^−/− mice to effectively normalize the fibrotic responses between these two systems led us to hypothesize that inflammatory cytokines promote the loss of Thy-1 and a coincident activated myofibroblastic phenotype among fibroblasts. Using an unbiased approach with a publicly available RNA sequencing (RNA-Seq) dataset ([91]GSM2072332) ([92]51), we identified the cytokine receptors expressed on fibroblasts and determined the T helper 1 (T[H]1) [interleukin-1α (IL-1α), IL-1β, TNFα, and interferon-γ (IFN-γ)], T helper 2 (T[H]2) [interleukin-4, interleukin-6 (IL-6), and interleukin-13], and T helper 17 [interleukin-17 (IL-17)] cytokines that are capable of signaling within fibroblasts. Fibroblasts (CCL-210) treated with T[H]1 cytokines (IL-1α, IL-1β, IFN-γ, and TNFα) all display elevation in expression of the myofibroblastic marker α-SMA (fig. S5). We additionally observed significantly elevated fibroblast spreading on soft (2 kPa) fibronectin-coated substrates when treated with IL-1β and TNFα and elevated but not significantly increased spread area when treated with IL-17, IFN-γ, and transforming growth factor–β (TGF-β). Only IL-1β and TNFα, and to a lesser extent IFN-γ, induced a loss of Thy-1 surface expression, and only IL-1β and TNFα show consistently promoted α-SMA expression. These short-term, acute cytokine stimulations indicate immune-stromal signaling that results in alteration of fibroblast phenotype but fall short of establishing the emergence of a persistent Thy-1–negative fibroblast subpopulation. Cytokine-mediated loss of Thy-1 recapitulates the traditional Thy-1–negative phenotype We therefore treated fibroblasts, seeded on soft (~2 kPa) fibronectin-coated hydrogels longitudinally with IL-1β and TNFα individually or in combination for 3 to 7 days. As in short stimulations, IL-1β or TNFα treatment for 72 hours resulted in a significant increase in the Thy-1–negative population (~20% of the total population) compared to fibroblasts in media. The prominence of the Thy-1–negative population was enhanced when stimulated with a cocktail of IL-1β and TNFα ([93]Fig. 3A). Stimulation over a 7-day period further intensified the proportion of Thy-1–negative fibroblasts, reaching ~30% in IL-1β and TNFα treatments and the combination yielding fibroblasts that were 40% Thy-1–negative (fig. S6). Concomitant with Thy-1 loss, fibroblasts displayed significantly greater cell spreading on soft hydrogels stimulated with IL-1β and TNFα for 72 hours despite showing no discernable differences in focal adhesion size ([94]Fig. 3, B and C). Fibroblast spreading on soft hydrogels suggests altered mechanosensory signaling, and our observations here are consistent with our prior discovery of Thy-1’s role in enabling proper mechanotransduction and stiffness sensing in fibroblasts ([95]20). Only IL-1β stimulation leads to a significant increase in α-SMA expression ([96]Fig. 3D), suggesting a transition to a myofibroblast phenotype. This suggests cooperative, but not overlapping, roles between IL-1β and TNFα signaling in fibroblasts. Fig. 3. Cytokine-mediated Thy-1 loss. [97]Fig. 3. [98]Open in a new tab (A) Flow cytometric analysis of cytokine-treated CCL-210 human lung fibroblasts measuring the emergence of a Thy-1–negative fibroblast subpopulation. Representative histogram of six independent experiments. (B) Fluorescence confocal images of CCL-210 human lung fibroblasts on fibronectin-coated 2-kPa polyacrylamide hydrogels for 48 hours. Staining is for vinculin (green), F-actin (red), and DAPI (blue). (C) F-actin images were used to calculate cell area, and vinculin images were used to measure focal adhesion area. (D) Flow cytometric analysis of cytokine-treated CCL-210 human lung fibroblasts measuring α-SMA expression depicted as geometric mean fluorescence intensity (gMFI). (E) Measuring Thy-1 expression using flow cytometry in CCL-210 human lung fibroblasts that have been sorted based on Thy-1 expression to identify if Thy-1 loss is deterministic. FITC, fluorescein isothiocyanate. ****P < 0.0001, ***P < 0.001, *P < 0.05, determined with one-way ANOVA with Tukey’s multiple comparison test. n = 5 biological replicates. Scale bars, 100 μm. Thy-1^+ and Thy-1^− fibroblast subpopulations were stable and deterministic The existence of a true subpopulation implies both a deterministic emergence and population persistence. Given the grossly Thy-1^+ nature of naïve fibroblasts (only 5 to 10% are inherently Thy-1^−), this would imply that there is a hidden or cryptic fibroblast subpopulation predetermined to lose Thy-1 in response to IL-1β and/or TNFα that then persists in the absence of the stimuli and/or is resistant to additional stimulation. To establish the presence of a deterministic subpopulation, we treated fibroblasts for 72 hours with either IL-1β or TNFα. Fibroblasts were then sorted based on Thy-1 expression into negative and positive populations. Each population was then subsequently stimulated with either media alone or a second dose of the initial cytokine for another 48 hours. Thy-1^+ fibroblasts emerging from the initial cytokine stimulation were both stable, i.e., remained Thy-1^+ in subsequent culture in media alone, and deterministic, i.e., resistant to Thy-1 loss upon a second stimulation. Similarly, Thy-1^− fibroblasts resulting from IL-1β or TNFα stimulation were also stable and deterministic ([99]Fig. 3E). These data indicate that there are subpopulations within the native fibroblast population that are resistant to cytokine-induced loss of Thy-1 and others predisposed to lose Thy-1 and remain Thy-1–negative. The emergent Thy-1^− fibroblasts represent a cryptic subpopulation that emerge specifically in response to IL-1β and/or TNFα. scRNA-Seq reveals Thy-1^− fibroblasts possess a distinct inflammatory signature While Thy-1^+/− subpopulations help define the heterogeneity of fibroblasts, they certainly do not fully encompass the true heterogeneity within the population. Recently, scRNA-Seq analysis of fibroblasts from various tissues and diseases has elucidated as many as four to seven transcriptionally distinct fibroblast subpopulations ([100]30, [101]31, [102]52–[103]54). We similarly took this approach to uncover both the endogenous heterogeneity of our fibroblast population and how those subpopulations change and new subpopulations emerge (such as Thy-1^−) in response to inflammatory signaling. Our scRNA-Seq analysis (figs. S7 to S10) revealed five transcriptionally distinct clusters of cells among untreated fibroblasts cultured on soft (2 kPa) hydrogels, a degree of heterogeneity that matches what has been observed among fibroblasts in mouse and human tissues. Following 72-hour stimulation with IL-1β and TNF, two new clusters emerge (clusters 2 and 5) along with the expansion of an existing cluster (cluster 0; [104]Fig. 4, A and B), all of which possess a distinct transcriptional signature based on differential expression analysis ([105]Fig. 4C). At the transcriptional level, subpopulations 0, 2, and 5 also displayed reduced Thy-1 expression ([106]Fig. 4B). According to Slingshot pseudotime analysis that predicts cell trajectory ([107]55), cluster 0 represents an endogenous, native Thy-1 low population, whereas clusters 2 and 5 are the emergent, cryptic Thy-1 low subpopulations observed in prior experiments ([108]Fig. 4D). The top markers of clusters 0, 2, and 5 are inflammation related: IL-1RI, IL-6, IL-1β, interleukin-8 (IL-8), interleukin-33, and interleukin-11 (IL-11) (fig. S11 and [109]Fig. 4C). These transcriptional signatures were further confirmed by KEGG pathway enrichment analysis that identified overexpression of transcripts related to phosphatidylinositol 3-kinase (PI3K)–Akt, TNF, IL-17, and NF-κB signaling pathways ([110]Fig. 4E). Intriguingly, the top gene identifying cluster 5 is IL-1β, the inflammatory cytokine that induces Thy-1 loss and the emergence of the profibrotic fibroblast subpopulations. The top markers and KEGG pathway analysis for the Thy-1 low subpopulations indicates a clear inflammatory transcriptional signature. This inflammatory signature was further corroborated by analyzing a publicly available dataset ([111]GSE211834) from an in vivo bleomycin-induced skin fibrosis model where WT and Thy1^−/− dermal fibroblasts underwent RNA-Seq analysis. Using KEGG pathway analysis, we identified that Thy1^−/− dermal fibroblasts in this model displayed an enrichment for more inflammatory pathways (fig. S12), many of which were the same pathways that emerged in our scRNA-Seq dataset. Together, these data provide a compelling argument that initial signaling by T[H]1 cytokines associated with the acute inflammatory response to biomaterials induces a profibrotic immunofibroblast capable of driving a positive feedback autocrine signaling loop capable of sustaining fibrotic progression. Fig. 4. Cytokine-mediated changes in fibroblast heterogeneity. [112]Fig. 4. [113]Open in a new tab CCL-210 fibroblasts were treated with inflammatory cytokines on 2-kPa fibronectin-coated hydrogels for 72 hours. Cells were collected for scRNA-Seq. (A) Uniform Manifold Approximation and Projection (UMAP) clustering projection showing subpopulations of CCL-210 fibroblasts in media alone or treated with IL-1β and TNFα. (B) Violin plot demonstrating Thy-1 expression across subpopulations and fraction of cells coming from each subpopulation based on treatment group. (C) Violin plots of top 2 differentially expressed genes that serve as markers identified for each subpopulation. GJA1, gap junction protein alpha 1; TAGLN, transgelin; VIM, vimentin; COL8A1, collagen 8 alpha 1; COL1A1, collagen 1 alpha 1; COL1A2, collagen 1 alpha 2; CENPF, centromere protein F; TOP2A, topoisomerase II alpha; MGP, matrix Gla Protein. (D) Slingshot pseudotime analysis demonstrating cell trajectory throughout subpopulations. (E) KEGG pathway enrichment analysis identifying up-regulated pathways corresponding to differentially expressed genes for each cluster. ER, endoplasmic reticulum. DISCUSSION Biomaterial-mediated fibrosis and the foreign body response are the critical obstacles to implant success. There has been extensive work demonstrating the role chronic inflammatory signaling has in promoting implant failure and efforts to modulate the immune system to attenuate fibrosis. Hu et al. ([114]56) demonstrated that Toll-like receptor 2–modulating biomaterials demonstrated the ability to promote islet-xenograft survival. Chung et al. ([115]57) showed that reducing IL-17 signaling or treatment with a senolytic agent reduces the fibrotic response to implanted biomaterials. Another study demonstrated that modulating macrophage activation state is critical to promoting implant vascularization, a key step in promoting tissue ingrowth as opposed to fibrotic encapsulation ([116]58). Sadtler et al. ([117]59) showed that proregenerative biomaterials require T[H]2 cells to guide macrophage activation to guide tissue repair. While inflammatory signaling and various immune cell populations are critical to promoting or avoiding fibrotic encapsulation, it is the fibroblasts and not immune cells that are the primary contributors to scar formation. Therefore, it is critical to understand how immune-fibroblast cross-talk contributes to biomaterial-mediated fibrosis. Here, we demonstrate that implants that undergo fibrotic encapsulation display robust NF-κB signaling as well as IL-1RI^+ fibroblasts within the fibrous capsule. We demonstrate that IL-1β treatment promoted α-SMA expression, increased cell area indicating increased cell contractility, and loss of Thy-1 expression. Thy-1 loss has been shown to be critical to various forms of tissue fibrosis and promotes dysfunctional mechanotransduction among fibroblast subpopulations that undergo myofibroblastic differentiation independent of the mechanical cues from the surrounding microenvironment ([118]16, [119]20, [120]21, [121]60). When nonfibrotic NP gels and regenerative MAP gels were implanted in Thy1^−/− mice, these MAP gels induced a fibrotic response that resembled the encapsulation seen in response to bulk NP gels. Our data show that the ability to evade fibrotic remodeling by MAP gels is not necessarily a salient feature of the material per se and can be reversed by simply tuning the fibroblast cell milieu within the material microenvironment. scRNA-Seq determined that IL-1β and TNFα treatment altered the fibroblast subpopulations and promoted the emergence of a Thy-1–negative “immunofibroblast” defined by cytokine, chemokine, and cytokine receptor expression. These findings provide a critical step forward in our understanding of responses to implanted biomaterials by identifying a critical subpopulation of fibroblasts that can promote fibrosis in traditionally nonfibrotic materials. Furthermore, we identify a signaling axis that promotes the emergence of this potent fibrotic and immune-responsive fibroblast subpopulation. These findings might inform biomaterial design (i.e., targeting IL-1R signaling or Thy-1–negative fibroblasts) and present an opportunity for a previously unidentified in vitro approach for biomaterial diagnostics. One key limitation of this study is the use of global Thy-1 knockout mice as opposed to fibroblast-specific Thy-1 knockout mice. As the fibroblast field narrows in on a reliable pan-fibroblast marker, our future studies investigating immune-stromal cross-talk in biomaterial-mediated fibrosis will certainly use them. As is, our findings paint an interesting picture because they imply that if Thy-1 expression is restored, then perhaps traditionally fibrotic implants can become nonfibrotic. This hypothesis was validated in a recent study where soluble Thy-1 was delivered intratracheally and shown to reverse fibrosis ([122]61). Designing or modifying biomaterials to promote Thy-1 expression in an effort to avoid fibrotic encapsulation would be a critical next step in applying these findings. Macrophage/monocyte-to-myofibroblast transitions ([123]62), so-called “fibrocytes,” have been well described in fibrotic responses. Here, in an unexpected twist, elements of the transcriptional signature of these unique fibroblasts include traditional macrophage markers, possibly implicating a form of myofibroblast-to-macrophage transition such that fibroblasts are capable of sustaining a macrophage-like response well beyond the termination of the acute inflammatory phase. Immune-stromal communication has been previously reported in disease ([124]63) and endogenous repair ([125]29) where inflammation instructs fibrosis, yet our data suggest that fibroblasts can “talk back,” driving chronic inflammation. This inflammatory phenotype of fibroblasts has largely been observed in the context of cancer but has yet to be discussed within the biomaterials field ([126]34–[127]38). Tackling these unexpected and cryptic contributors to inflammation and fibrosis will be the key to future success in biomaterial development and the longevity of implant function. MATERIALS AND METHODS Hydrogel fabrication Four-arm PEG-maleimide (PEG-Mal; 10 and 20 kDa) was purchased from Nippon Oil Foundry Inc. (Japan) and used as the backbone. RGD peptide (Ac-RGDSPGGC-NH[2]) and the matrix metalloproteinase 2 (MMP-2) degradable cross-linker (Ac-GCGPQGIAGQDGG-NH[2]) were purchased from WatsonBio Sciences (United States). All materials were dissolved in either ultrapure water or 0.1% trifluoroacetic acid solutions (to prevent disulfide bond formation) and aliquoted at specific volumes to ensure precision. Aliquots were lyophilized and stored at −20°C until use. A heterofunctional maleimide/methacrylamide four-arm PEG macromer (MethMal) was synthesized as previously reported ([128]64). A 6.5 wt % gel was prepared by dissolving PEG-Mal, MethMal, and RGD in 10X phosphate-buffered saline (PBS) (pH = 2). This solution was combined with an MMP cross-linker and 5 μM biotin-maleimide, which were dissolved in ultrapure water. The final concentrations in the hydrogel solution were 81.31 mg/ml PEG-Mal, 14.90 mg/ml MMP, 0.80 mg/ml RGD, and 8.02 mg/ml MethMal. Microgels were produced using a microfluidic polydimethylsiloxane mold created using a previously published design ([129]65) in a dust-free hood. Briefly, a 1% Pico-Surf surfactant (Sphere Fluidics) solution diluted in NOVEC 7500 oil (3 M) was run through the oil channel and the gel formulations described above were in the aqueous channel. Using a syringe pump, the surfactant and gel solutions were run at 5 ml/hour in the device and collected in a conical tube. The resultant particles were mixed with a triethylamine (TEA) solution (20 μl of TEA/ml of gel) to increase the pH and accelerate gelation. Microgels were washed three times with NOVEC 7500 oil (1X volume of gel). Next, microgels were combined with PBS (5X volume of gel) and washed three times with NOVEC 7500 oil (1X volume of gel), allowing separation of the oil and aqueous solution by settling. Last, the oil was removed and the microgels were washed three times with PBS (3X volume of gel) and hexanes (3X volume of gel). After the final wash, microgels reacted overnight at 37°C with a 100 mM N-acetyl-l-cysteine solution in 1X PBS to cap any excess maleimides. Microgels were removed from solution via centrifugation (4696g x 5min), and all further steps were performed in a biosafety cabinet. Microgels were washed three times with 70% isopropyl alcohol followed by four washes with sterile 1X PBS (pH = 7.4) before being mixed with a photoinitiator solution. NP gel fabrication In a biosafety cabinet, an identical 6.5 wt % gel with a PEG-Mal backbone was prepared by dissolving PEG-Mal, MethMal, and RGD in sterile-filtered 2X PBS (pH = 6.74). The MMP cross-linker and 5 μM biotin-maleimide were dissolved in ultrapure water. The solutions were combined and loaded into a syringe for injection. The final concentrations in the NP gel solution were 81.31 mg/ml PEG-Mal, 14.90 mg/ml MMP, 0.80 mg/ml RGD, and 8.02 mg/ml MethMal. Mice and subcutaneous implantation of hydrogels All animal care and experiments followed guidelines by the University of Virginia’s Institutional Animal Care and Use Committee. C57BL/6 (WT) and Thy1^−/− mice were purchased from the Jackson Laboratory. Ten-week-old male and female mice were used for all studies. For subcutaneous implantation of hydrogels, 10-week-old C57BL/6 mice were anesthetized with aerosolized isoflurane and received three 80-μl subcutaneous dorsal-side injections of MAP hydrogel solution and three 80-μl injections of NP hydrogel solution as a contralateral control. After 3 weeks, mice were euthanized via carbon dioxide asphyxiation and the hydrogels and surrounding subcutaneous tissue were collected and frozen in optimal cutting temperature compound over dry ice. Sections (10 μm) were cut using a CryoStar NX50 cryostat and mounted on SuperFrost Plus slides for downstream histology and immunofluorescence. Histology and analysis Slide-mounted 10-μm sections of hydrogel samples were fixed with 4% paraformaldehyde and then stained with hematoxylin and eosin (H&E), Masson’s trichrome, and picrosirius red using standard procedures. The subsequent histology images were blinded and analyzed for fibrous capsule thickness, which we defined as the collagenous tissue surrounding the circumference of the hydrogel implant below the skin, and foreign body giant cell presence, which we defined as multinucleated cells within the fibrotic capsule with a large bundle or ring of nuclei ([130]66). Picrosirius red staining was imaged under polarized light using a Zeiss Axioscan 7 slide scanner, and collagen deposition was quantified as percent area of positive picrosirius red staining. Masson’s trichrome and picrosirius red staining was done with the help of the Research Histology core at University of Virginia (UVA). Tissue immunofluorescence and quantification Slide-mounted 10-μm sections were fixed with 4% paraformaldehyde and blocked with 3% normal goat serum (NGS) + 0.1% Triton in 1X PBS for intracellular antigens. Primary antibody dilutions were prepared in 3% NGS in 1X PBS as follows: anti-mouse α-SMA (1A4, Thermo Fisher Scientific) at 1:200, rat anti-mouse Thy-1.2 (53-2.1, BD Biosciences) at 1:100, rat anti-mouse phospho–NF-κB p65 (Ser^536; 93H1, Cell Signaling Technologies) at 1:100, and biotinylated anti-mouse IL-1RI (JAMA-147, BioLegend) at 1:100. The primary antibody incubation was done overnight at 4°C, then washed three times with 1X PBS, and then stained with a secondary antibody for 1 hour at room temperature. The secondary antibodies (goat anti-mouse Alexa Fluor 488, goat anti-rat Alexa Fluor 555, and goat anti-rabbit Alexa Fluor 647; Invitrogen) were all prepared in 3% NGS in 1X PBS at a dilution of 1:1000. 4′,6-Diamidino-2-phenylindole (DAPI) was used as a counterstain at 300 nM in 1x PBS for 5 min at room temperature and then mounted in mounting medium ProLong Diamond Antifade (Thermo Fisher Scientific). Slides were imaged on a Keyence BZ-X810 microscope using the 10X objective. To account for autofluorescence, each sample was treated with the Vector TruView autofluorescence quenching kit to diminish autofluorescence (Vector Laboratories). α-SMA expression was quantified by defining a region of interest in Fiji around the fibrotic capsule within the field of view and measuring the sum of the α-SMA signal pixel intensity as defined by raw integrated density in Fiji. Phospho-p65^+ cells were measured by recording the number of cells that expressed phosphop-p65 in thresholded images per field of view using the “Analyze Particles” function in Fiji ([131]67). Exposure settings and thresholding approach were the same across all images and incorporated the use of primary delete images (fig. S13). Antibodies were validated via immunofluorescence and flow cytometry on lung tissue, which is known to be high in all the observed markers. Polyacrylamide hydrogels for cell culture Human lung fibroblasts (CCL-210) were purchased from the American Type Culture Collection were cultured [Dulbecco’s modified Eagle’s medium, 10% fetal bovine serum (FBS), and 1% penicillin/streptomycin] and used between passages 2 and 10. To recapitulate the biophysical microenvironment and remove substrate stiffness as a conflating factor in this study, fibroblasts were cultured on 2-kPa polyacrylamide hydrogels that were purchased from Matrigen and came chemically activated ready to bind matrix proteins and were subsequently coated with fibronectin (10 μg/ml). For immunocytochemistry of fibroblasts on hydrogels, cells were fixed with 4% paraformaldehyde and blocked with 3% NGS + 0.1% Triton in 1X PBS for intracellular antigens. Primary antibody dilutions were prepared in 3% NGS in 1X PBS as follows: mouse anti-human vinculin (VIN-54, Abcam) at 1:200 and phalloidin conjugated with Alexa Fluor 546 (Invitrogen). The primary antibody incubation was done for 1 hour at room temperature, then washed three times with 1X PBS, and then stained with a secondary antibody for 1 hour at room temperature. The secondary antibody (goat anti-mouse Alexa Fluor 488, Invitrogen) was prepared in 3% NGS in 1X PBS at a dilution of 1:1000. DAPI was used as a counterstain at 300 nM in 1x PBS for 5 min at room temperature and then mounted in mounting medium ProLong Diamond Antifade (Thermo Fisher Scientific). Imaging was done at room temperature on a Nikon Eclipse Ti microscope with an UltraView VoX imaging system (PerkinElmer) using a Nikon N Apo LWD 40X water objective (numerical aperature: 1.15). Cell and focal adhesion area were measured using Fiji software. Flow cytometry and FACS Fibroblasts were lifted from hydrogels for flow cytometry using TrypLE Express enzyme (Thermo Fisher Scientific) and subsequently stained with the Zombie Near-Infrared Fixable Viability dye (BioLegend) then fixed and permeabilized using the FIX & PERM Cell Permeabilization Kit (Thermo Fisher Scientific). After fixation, fibroblasts were surface stained for Thy-1 using the allophycocyanin (APC) anti–Thy-1 antibody (1:100, BioLegend). For α-SMA staining, after surface staining and washing, fibroblasts were incubated in the permeabilization buffer with the phycoerythrin anti–α-SMA antibody (1A4, R&D) at a 1:25 dilution. For analyzing cells from implanted biomaterials, implants were digested with Liberase TM (Roche) and stained with BV711 anti-CD45 antibody (1:100, BioLegend), BV605 anti-EpCAM antibody (1:100, BioLegend), and BV421 anti-CD31 antibody (1:100, BioLegend) to yield a fibroblast population that was defined as CD45^−, EpCAM^−, and CD31^− and subsequently stained with APC anti–Thy-1 antibody. All staining was done using fluorescence-activated cell sorting (FACS) buffer (1X PBS with 10% FBS and 0.05% sodium azide) and staining was done in the presence of Fc block (1:100, BioLegend). Flow cytometry was done on a BD LSR Fortessa, and cell sorting was done using an Influx Cell Sorter through the UVA Flow Cytometry Core Facility (RRID: SCR_017829). scRNA-Seq and analysis Fibroblasts were lifted from hydrogels using TrypLE Express enzyme, and viability was confirmed to be greater than 80% using Trypan blue staining with the Countess II FL Automated Cell Counter (Thermo Fisher Scientific). Cells were resuspended in 0.04% UltraPure BSA in PBS (Thermo Fisher Scientific). A total of 1500 fibroblasts in each group were targeted. After 72 hours of cytokine treatment, cells will be collected and then we used the 10X Genomics Chromium platform for automated single-cell barcoding and library preparation on an eight-channel microfluidic chip. Then, we used the Illumina NextSeq 500 Sequencing System for high-throughput sequencing that allows for 400M reads, permitting us to have broad coverage of each cell’s transcriptome. The Genome Analysis and Technology Core at UVA (RRID: SCR_018883) assisted in single-cell barcoding, library construction, and sequencing. Gene-barcode matrices were analyzed in R using Seurat v3 ([132]68). Cells were filtered for 2500 to 9000 reads per unique molecular identifiers and less than 10% of mitochondrial gene content. Significant principal components analysis of variation was calculated using JackStraw ([133]69) test with 100 repetitions, and clusters were defined using 20 principal components of variation. Our analysis was done combining together the untreated and treated groups so that we might be able to simultaneously analyze all subpopulations. Slingshot pseudotime trajectory analysis was used to determine the trajectory of fibroblasts across subpopulations ([134]55). KEGG pathway analysis uses transcript information to understand higher-order changes in pathways or biological systems, and this analysis was done for both the scRNA-Seq dataset and the publicly available dataset comparing WT versus Thy1^−/− dermal fibroblasts ([135]GSE211834). We used the Connectome package ([136]47) that helps explore cell-cell signaling networks within scRNA-Seq datasets and applied it to a scRNA-Seq dataset ([137]GSE175890) that compares different implanted biomaterials in a volumetric muscle loss model to identify bidirectional signaling between macrophages and fibroblasts in the presence of biomaterials ([138]46). Our analysis for differential expression analysis was determined using the nonparametric Wilcoxon rank sum test. Code is available on request. Statistical analysis Statistics were performed using GraphPad Prism 9 (GraphPad, San Diego, CA). For multiple comparisons along one variable, we performed one-way analysis of variance (ANOVA) followed by Tukey’s post hoc test. For multiple variable comparisons, we performed two-way ANOVA followed by Tukey’s post hoc test. The number of mice used for each analysis was determined by power analysis (power = 0.80, α = 0.05). Statistical significance was defined at P values < 0.05. Data were visualized, and statistical analysis was done using GraphPad Prism (version 9). Acknowledgments