Category Archives: Adult Stem Cells

COVID-19 impact: Stem Cells Market to Witness Steady Expansion During 2019-2026 – Personal Injury Bureau UK

The report on the Stem Cells market provides a birds eye view of the current proceeding within the Stem Cells market. Further, the report also takes into account the impact of the novel COVID-19 pandemic on the Stem Cells market and offers a clear assessment of the projected market fluctuations during the forecast period. The different factors that are likely to impact the overall dynamics of the Stem Cells market over the forecast period (2019-2029) including the current trends, growth opportunities, restraining factors, and more are discussed in detail in the market study.

The Stem Cells market study is a well-researched report encompassing a detailed analysis of this industry with respect to certain parameters such as the product capacity as well as the overall market remuneration. The report enumerates details about production and consumption patterns in the business as well, in addition to the current scenario of the Stem Cells market and the trends that will prevail in this industry.

Get Free Sample PDF (including COVID19 Impact Analysis, full TOC, Tables and Figures) of Market Report @ https://www.researchmoz.com/enquiry.php?type=S&repid=2649650&source=atm

What pointers are covered in the Stem Cells market research study?

The Stem Cells market report Elucidated with regards to the regional landscape of the industry:

The geographical reach of the Stem Cells market has been meticulously segmented into United States, China, Europe, Japan, Southeast Asia & India, according to the report.

The research enumerates the consumption market share of every region in minute detail, in conjunction with the production market share and revenue.

Also, the report is inclusive of the growth rate that each region is projected to register over the estimated period.

The Stem Cells market report Elucidated with regards to the competitive landscape of the industry:

The competitive expanse of this business has been flawlessly categorized into companies such as

Regional and Country-level Analysis The report offers an exhaustive geographical analysis of the global Stem Cells market, covering important regions, viz, North America, Europe, China, Japan, Southeast Asia, India and Central & South America. It also covers key countries (regions), viz, U.S., Canada, Germany, France, U.K., Italy, Russia, China, Japan, South Korea, India, Australia, Taiwan, Indonesia, Thailand, Malaysia, Philippines, Vietnam, Mexico, Brazil, Turkey, Saudi Arabia, UAE, etc. The report includes country-wise and region-wise market size for the period 2015-2026. It also includes market size and forecast by each application segment in terms of revenue for the period 2015-2026. Competition Analysis In the competitive analysis section of the report, leading as well as prominent players of the global Stem Cells market are broadly studied on the basis of key factors. The report offers comprehensive analysis and accurate statistics on revenue by the player for the period 2015-2020. It also offers detailed analysis supported by reliable statistics on price and revenue (global level) by player for the period 2015-2020. On the whole, the report proves to be an effective tool that players can use to gain a competitive edge over their competitors and ensure lasting success in the global Stem Cells market. All of the findings, data, and information provided in the report are validated and revalidated with the help of trustworthy sources. The analysts who have authored the report took a unique and industry-best research and analysis approach for an in-depth study of the global Stem Cells market. The following players are covered in this report: CCBC Vcanbio Boyalife Beikebiotech Stem Cells Breakdown Data by Type Umbilical Cord Blood Stem Cell Embryonic Stem Cell Adult Stem Cell Other Stem Cells Breakdown Data by Application Diseases Therapy Healthcare

Do You Have Any Query Or Specific Requirement? Ask to Our Industry [emailprotected] https://www.researchmoz.com/enquiry.php?type=E&repid=2649650&source=atm

Exclusive details pertaining to the contribution that every firm has made to the industry have been outlined in the study. Not to mention, a brief gist of the company description has been provided as well.

Substantial information subject to the production patterns of each firm and the area that is catered to, has been elucidated.

The valuation that each company holds, in tandem with the description as well as substantial specifications of the manufactured products have been enumerated in the study as well.

The Stem Cells market research study conscientiously mentions a separate section that enumerates details with regards to major parameters like the price fads of key raw material and industrial chain analysis, not to mention, details about the suppliers of the raw material. That said, it is pivotal to mention that the Stem Cells market report also expounds an analysis of the industry distribution chain, further advancing on aspects such as important distributors and the customer pool.

The Stem Cells market report enumerates information about the industry in terms of market share, market size, revenue forecasts, and regional outlook. The report further illustrates competitive insights of key players in the business vertical followed by an overview of their diverse portfolios and growth strategies.

You can Buy This Report from Here @ https://www.researchmoz.com/checkout?rep_id=2649650&licType=S&source=atm

Some of the Major Highlights of TOC covers:

See the original post:
COVID-19 impact: Stem Cells Market to Witness Steady Expansion During 2019-2026 - Personal Injury Bureau UK

Nuclear softening expedites interstitial cell migration in fibrous networks and dense connective tissues – Science Advances

INTRODUCTION

After injury or tissue damage, cells must migrate to the wound site and deposit new tissue to restore function (1). While many tissues provide a permissive environment for such interstitial [three-dimensional (3D)] cell migration (i.e., skin), adult dense connective tissues (such as the knee meniscus, articular cartilage, and tendons) do not support this migratory behavior. Rather, the extracellular matrix (ECM) density and micromechanics increase markedly with tissue maturation (2, 3) and, as a consequence, act as a barrier for cells to reach the wound interface. It follows then that healing of these tissues in adults is poor (4, 5) and that wound interfaces remain susceptible to refailure over the long term due to insufficient repair tissue formation. Similarly, fibrous scaffolds used in repair applications also impede cell infiltration when the scaffolds become too dense (6).

This raises an important conundrum in dense connective tissues and repair scaffolds; while the dense ECM and fibrous scaffold properties are critical for mechanical function, they, at the same time, can compromise cell migration, with endogenous cells locked in place and unable to participate in repair processes. This concept is supported by in vitro studies documenting that, in 3D collagen gels, the migration of mesenchymal lineage cells is substantially attenuated once the gel density and/or stiffness has reached a certain threshold (79). Consistent with this, our recent in vitro models exploring cell invasion into devitalized dense connective tissue (knee meniscus sections) showed reduced cellular invasion in adult tissues compared to less dense fetal tissues (3). The density of collagen in most adult dense connective tissues is 30 to 40 times higher than that used within in vitro collagen gel migration assay systems (2, 3), emphasizing the substantial barrier to migration that the dense ECM plays in these tissues.

To address this ECM impediment to successful healing, we and others have developed strategies to loosen the matrix (via local release of degradative enzymes) in an attempt to expedite repair and/or encourage migration to the wound site (10), with promising results both in vitro and in vivo (10, 11). Despite the potential of this approach, it is cognitively dissonant to disrupt ECM to repair it, and any such therapy would have to consider any adverse consequences on tissue mechanical function.

This led us to consider alternative controllable parameters that might regulate interstitial cell mobility while preserving the essential mechanical functionality of the matrix. It is well established that increasing matrix density decreases the effective pore size within dense connective tissues. The nucleus is the largest (and stiffest) organelle in eukaryotic cells (12), and it must physically deform as a cell passes through constructures that are smaller than its own smallest diameter (9). When artificial pores of decreasing diameter are introduced along an in vitro migration path (e.g., in an in vitro Boyden chamber system), cell motion can be completely arrested (13). If cells are forced to transit through these tight passages, then nuclear rupture and DNA damage can occur (14, 15). Conversely, under conditions where nuclear stiffness is low, as is the case in neutrophils (16) and some particularly invasive cancer cells (17), migration through small pores occurs quite readily.

Given the centrality of the nucleus in migration through small pores, methods to transiently regulate nuclear stiffness or deformability might therefore serve as an effective modulator of interstitial cell migration through dense tissues and scaffolds. Nuclear stiffness is defined by two primary featuresthe density of packing of the genetic material contained within (i.e., the heterochromatin content) and the intermediate filament network that underlies the nuclear envelope (the nuclear lamina, composed principally of the proteins Lamin B and Lamin A/C) (12, 16, 18, 19). Increasing chromatin condensation increases nuclear stiffness, while decreasing Lamin A/C content decreases nuclear stiffness (19, 20). Both increasing the stiffness of the microenvironment in which a cell resides (21) and the mechanical loading history of a cell promotes heterochromatin formation and Lamin A/C accumulation (2224), resulting in stiffer nuclei. Since both matrix stiffening and mechanical loading are features of dense connective tissue maturation, these inputs may drive nuclear mechanoadaptation (25), resulting in endogenous cells with stiff nuclei that are locked in place.

On this basis, the goal of this study was to determine whether nuclear softening could enhance migration through dense connective tissues and repair scaffolds to increase colonization of the wound site and the potential for repair by endogenous cells. We took the approach of transiently decreasing nuclear stiffness in adult meniscus cells through decreasing heterochromatin content [using Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor] that promotes chromatin relaxation (26) and confirmed the importance of nuclear stiffness by reducing Lamin A/C protein content (using lentiviral-mediated knockdown). Our experimental findings and theoretical models demonstrate that nuclear softening decreases the barriers to interstitial migration through small pores, both in vitro and in vivo, resulting in the improved colonization of dense fibrous networks and transit through native tissue by adult meniscus cells. By addressing the inherent limitations to repair imposed by nuclear mechanoadaptation that accompanies cell differentiation and ECM maturation, this work defines a promising strategy to promote the repair of damaged dense connective tissues in adults.

We first determined whether TSA treatment alters chromatin organization in adult meniscal fibrochondrocytes (MFCs). Super-resolution images of the core histone protein Histone-H2B in MFC nuclei were obtained by stochastic optical reconstruction microscopy (STORM) and revealed a notable organization of Histone-H2B inside MFC nuclei (STORM; Fig. 1A), which could not be observed with conventional microscopy (conventional; Fig. 1A). It has recently been shown that super-resolution images can be segmented at multiple length scales using Voronoi tessellation (27, 28). To segment the H2B super-resolution images, we carried out Voronoi tessellation, used a threshold to remove large polygons corresponding to regions of the nucleus containing sparse localizations, and color-coded the localizations with the same color if their polygons were connected in space and shared at least one edge. This segmentation showed that H2B localizations clustered to form discrete and spatially separated nanodomains in control nuclei [()TSA]. Nuclei treated with TSA, on the other hand, contained smaller domains. These results were quantitatively recapitulated by a decrease in the number of H2B localizations in individual domains and an overall decrease in the area of domains in MFCs treated with TSA [(+)TSA] (Fig. 1, B to D). These results are in line with a more folded chromatin confirmation in ()TSA cells, which opens and decondenses after TSA treatment. These results are also consistent with recent super-resolution analysis, which showed that TSA-treated fibroblasts have small nucleosome nanodomains that are more uniformly distributed in the nuclear space compared to control fibroblasts (29, 30). This decondensation was also confirmed in TSA-treated bovine mesenchymal stem cells (MSCs), where TSA treatment decreased the number and area of H2B nanodomains (fig. S1A). This increased acetylation at H3K9 (Ac-H3K9) was apparent at the nanoscale (fig. S1B) and via conventional fluorescence imaging of the nuclei (fig. S1C). Conversely, there were no significant changes in H3K27me3 with TSA treatment when evaluated using STORM or conventional fluorescent microscopy (fig. S1, D and E).

(A) Representative conventional fluorescent and STORM imaging of Histone-H2B in a control [top; ()TSA] or TSA-treated MFC nucleus [bottom; (+)TSA]. (B) Corresponding Voronoi-based image segmentation, which allows for visualization and quantification of Histone-H2B nanodomains. (C and D) Quantification of the number of H2B localizations per cluster and the cluster area with TSA treatment. The box, line, and dot correspond to the interdecile range (10th to 90th percentile), median, and mean, respectively, Mann-Whitney U test, n 10,584 clusters from five cells. Next to each Voronoi image, higher-magnification zoom-ins of the region inside the squares are shown. (E) TSA treatment for 3 hours decreases chromatin condensation in 4,6-diamidino-2-phenylindole (DAPI)stained nuclei (scale bar, 5 m), and the number of visible edges (left). Quantification of the chromatin condensation parameter (CCP) with TSA treatment [right; *P < 0.05 versus ()TSA, n = ~20]. (F) Schematic showing experimental design to evaluate nuclear deformability and changes in nuclear aspect ratio (NAR = b/a) with cell stretch. (G) Representative DAPI-stained nuclei on scaffolds before and after 15% stretch (left; scale bar, 20 m) and NAR at 3 and 15% stretch (n = 32 to 58 cells, *P < 0.05 versus ()TSA and +P < 0.05 versus 3%). (H) 2D wound closure assay shows no differences in gap filling in the presence or absence of TSA [()TSA; left: scale bar, 200 m; right: P > 0.05, n = 6). (I) Schematic of Boyden chamber chemotaxis assay (left) and migrated cell signal intensity through 3-, 5-, and 8-m-diameter pores, with and without TSA pretreatment [right; n = 5 samples per group, *P < 0.05 versus ()TSA and +P < 0.05 versus 3 m, means SD]. All experiments were carried out at least in triplicate, except for the wound closure assay (which was performed in duplicate). RFU, relative fluorescence units.

In addition, TSA treatment for 3 hours [(+)TSA] also resulted in marked chromatin decondensation in MFCs seeded on aligned (AL) nanofibrous scaffolds that are commonly used for dense connective tissue repair, as evidenced by decreases in the number of visible edges in 4,6-diamidino-2-phenylindole (DAPI)stained nuclei compared to control cells [()TSA] and a reduction (~40%) in the image-based chromatin condensation parameter (CCP) (Fig. 1E).

To assess whether this TSA-mediated chromatin decondensation changed nuclear stiffness and deformability, we stretched MFC-seeded AL scaffolds (from 0 to 15% grip-to-grip strain) and determined the change in nuclear aspect ratio (NAR) (Fig. 1F). Nuclei that were pretreated with TSA [(+)TSA] showed increased nuclear deformation compared to control nuclei [()TSA] (Fig. 1G); however, TSA did not change cell/nuclear morphology (fig. S2, A to C) or cell migration on planar surfaces (Fig. 1H), and only minor changes in focal adhesions were observed (fig. S2, D and E). MFC spread area and traction force generation were also unaffected by TSA treatment when cells were plated on soft substrates (E = 10 kPa) (fig. S2, F to I). These observations suggest that TSA treatment decreases nuclear deformability by chromatin decondensation without changing overall cell migration capacity in 2D culture.

We next assessed the ability of MFCs to migrate through small pores using a commercial transwell migration assay (Fig. 1I). Cells treated with TSA [(+)TSA] (200 ng/ml) showed enhanced migration compared to controls [()TSA] across all pore sizes, including 3-m pores that supported the lowest migration in controls (Fig. 1I). This improved migration with TSA treatment was dose dependent (fig. S3). Together, these data show that while TSA treatment does not change cell morphology, contractility, or planar migration on 2D substrates, chromatin relaxation increases MFC nuclear deformability, which improves cell migration through micron-sized pores.

Having observed increased migration through rigid micron-sized pores with nuclear softening, we next assayed whether TSA treatment would enhance migration through dense fibrillar networks. A custom microfluidic cell migration chamber was designed, consisting of a top reservoir containing basal medium (BM), a bottom reservoir containing BM supplemented with platelet-derived growth factor (PDGF) as a chemoattractant and an interposed nanofibrous poly(-caprolactone) (PCL) layer (labeled with CellTracker Red, ~150-m thickness) (Fig. 2, A and B). With this design, a gradient of soluble factors is presented across the fibrous layer, as evidenced by Trypan blue diffusion over time (Fig. 2C).

(A) Schematic (top) and a top view (bottom) of the PDMS [poly(dimethylsiloxane)]/nanofiber migration chamber. (B) Schematic showing meniscus cells (green) seeded onto fluorescently labeled nanofibers interposed between the top reservoir containing BM and a bottom reservoir containing BM supplemented with PDGF (100 ng/ml) as a chemoattractant. (C) Visual representation of soluble factor gradient in microdevice showing the slow accumulation of trypan blue in the upper chamber as a function of time. (D) Experimental schematic showing meniscus cell (MFC) isolation and seeding onto nanofiber substrates (passage 1, isolated from adult bovine menisci). One day after seeding, TSA or PDGF was added to the top reservoir or the bottom reservoir, respectively, and cells were cultured for additional 2 days. On day 3, scaffolds were imaged by confocal microscopy to determine the degree of cell penetrance into the scaffold. (E) 3D confocal reconstructions of cell (green) migration through AL or non-AL (NAL) nanofibrous networks (AL or NAL; red) with and without TSA treatment. Scale bar, 30 m. (F) Cross-sectional views of cells (green) within nanofibrous substrates (red). Scale bar, 30 m. Quantification of the percentage of infiltrated cells (G) [n = 5 to 8 images, *P < 0.05 versus ()TSA and +P < 0.05 versus AL, means SD] and cell infiltration depth (H) [n = 33 cells, *P < 0.05 versus ()TSA and +P < 0.05 versus AL, means SEM, normalized to the ()TSA/AL group]. Quantification of the percentage of infiltrated cells (I) [n = 5 images, *P < 0.05 versus ()TSA, P < 0.05 versus 0% poly(ethylene oxide) (PEO), and aP < 0.05 versus 25% PEO, means SD] and cell infiltration depth (J) [n = 33 cells, *P < 0.05 versus ()TSA, P < 0.05 versus 0% PEO, and aP < 0.05 versus 25% PEO, means SD] normalized to the control PCL/0% PEO group] as a function of PEO content. All experiments were carried out in triplicate.

MFCs were seeded atop the fibrous layer, and their migration was evaluated as a function of nuclear deformability (TSA) and fiber alignment [AL or non-AL (NAL)]. MFCs were cultured in BM for 1 day for attachment and then were treated for 2 days either with or without TSA (Fig. 2D). Confocal imaging (Fig. 2, E and F, and movie S1, A and B) and scanning electron microscopy (fig. S4A) showed increased MFC invasion into the fibrous networks with TSA treatment [(+)TSA] when compared to untreated MFCs [()TSA]. Without TSA, MFCs remained largely on the surface of the fibers with some cytoplasmic extensions into the fibers (fig. S4B), whereas TSA treatment increased the number of nuclei entering the fiber network (fig. S4C). When quantified, infiltration was higher in the NAL group compared to the AL group (P < 0.05; Fig. 2, G and H), likely due to the increased pore size in the NAL scaffolds (6, 31), and TSA treatment improved migration to similar levels in both NAL and AL groups (P < 0.05; Fig. 2, G and H). As expected, cells in AL scaffolds showed higher aspect ratios and solidity compared to cells on NAL scaffolds, yet TSA treatment did not influence cell morphology (fig. S4D). Nuclei in NAL groups were rounder (lower NAR) than in AL groups, and TSA treatment resulted in more elongated nuclei (higher NAR) in both AL and NAL groups (fig. S4E). While promoting cell invasion, TSA treatment did not result in any change in DNA damage (as assessed by phospho-histone H2AX-positive nuclei; fig. S4F) and slightly reduced cell proliferation at this time point (fig. S4G). Thus, it appears that TSA increased nuclear deformability, resulting in enhanced cell migration into these dense fibrous networks.

To verify that nuclear softening is the primary mechanism for enhanced migration into fibrous networks, we also knocked down Lamin A/C in MFCs before seeding. In previous studies, cells lacking Lamin A/C showed increased nuclear deformability and increased mobility in collagen gels and through small pores in Boyden chambers (13, 32). Consistent with these studies (12, 19, 33), reduction of Lamin A/C protein levels in MFCs and MSCs (fig. S5, A to C) increased nuclear deformability in response to applied stretch (fig. S5D). When MFCs with Lamin A/C knockdown were seeded onto fibrous networks, a greater fraction entered into the scaffold and reached greater infiltration depths (fig. S5, E to G). To further illustrate that nuclear stiffening reduces migration, we cultured MSCs in transforming growth factor3 (TGF-3)containing media for 1 week before seeding onto the fibers. As we reported previously (23), these conditions induce differentiation in MSCs, resulting in stiffer nuclei with increased chromatin condensation and decreased nuclear deformability. Compared to undifferentiated MSCs, these differentiated MSCs were found largely on the scaffold surface (fig. S6, A to D) and had a lower infiltration rate and depth. While many factors change during cell differentiation, these findings also support that a less deformable nucleus is an impediment to interstitial cell migration. Together, these studies support that a stiff nucleus is a limiting factor in the invasion of the small pores of dense fibrous networks.

To investigate the combined role of porosity and nuclear softening on migration, we next fabricated fibrous networks through the combined electrospinning of both PCL and poly(ethylene oxide) (PEO), where PEO acts as a sacrificial fiber fraction to enhance porosity (6, 31). Consistent with our previous findings, cell infiltration percentage and depth progressively increased as a function of increasing PEO content (Fig. 2, I and J). When nuclei were softened with TSA treatment, we observed greater infiltration into low-porosity scaffolds (PEO content, <25%), but no difference in high porosity scaffolds (Fig. 2, I and J). This suggests that increasing nuclear deformability is only beneficial in the context of dense networks, where the nucleus impedes migration.

To better define the relationship between pore size and nuclear stiffness on cellular migration, we developed a computational model to predict the critical force (Fc) required for the nucleus to enter a small channel (Fig. 3). This model was motivated by studies of cellular transmigration through endothelium in the context of cancer invasion, where the surrounding matrix properties (stiffness), endothelium properties (stiffness and pore size), and the cell properties (in particular, the nuclear stiffness) appear to regulate transmigration (34). Here, we considered cell migration into a narrow and long channel to mimic migration into a porous fiber network, where network properties are defined by fiber density (Fig. 3A). When the cell enters the channel, the resistance force encountered by the nucleus increases monotonically as the cell advances, reaching a maximal resistance force (defined as the critical force, Fc). Following this, the nucleus snaps through the opening, leading to a drop in the resistance force, which vanishes after the nucleus fully enters the channel (Fig. 3B and movie S2). Thus, the cells must generate a sufficient force to overcome this critical force to migrate into a channel. As the channel size (rg) becomes smaller and the ECM modulus (EECM) becomes greater, the critical force required for the nucleus to enter the channel increases (Fig. 3C and fig. S7). As this required force increases, it eventually exceeds the force generation capacity of the cell, resulting in a situation where the cell cannot enter the pore.

(A) Schematic showing a nucleus (blue) above a narrow channel representing the small pores in a dense fiber network (orange). The geometric parameters are the radius of the nucleus (rn) and the half width of the channel (rg). The stiffness parameters are the modulus of the nucleus (En) and the fiber network (EECM). The nucleus is treated as a spheroid for simplicity. (B) Simulation of a nucleus moving into and through the channel in the dense fiber network. The normalized resistant force (F/Enrn2) encountered by the nucleus is plotted as a function of the normalized displacement of the nucleus (un/rn). The maximum normalized resistance force is defined as the critical force. (C) The critical force as a function of the normalized ECM modulus (with respect to En) and normalized channel size (with respect to rn). The critical force is larger as the ECM becomes stiffer or the channel becomes smaller. (D) The critical force decreases as the PEO content increases. TSA treatment also decreases the critical force, particularly for dense networks (low PEO content). (E) Normalized NAR after entry into the channel increases as the ECM becomes stiffer or the nucleus becomes softer (both lead to a larger normalized ECM modulus, EECM/En).

To better understand the influence of PEO content (affecting both the channel size and ECM modulus) and dose of TSA (affecting nuclear modulus) on cell migration, we used the normalized critical force data obtained from the model. Our previous work (6) defined the influence of PEO content on matrix mechanical properties and pore size; the effect of TSA on nuclear stiffness has also been measured quantitatively by other groups (26). Using these data, we predicted the critical force at different PEO contents for both TSA-treated and control cells (Fig. 3D). Results from this model showed that critical force decreased monotonically as PEO content increased, given that a higher PEO content results in larger pores (31). This indicates that infiltrated cell numbers should increase as the PEO content increases, consistent with our experimental results. Likewise, since TSA results in a softer nucleus (26), the critical force drops significantly compared to control conditions. This is particularly important at low PEO contents (denser networks), where the critical force for TSA-treated nuclei drops markedly. In networks with larger pores, the difference in critical force between TSA-treated groups vanishes. We included the model to gain, in general, insight into how a change in nuclear deformability (with TSA) might broadly affect cell migration in 3D and chose a simple configuration to gain some initial insight. While this model is simple (i.e., it does not represent the geometry of our fiber networks or native tissue), its predictions were consistent with our experimental findings, where the percentage of infiltrated cells was higher with TSA treatment at 0% PEO but the difference between groups disappeared at 50% PEO (Fig. 2I). The model also predicted that the NAR (after fully embedded in the channel) should increase as the nucleus becomes softer or the ECM becomes stiffer [with both resulting in a larger normalized ECM modulus (Fig. 3E), EECM/En]; this also is consistent with our experimental results showing that the NAR of TSA-treated nuclei within scaffolds was larger than nuclei in the control group.

The above data demonstrate that TSA treatment decreases chromatin condensation for a sufficient period of time to permit migration. However, prolonged exposure to this agent may have deleterious effects on cell phenotype and function. To assess this, we queried how long changes in MFC nuclear condensation persist after TSA withdrawal. MFCs were treated with TSA for 1 day as above, followed by five additional days of culture in fresh BM (Fig. 4A). Consistent with our previous findings, TSA decreased chromatin condensation and CCP after 1 day of treatment (Fig. 4, B and C). Upon removal of TSA, CCP values progressively increased, reaching baseline levels by day 5 (Fig. 4, B and C). A similar finding was noted in H2B localizations and domain area via STORM imaging, where these values returned to baseline levels within 5 days of TSA withdrawal (fig. S8, A to C). Similarly, nuclei in MFCs treated with TSA showed increased deformation compared to control MFC nuclei that were not treated with TSA (Fig. 4D) and increased Ac-H3K9 levels (Fig. 4, E and F), but these values gradually returned to the baseline levels within 5 days with TSA removal (Fig. 4, D to F). Over this same time course, proliferation was decreased in TSA-treated cells but returned to baseline levels within 5 days of TSA withdrawal on both tissue culture plastic (TCP) and on AL nanofibrous scaffolds (fig. S8, D and E). No change in levels of apoptosis (caspase activity) was observed over this time course (fig. 8F). Further, to investigate phenotypic behavior of cells after TSA treatment in the context of tissue repair, we next assayed whether cells exposed to TSA showed alterations in fibrochondrogenic gene expression and collagen production in MFCs. Although the sample size was small in this study, we did not detect a significant change in gene expression for any of the major collagen isoforms or proteoglycans normally produced by meniscus cells (fig. S9A). To further assess this, MFCs were treated with TSA for 1 day, followed by culture in fresh BM or TGF-3 containing chemically defined media (to accelerate collagen production) for an additional 3 days. Collagen produced by these cells and released to the media was not altered by TSA treatment (fig. S9B). Together, these data support that TSA treatment decreases chromatin condensation by increasing acetylation of histones in MFCs but this change is transient and baseline levels are restored gradually after TSA is removed, without alterations in collagen production.

(A) Schematic showing experimental setup; adult MFCs seeded on AL nanofibrous scaffolds were treated with/without TSA in BM for 1 day, followed by culture in fresh BM without TSA for an additional 5 days. (B) Representative DAPI-stained nuclei (top) and corresponding detection of visible edges (bottom) (scale bar, 3 m) and (C) CCP for time points indicated in (A) (red line; BM control at day 0, n = ~20 nuclei, *P < 0.05 versus Ctrl, means SEM). (D) NAR with 3 and 15% of applied stretch (normalized to NAR with 0%, n = 65 ~80 cells, *P < 0.05 versus 3%, +P < 0.05 versus Ctrl, P < 0.05 versus day 0, and aP < 0.05 versus day1, means SEM). (E) Immunostaining for Ac-H3K9 (green) in nuclei (blue) and quantification of mean intensity of the immunostaining (F) (n = ~28 cells, *P < 0.05 versus Ctrl and +P < 0.05 versus day 0, means SEM]. a.u., arbitrary units. All experiments were carried out in triplicate.

Given that transient TSA treatment softened MFC nuclei, resulting in enhanced interstitial cell migration, and did not perturb collagen production in the short term, we next investigated longer-term maturation of a tissue engineered construct with TSA treatment. For this, MFCs were seeded onto AL-PCL/PEO 25% scaffolds and cultured in TGF-3 containing chemically defined media for 4 weeks with/without TSA treatments (once a week for 1 day) as illustrated in Fig. 5A. In controls [()TSA], collagen deposition occurred mostly at the construct border (Fig. 5B), but both deposition and cell distribution were improved with TSA treatment [(+)TSA] (Fig. 5, B and C). Quantification showed that ~50% of cells were located within 50 m of the scaffold edge in controls [()TSA], while TSA treatment [(+)TSA] increased the number of cells deeper within the scaffold (250- to 400-m range; Fig. 5D).

(A) Experimental schematic showing MFCs seeded on PCL/25% PEO nanofibrous scaffolds that were cultured in chemically defined media for 4 weeks with TSA treatment once per week. After 4 weeks, ECM production and cell infiltration with/without TSA treatment were evaluated. Representative cross sections of MFC-laden nanofibrous constructs at week 4 stained for collagen (B) and cell nuclei (C). Scale bar, 100 m. (D) Quantification of MFC infiltration with/without TSA treatment (n = 3 images from three separate samples, *P < 0.05 versus ()TSA, means SEM). Experiments were carried out in duplicate. PSR, Picrosirius Red.

Toward meniscus repair, it is important to evaluate MFC migration through the dense fibrous ECM of meniscus tissue in the context of TSA treatment. For this, adult meniscus explants (, 5 mm) were cultured for ~2 weeks, donor cells in these vital explants were stained with CellTracker, and the explants were placed onto devitalized tissue substrates and cultured for an additional 48 hours, with/without TSA treatment [(/+)TSA] (Fig. 6A). During this 48-hour period, the cells derived from the donor explants adhered to the tissue substrates (Fig. 6B). In control groups [()TSA], cells were found predominantly on the substrate surface, whereas TSA-treated MFCs were found below the substrate surface (Fig. 6, B and C). Quantification showed that both the percent infiltration and the infiltration depth were significantly greater with TSA treatment (Fig. 6D).

(A) Schematic showing processing of vital tissue explants and devitalized tissue sections for invasion assay. Cell migration from the vital tissue and infiltration into the devitalized tissue section were evaluated by confocal microscopy. (B) 3D reconstructions (scale bar, 200 m) and (C) cross-sectional views (scale bar, 50 m) of cells (green) migrating through the devitalized tissue sections (blue), with and without TSA treatment. (D) Quantification of the percentage of infiltrated cells [n = 6 images, *P < 0.05 versus ()TSA, means SD] and cell infiltration depth [n = ~40 cells, *P < 0.05 versus ()TSA, means SEM]. Experiments were carried out in triplicate. (E) Electrospinning schematic showing two independent fiber jets collected simultaneously onto a common rotating mandrel. Discrete fiber populations are composed of PEO containing TSA and PCL. (F) Experimental schematic showing meniscus cell seeding onto nanofiber substrates. One day after seeding, the composite PCL/PEO TSA-releasing (PPT) scaffold was added to the microfluidic chamber reservoir, and cells were cultured for an additional 2 days, followed by confocal imaging. (G) 3D confocal reconstructions of cell (green) migration through AL nanofibrous networks with and without scaffold-mediated TSA delivery (scale bar, 100 m) and quantifications of the percentage of infiltrated cells [n = 5 images, *P < 0.05 versus ()TSA, +P < 0.05 versus (+)TSA, and #P < 0.05 versus 100 ng, means SD; biomolecule loading (mass per scaffold) is based on electrospinning parameters and scaffold mass]. (H) Schematic of repair construct assembly and subcutaneous evaluation in a rat model. (I) Images of DAPI-stained nuclei (blue) at the center of repair constructs after 1 week of subcutaneous implantation, with and without TSA delivery. Dashed lines indicate tissue-scaffold interfaces; dotted lines indicate separation into outer one-third (A), middle (B), and inner one-third (C) sections for quantification. Scale bar, 300 m. (J) Number of cells within each region of the scaffold with and without biomaterial-mediated TSA release (n = 3 samples from three different animals, *P < 0.05 versus PCL/PEO).

Next, we developed an assay to evaluate endogenous cell migration within native tissue. For this, tissue explants (, 6 mm) were excised from adult menisci, and the cells on the periphery of the explants were devitalized using a two-cycle freeze-thaw process (freezing in 20C for 30 min, followed by thawing at room temperature for 30 min, repeated twice on day 2; fig. S10A). This resulted in a ring of dead cells at the periphery of the tissue and a vital core. Processed explants were then treated with TSA for 1 day (day 1) and cultured in fresh media for an additional 3 days (fig. S10A). At the end of culture, living cells along the explant border were quantified. In controls that had not been treated by freeze-thaw (Ctrl), live cells occupied the periphery (fig. S10, B and D). With the two-cycle freeze-thaw process, there was a significant decrease in the number of live cells in this region (fig. S10, B and D), while cells in the center of the explant remained vital (day 2; fig. S10, B and D). With TSA treatment [(+)TSA], the number of vital cells that had migrated from the vital core to the periphery was significantly increased (day 3; fig. S10, C and D).

Last, to demonstrate the clinical potential of these findings, we developed an integrated biomaterial implant system to improve tissue repair in vivo (10, 35) via TSA delivery (Fig. 6E). Here, TSA was released from the PEO fiber fraction of a composite nanofibrous scaffold when this fiber fraction dissolves when placed in an aqueous environment. To first demonstrate bioactivity of the scaffold, we directly included small segments of these TSA-releasing composite scaffolds in the top chamber of the microfluidic migration device to treat seeded MFCs (Fig. 6F). Consistent with findings from soluble delivery, the percentage of infiltrated cells increased with the addition of the TSA-releasing composite scaffold (Fig. 6G): scaffolds releasing ~200 ng of TSA resulted in similar cell migration as direct addition of TSA (200 ng/ml) to the chamber (Fig. 6G). These results show our ability to deliver TSA to the wound site in a controlled fashion. To determine whether these TSA-releasing scaffolds could improve interstitial migration of endogenous meniscus cells in an in vivo setting, we subcutaneously placed meniscal repair constructs in nude rats with empty (PCL/PEO) or TSA-releasing scaffolds (PCL/PEO/TSA) interposed between the cut surfaces and histologically evaluated cellularity of the tissue and implant at 1 week (Fig. 6H). Results showed that interfacial cellularity was markedly higher for repair constructs with the scaffolds releasing ~100 ng of TSA (PCL/PEO/TSA) compared to control scaffolds (PCL/PEO; Fig. 6I), with cells occupying the full thickness of the TSA-releasing scaffold (Fig. 6J). Together, these data indicate that biomaterial-mediated nuclear softening of endogenous meniscus cells increases their capacity for interstitial migration through the tissue and into the scaffold in an in vivo setting.

PCL nanofibrous scaffolds were fabricated via electrospinning as in (6). Briefly, a PCL solution (80 kDa; Shenzhen Bright China Industrial Co. Ltd., China; 14.3% (w/v) in 1:1 tetrahydrofuran and N,N-dimethylformamide) was extruded through a stainless steel needle (2.5 ml/hour, 18-gauge, charged to +13 kV). To form NAL scaffolds, fibers were collected on a mandrel rotating with a surface velocity of <0.5 m/s. For AL scaffolds, fibers were collected at a high surface velocity (~10 m/s) (36). In some studies, to enhance cell infiltration, PCL/PEO (PEO, 200 kDa; Polysciences Inc., Warrington, PA) composite AL fibrous scaffolds were produced by coelectrospinning two fiber fractions onto the same mandrel, as in (6). For this, solutions of PCL (14.3%, w/v) and PEO (10%, w/v, in 90% ethanol) were electrospun simultaneously onto a centrally located mandrel (~10 m/s, 2.5 ml/hour). Resulting composite scaffolds were produced with PEO content of 0, 25, and 50% by scaffold dry mass. To visualize fibers, CellTracker Red (0.0005%, w/v) was mixed into the PCL solutions before electrospinning. Scaffolds were hydrated and sterilized in ethanol (100, 70, 50, and 30%; 30 min per step) and incubated in a fibronectin (20 g/ml) solution overnight to enhance initial cell attachment. TSA-releasing scaffolds contained a semipermanent (very slow degrading) fiber population (PCL) and a transient (water soluble) fiber population (PEO). The PEO fibers released TSA as they dissolve. To form this fiber fraction, TSA was added to the PEO solution (1% wt/vol) 2 days before spinning. PCL (10 ml) and PEO/TSA (10 ml) solutions were loaded into individual syringes and electrospun simultaneously by coelectrospinning onto a common centrally located mandrel, as above. Estimates of TSA content (mass per scaffold) were based on electrospinning parameters and the mass of each fiber fraction (Fig. 6E).

MFCs were isolated from the outer zone of adult bovine (20 to 30 months; Animal Technologies Inc.) or porcine menisci (6 to 9 months; Yucatan, Sinclair BioResources). For this, meniscal tissue segments were minced into ~1-mm3 cubes and placed onto TCP and incubated at 37C in a BM consisting of Dulbeccos modified Eagles medium (DMEM) with 10% fetal bovine serum and 1% penicillin/streptomycin/fungizone (PSF). Cells gradually emerged from the small tissue segments over 2 weeks, after which the remaining tissue was removed and the cells were passaged one time before use. MSCs were isolated from juvenile bovine bone marrow as in (37) and expanded in BM. To induce MSC fibrochondrogenesis, passage 1 MSCs were seeded on AL PCL scaffolds and cultured in a chemically defined serum free medium consisting of high glucose DMEM with 1 PSF, 0.1 M dexamethasone, ascorbate 2-phosphate (50 g/ml), l-proline (40 g/ml), sodium pyruvate (100 g/ml), insulin (6.25 g/ml), transferrin (6.25 g/ml), selenous acid (6.25 ng/ml), bovine serum albumin (BSA; 1.25 mg/ml), and linoleic acid (5.35 g/ml) (Life Technologies, NY, USA). This base medium (Ctrl) was further supplemented with TGF-3 (10 ng/ml) to induce differentiation (Ctrl/Diff, R&D Systems, Minneapolis, MN). Cell-seeded constructs were cultured in this medium for up to 7 days.

MFCs or MSCs were plated into eight-well Lab-Tek 1 cover glass chambers (Nunc), followed by preculture in BM for 2 days. At this time point, cells were treated with TSA for 3 hours, followed by fixation in methanol-ethanol (1:1) at 20C for 6 min. After a 1-hour incubation in blocking buffer containing 10 weight % BSA (Sigma-Aldrich) in phosphate-buffered saline (PBS), samples were incubated overnight with anti-H2B (1:50; abcam1790, Abcam), anti-H3K4me4 (1:100; MA5-11199, Thermo Fisher Scientific), or anti-H3K27me3 (1:100; PA5-31817, Thermo Fisher Scientific) at 4C. Next, samples were washed and incubated for 40 min with secondary antibodies custom labeled with activator-reporter dye pairs (Alexa Fluor 405Alexa Fluor 647, Invitrogen) for STORM imaging (29, 38). All imaging experiments were carried out with a commercial STORM microscope system from Nikon Instruments (N-STORM). For imaging, the 647-nm laser was used to excite the reporter dye (Alexa Fluor 647, Invitrogen) to switch it to the dark state. Next, a 405-nm laser was used to reactivate the Alexa Fluor 647 in an activator dye (Alexa Fluor 405)facilitated manner. An imaging cycle was used in which one frame belonging to the activating light pulse (405 nm) was alternated with three frames belonging to the imaging light pulse (647 nm). Imaging was carried out in a previously described imaging buffer [Cysteamine (#30070-50G, Sigma-Aldrich), GLOX solution: 1 glucose oxidase (0.5 mg/ml), 1 catalase (40 mg/ml) (all from Sigma-Aldrich), and 10% glucose in PBS] (39). STORM images were analyzed and rendered using custom-written software (Insight3, gift of B. Huang, University of California, San Francisco, USA) as previously described (39). For quantitative analysis, a previously described method was adapted that segments super-resolution images based on Voronoi tessellation of the fluorophore localizations (27, 28). Voronoi tessellation of a STORM image assigns a Voronoi polygon to each localization, such that the polygon area is inversely proportional to the local localization density (40). The spatial distribution of localizations is represented by a set of Voronoi polygons such that smaller polygon areas correspond to regions of higher density. Domains were segmented by grouping adjacent Voronoi polygons with areas less than a selected threshold, and imposing a minimum of three localizations per domain criteria generates the final segmented dataset.

MFCs (P1) were seeded onto AL PCL (0% PEO) scaffolds in BM for 2 days. To induce chromatin decondensation, TSA, a HDAC inhibitor (26) was added to the media for 3 hours. Chromatin condensation state and nuclear deformability were evaluated 3 hours after TSA treatment. For chromatin condensation analysis, constructs were fixed in 4% paraformaldehyde for 30 min at 37C, followed by PBS washing and permeabilization (with 0.05% Triton X-100 in PBS supplemented with 320 mM sucrose and 6 mM magnesium chloride). Nuclei were visualized by DAPI (ProLong Gold Antifade Reagent with DAPI, P36935, Molecular Probes, Grand Island, NY) and imaged at their mid-section using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL). Edge density in individual nuclei was measured using a Sobel edge detection algorithm in MATLAB to calculate the CCP as described in (24).

To assess nuclear deformability, the NAR (NAR = a/b) was evaluated before (0%) and after 9 and 15% grip-to-grip static deformation of constructs. Nuclear shape was captured on an inverted fluorescent microscope (Nikon T30, Nikon Instruments, Melville, NY) equipped with a charge-coupled device camera at each deformation level. NAR was calculated using a custom MATLAB code. Changes in NAR were tracked for individual MSC nuclei at each strain step as in (41).

To assess MFC migration on 2D substrates, a scratch assay was performed with or without TSA treatment. For this, passage 1 MFCs were plated into a six-well tissue culture dish (2 105 cells per well) and cultured to confluence (for 2 to 3 days). Confluent monolayers were then scratched with a 2.5-l pipette tip, and cell debris was removed via PBS washing. Images were taken using an inverted microscope at regular intervals and wound closure computed using ImageJ.

In addition, as an initial assessment of MFC migration, 96-well transwell migration assay kits (Chemicon QCM 96-well Migration Assay; membrane pore size, 3, 5, or 8 m) were used to assess cell migration. Briefly, human recombinant PDGF-AB (100 ng/ml in 150 l of BM; Prospect Bio) was added to the bottom chamber, and passage 1 MFCs (50,000 cells per well) were seeded into the top chamber. Cells were allowed to migrate for 18 hours at 37C with/without TSA treatment. In some studies, different dosages of TSA (0 to 800 nM) were applied (at a pore size of 5 m).

To assess initial cell migration through dense nanofiber networks, a custompoly(dimethylsiloxane) (PDMS) migration assay chamber was implemented (Fig. 2A). Top and bottom pieces containing holes (top, 6, 7, 6 mm in diameter; bottom, 6, 5, 6 mm in diameter) and a channel (bottom, 2 mm in width and 20 mm in length) were designed via SOLIDWORKS software for 3D printed templates (Acura SL 5530, Protolabs), and these were cast from the templates with PDMS (Sylgard 184, Dow Corning). To assemble the multilayered chamber, bottom PDMS pieces, the periphery of PCL electrospun fiber networks, and top PDMS pieces were coated with uncured PDMS base and curing agent mixture (10:1 ratio) and placed on cover glasses sequentially. For firm adhesion of each layer, chambers were incubated at 40C overnight. The final device consisted of a top reservoir containing BM and a bottom reservoir containing BM + PDGF (100 ng/ml) as a chemoattractant (Fig. 2A). To simulate chemoattactant diffusion from bottom to top reservoirs, trypan blue 0.4% solution (MP Biomedicals) was introduced to one of the side holes to fill the bottom reservoir, and the central top reservoir was filled with PBS. Cell migration chambers were kept in incubator (37C, 5% CO2), and images were obtained at regular intervals (Fig. 2D).

Fluorescently labeled (CellTracker Red) AL or NAL nanofibrous PCL scaffolds (thickness, ~150 m) were interposed between the reservoirs, and MFCs (2000 cells, passage 1) were seeded onto the top of each scaffold, followed by 1 day before culture in BM. Cells in chambers were cultured in BM with/without TSA for an additional 2 days. At the end of 3 days, cells were fixed and visualized by actin/DAPI staining. Confocal z-stacks were obtained at 40 magnification, and maximum z-stack projections were used to assess cellular morphology (cell/nuclear aspect ratio, area, circularity, and solidity). The percentage of infiltrated cells was quantified from confocal z stacks, with cells located beneath fibers categorized as infiltrated (fig. S3C) and the infiltration depth measured on cross-sectional images using ImageJ. For scanning electron microscopy imaging, additional samples were fixed and dehydrated in ethanol (30, 50, 70, and 100%, 60 min per step) and then hexamethyldisilane for terminal dehydration under vacuum.

Details on the model have been described previously (34). Briefly, to understand the influence of both intracellular and extracellular cues on cell migration through the fibrous ECM, we considered a model in which a cell with a spherical nucleus of radius rn is invading ECM through a deformable gap (with radius rg) smaller than the diameter of the nucleus (Fig. 3A). For simplicity, the nucleus is modeled by a spheroid and treated as a compressible neo-Hookean hyperelastic material to capture the mechanical response. An infinitely long small channel is created in the ECM to mimic the path a cell would migrate through in the migration assay. A neo-Hookean hyperelastic material was used to capture the ECM mechanical properties. The model parameters are shown in Table 1.

To assess how fast the TSA-mediated MFC chromatin organization and deformability was restored after TSA removal, MFCs seeded on AL scaffolds were treated with TSA for 1 day, followed by additional culture for 5 days in fresh BM (Fig. 4A). At each time point, the CCP and nuclear deformability were evaluated as described above. In addition, Ac-H3 levels in MFC nuclei were assessed by immunostaining with an Ac-H3K9 monoclonal antibody (MA5-11195, Thermo Fisher Scientific; 1:400, overnight at 4C). All images were collected using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL) at 63 magnification, with staining intensity quantified using ImageJ.

For long-term evaluation of matrix production after TSA treatment, MFCs were seeded on PCL/PEO 25% AL nanofibrous scaffolds (P1, 105 cells, 1 cm by 1 cm by 0.1 cm) and were cultured in TGF-3 containing chondrogenic media for 4 weeks. TSA was applied once each week for 24 hours. After 4 weeks, constructs were fixed with 4% paraformaldehyde and embedded in CryoPrep frozen section embedding medium [optimal cutting temperature (OCT) compound, Thermo Fisher Scientific, Pittsburgh, PA]. Using a cryostat microtome (Microm HM-500 M Cryostat, Ramsey, MN), constructs were sectioned to 8 m in thickness through their depth and stained with Picrosirius Red and DAPI to visualize collagen and nuclei, respectively. Stained sections were visualized and imaged by brightfield and fluorescent microscopy (Nikon Eclipse TS 100, Melville, NY). To quantify cell infiltration in the scaffolds, the number of migrated cells as a function of scaffold depth was determined for each experimental group (n = 3 scaffolds per group) using ImageJ.

To isolate fresh MFCs, cylindrical tissue explants (6 mm in diameter and 3 mm in height) were excised using biopsy punches from the middle zone of the meniscus, and these explants incubated in BM for ~2 weeks to allow cells to occupy the periphery. To fabricate devitalized tissue substrates, additional cylindrical tissue explants (8 mm in diameter) were embedded in OCT sectioning medium (Sakura Finetek, Torrance, CA) and axially cut (to ~50 m in thickness) using a cryostat microtome. These devitalized sections were placed onto positively charged glass slides and stored at 20C until use. After ~2 weeks of in vitro culture, the living explants were incubated in 5-chloromethylfluorescein diacetate (5 g/ml) (CellTracker Green, Thermo Fisher Scientific, Waltham, MA) in serum-free media (DMEM with 1% PSF) for 1 hour to fluorescently label cells in the explants. The explants were placed atop tissue substrates to allow for cell egress onto and invasion into the sections, and slides with explants were incubated at 37C with/without TSA treatment in BM for 2 days, at which point maximum z-stack projections were acquired using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL). Cell infiltration depth was measured as the distance between the apical tissue surface and the basal cell surface using a custom MATLAB code (3), and the total number of cells and the number of migrated cells (those entirely embedded within the tissue) were counted (n = 3 per group) using ImageJ.

In addition, to observe endogenous meniscus cell migration in the native ECM, a tissue-based migration assay was developed. Cylindrical meniscus tissue explants (6 mm in diameter and ~6 mm in height) were excised from the middle zone of adult menisci. To kill the cells on the border of the tissue, explants were frozen at 20C for 30 min and then thawed at room temperature for 30 min; this process was repeated twice (two-cycle) (day 2; fig. S10A). After devitalizing the periphery, explants were cultured in BM for 1 day, and TSA was added for 1 day (day 1; fig. S10A). After TSA treatment, explants were washed with PBS (day 0; fig. S10A), followed by culture in fresh BM for an additional 3 days. At day 3, LIVE/DEAD staining was performed, and explants cross sections were imaged (day 3; fig. S10A). Images were acquired from eight regions distributed evenly around the boundary (Leica TCS SP8, Leica Microsystems Inc., IL). The number of live cells located within 1 mm of the boundary was determined using ImageJ.

To evaluate the impact of biomaterial-mediated TSA delivery on endogenous meniscus cell migration in an in vivo setting, a nude rat xenotransplant model was used, as in (10). All animal procedures were approved by the Animal Care and Use Committee of the Corporal Michael Crescenz VA Medical Center. Before subcutaneous implantation, horizontal defects were created in adult bovine meniscal explants (8 mm in diameter and 4 mm in height, n = 3 donors; Fig. 6H). Electrospun PCL/PEO scaffolds with/without TSA were prepared (6 mm in diameter with a 2-mm-diameter central fenestration). Control PCL/PEO scaffolds or scaffolds releasing TSA (PCL/PEO/TSA, ~100 ng) were placed into the defect, which was closed with absorbable sutures. The repair construct was implanted subcutaneously into the dorsum of male athymic nude rats (n = 3, Hsd:RH-Foxn1rnu, 8 to 10 weeks old, ~300 g, Harlan) (Fig. 6H) (10). At 1 week, rats were euthanized, and constructs were removed from the subcutaneous space. Samples were fixed with para-formaldehyde and embedded in OCT sectioning medium (Sakura Finetek, Torrance, CA), sectioned to 8 m in thickness, stained with DAPI for cell nuclei, and imaged using a fluorescence microscope. Cell number in the center and edges of the implanted scaffold were determined using ImageJ.

Statistical analysis was performed using Student t tests or analysis of variance (ANOVA) with Tukeys honestly significantly different post hoc tests (SYSTAT v.10.2, Point Richmond, CA). For datasets that were not normally distributed, nonparametric Mann-Whitney or Kruskal-Wallis tests were performed, followed by post hoc testing with Dunns correction using GraphPad Prism version 6 (GraphPad Software Inc., La Jolla, CA, USA). Results are expressed as the means SEM or SD, as indicated in the figure legends. Differences were considered statistically significant at P < 0.05.

Acknowledgments: We acknowledge S. Gullbrand, D. H. Kim, and E. Henning for technical support. Funding: This work was supported by the NIH (R01 AR056624), the Department of Veterans Affairs (I01 RX000174), the NSF Science and Technology Center for Engineering Mechanobiology (CMMI-1548571), and the Penn Center for Musculoskeletal Disorders (P30 AR069619). Author contributions: S.-J.H., K.H.S., S.T., X.C., A.P.P., B.N.S., F.Q., V.B.S., M.L., J.A.B., and R.L.M. designed the studies. S.-J.H., K.H.S., S.T., X.C., A.P.P., and B.N.S. performed the experiments. S.-J.H., K.H.S., S.T., X.C., A.P.P., B.N.S., F.Q., V.B.S., M.L., J.A.B., and R.L.M. analyzed and interpreted the data. S.-J.H., S.T., X.C., V.B.S., M.L., J.A.B., and R.L.M. drafted the manuscript, and all authors edited the final submission. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Read the rest here:
Nuclear softening expedites interstitial cell migration in fibrous networks and dense connective tissues - Science Advances

Global Stem Cell Market Study and Forecast 2020-2025: Oncology Disorders Expected to Exhibit the Fastest Growth Rate – ResearchAndMarkets.com -…

DUBLIN--(BUSINESS WIRE)--The "Global Stem Cell Market: Growth, Trends and Forecasts (2020-2025)" report has been added to ResearchAndMarkets.com's offering.

The global stem cell market is experiencing growth, owing to the increasing number of clinical trials around the world.

North America, especially the United States, dominated the number of trials undergoing stem cell therapies. However, Asia-Pacific is growing at the highest growth rate. Stem cells are majorly used in regenerative medicine, especially in the field of dermatology. However, oncology is expected to grow at the highest growth rate, due to a large number of pipeline products present for the treatment of tumors or cancers. With the increase in the number of regenerative medicine centers, the stem cell market is also expected to increase in the future.

Stem cell banking is gaining importance with the support of government initiatives. The number of stem cell banks is increasing in developing countries, which is aiding the growth of the market. Also, increasing awareness about stem cell storage among the people has positively affected the market. Currently, the market is not well established in many therapeutic areas and has shown nascent success in history. However, it holds great potential in both the diagnosis and therapeutic fields.

Oncology Disorders Segment Expected to Exhibit the Fastest Growth Rate Over the Forecast Period

Cancer has a major impact on the world. According to the World Health Organization (WHO) 2018 data on cancer, the global cancer burden is estimated to have risen to 18.1 million new cases and 9.6 million deaths in 2018. Moreover, Cancer Research UK suggests that the population suffering from cancer is expected to increase in the future. As per the report, if recent trends in the incidence of major cancers and population growth are consistent, it is predicted there will be 27.5 million new cancer cases worldwide each year by 2040.

Stem cell transplants are procedures that restore blood-forming stem cells in people who have had theirs destroyed by the very high doses of chemotherapy or radiation therapy. Embryonic stem cells (ESC) are the major source of stem cells for therapeutic purposes, due to their higher totipotency and indefinite lifespan, as compared to adult stem cells with lower totipotency and restricted lifespan. These advantages along with the increasing incidence of cancer is expected to help the growth of stem cell market

North America Captured the Largest Market Share and is Expected to Retain its Dominance

North America dominated the overall stem cell market with the United States contributing to the largest share in the market. The United States (US) and Canada have a developed and well-structured health care system. These systems also encourage research and development. These policies encourage global players to enter the US and Canada. As a result, these countries enjoy the presence of many global market players. Additionally, Mexico is a developing nation with the benefit of being a neighbor to the United States. This allows many companies to penetrate in Mexico as well. This helps the growth in the region.

Competitive Landscape

The stem cell market is highly competitive and consists of several major players. In terms of market share, few of the major players currently dominate the market. The presence of major market players, such as Thermo Fisher Scientific (Qiagen NV), Sigma Aldrich (A Subsidiary of Merck KGaA), Becton, Dickinson and Company, and Stem Cell Technologies, is in turn, increasing the overall competitive rivalry in the market. The product advancements and improvement in stem cell technology by the major players are increasing the competitive rivalry.

Key Topics Covered

1 INTRODUCTION

1.1 Study Deliverables

1.2 Study Assumptions

1.3 Scope of the Study

2 RESEARCH METHODOLOGY

3 EXECUTIVE SUMMARY

4 MARKET DYNAMICS

4.1 Market Overview

4.2 Market Drivers

4.2.1 Increased Awareness about Umbilical Stem Cell

4.2.2 Rising R&D Initiatives to Develop Stem Cell Therapies and Increasing Approvals for Clinical Trials in Stem Cell Research

4.2.3 Growing Demand for Regenerative Treatment Option

4.3 Market Restraints

4.3.1 Expensive Procedures

4.3.2 Regulatory Complications

4.3.3 Ethical and Moral Framework

4.4 Industry Attractiveness- Porter's Five Forces Analysis

5 MARKET SEGMENTATION

5.1 By Product Type

5.1.1 Adult Stem Cell

5.1.2 Human Embryonic Cell

5.1.3 Pluripotent Stem Cell

5.1.4 Other Product Types

5.2 By Application

5.2.1 Neurological Disorders

5.2.2 Orthopedic Treatments

5.2.3 Oncology Disorders

5.2.4 Injuries and Wounds

5.2.5 Cardiovascular Disorders

5.2.6 Other Applications

5.3 By Treatment Type

5.3.1 Allogeneic Stem Cell Therapy

5.3.2 Auto logic Stem Cell Therapy

5.3.3 Syngeneic Stem Cell Therapy

5.4 Geography

5.4.1 North America

5.4.2 Europe

5.4.3 Asia-Pacific

5.4.4 Middle-East & Africa

5.4.5 South America

6 COMPETITIVE LANDSCAPE

6.1 Company Profiles

6.1.1 Osiris Therapeutics Inc.

6.1.2 Pluristem Therapeutics Inc.

6.1.3 Thermo Fisher Scientific

6.1.4 Merck KGaA (Sigma Aldrich)

6.1.5 Becton, Dickinson and Company

6.1.6 Stem Cell Technologies Inc.

6.1.7 AllCells LLC

6.1.8 Miltenyi Biotec

6.1.9 International Stem Cell Corporation

6.1.10 Smith & Nephew PLC

7 MARKET OPPORTUNITIES AND FUTURE TRENDS

For more information about this report visit https://www.researchandmarkets.com/r/z5sdky

Go here to read the rest:
Global Stem Cell Market Study and Forecast 2020-2025: Oncology Disorders Expected to Exhibit the Fastest Growth Rate - ResearchAndMarkets.com -...

EHA25Virtual: Adult Patients With Sickle Cell Disease May Be at Increased Risk of Adverse Outcomes From COVID-19 – Yahoo Finance

THE HAGUE, Netherlands, June 13, 2020 /PRNewswire/ -- Sickle cell disease (SCD) and thalassemia are severe inherited blood disorders, often referred to as "hemoglobinopathies." They predominantly affect the Black and Asian ethnic minority populations in England. To ensure good standards and equitable access to care, the National Health Service in England has recently commissioned a model of regional care networks overseen by a new body, the National Haemoglobinopathy Panel. This organizational structure has enabled a rapid response to the COVID-19 epidemic and enabled collection of national data on new cases and outcomes to determine if hemoglobinopathy patients are at risk of adverse COVID-19 outcomes.

EHA Logo (PRNewsfoto/European Hematology Association)

We present an analysis on data collected up to June 5th indicating that the majority of cases have been mild, and in particular children do not appear to be at increased risk. However, the data suggests that adults with SCD may be more vulnerable to adverse outcomes. Therefore, we recommend that isolation precautions should be lifted cautiously, and that new therapies and vaccination for COVID-19, when available, should be prioritized for this patient group.

Presenter: Dr Paul Telfer Affiliation:Queen Mary University of London, Barts Health NHS Trust, London, UK Abstract:#LB2606 REAL-TIME NATIONAL SURVEY OF COVID-19 IN HEMOGLOBINOPATHY AND RARE INHERITED ANEMIA PATIENTS

About the EHA Annual Congress: Every year in June, EHA organizes its Annual Congress in a major European city. This year due to the COVID19 pandemic, EHA transformed its physical meeting into a Virtual Congress. The Congress is aimed at health professionals working in or interested in the field of hematology. The scientific program topics range from stem cell physiology and development to leukemia; lymphoma; diagnosis and treatment; red blood cells; white blood cells and platelet disorders; hemophilia and myeloma; thrombosis and bleeding disorders; as well as transfusion and stem cell transplantation. Embargo: Please note that our embargo policy applies to all selected abstracts in the Press Briefings. For more information, see our EHA Media and Embargo policy here.

Website: ehaweb.org

Logo - https://media.zenfs.com/en/prnewswire.com/b19270e94cffa3b03349741d8b8900b6

View original content to download multimedia:http://www.prnewswire.com/news-releases/eha25virtual-adult-patients-with-sickle-cell-disease-may-be-at-increased-risk-of-adverse-outcomes-from-covid-19-301075093.html

SOURCE European Hematology Association (EHA)

More:
EHA25Virtual: Adult Patients With Sickle Cell Disease May Be at Increased Risk of Adverse Outcomes From COVID-19 - Yahoo Finance

Tolero Pharmaceuticals Presents Findings from Phase 1 Zella 101 Clinical Study Evaluating Investigational Agent Alvocidib in Patients with Newly…

SALT LAKE CITY, Utah, June 12, 2020 /PRNewswire/ --Tolero Pharmaceuticals, Inc., a clinical-stage company focused on developing novel therapeutics for hematological and oncological diseases, today presented data from the completed Phase 1 Zella 101 study evaluating the investigational agent alvocidib, a potent CDK9 inhibitor, in adult patients with newly diagnosed acute myeloid leukemia (AML). These results were presented in a poster presentation at the 25th European Hematology Association (EHA) Virtual Congress, being held June 11-14, 2020.

Updated findings from the Phase 1, dose-escalation, safety and biomarker study of alvocidib followed by cytarabine and daunorubicin (7+3) induction therapy showed encouraging clinical activity and a tolerable safety profile in adults with newly diagnosed AML. In the study, 71% (n=22 of 31) of evaluable patients achieved complete remission (CR), with an overall response rate (ORR) of 77% (n=24 of 31). Additionally, an exploratory cohort of the study found that 89% (n=8 of 9) of patients achieved measurable residual disease (MRD)-negativity.At a median of 9.2 months follow-up, overall survival was not reached, with 62% of patients alive at data cut-off.1

The maximum tolerated dose of alvocidib was determined to be 30 mg/m2 IV bolus followed by 60 mg/m2 IV over 4 hours and no dose-limiting toxicities (DLTs) were observed. The most frequently observed treatment-emergent, nonhematologic adverse events of Grade 3 or higher were diarrhea, tumor lysis syndrome and hypocalcemia, which all resolved with supportive care.1

"AML is an aggressive blood cancer which can progress rapidly and remains difficult to treat. We are pleased with the clinical responses, including overall survival, observed in newly diagnosed AML patients treated with alvocidib followed by standard induction therapy. In addition, the high level of MRD-negativity, a meaningful indicator of durable response, is particularly encouraging," said David J. Bearss, Ph.D., Chief Executive Officer, Tolero Pharmaceuticals, and Chief Scientific Officer and Global Head of Research, Global Oncology. "We are excited to continue the advancement of this program and further investigate the potential role of alvocidib in contributing to a durable complete remission and achievement of MRD-negativity."

Below are the details for the presentation:

Abstract Title

Details

Author

Alvocidib Followed by 7+3 Induction in Newly Diagnosed AML Achieves High Rates of MRD-Negative CR: Results of a Phase 1 Dose Escalation Study

Poster# 551

June 12, 2020

8:30 a.m. CEST

e-Poster Presentation

Joshua F. Zeidner, M.D., University of North Carolina

About Alvocidib

Alvocidib is an investigational small molecule inhibitor of cyclin-dependent kinase 9 (CDK9) currently being evaluated in the ongoing Phase 2 Zella 202 study in patients with acute myeloid leukemia (AML) who have either relapsed from or are refractory to venetoclax in combination with azacitidine or decitabine (NCT03969420).Alvocidib is also being evaluated in Zella 102, a Phase 1b/2 study in patients with myelodysplastic syndromes (MDS) in combination with azacitidine or decitabine (NCT03593915) and in a Phase 1 study in patients with relapsed or refractory AML in combination with venetoclax (NCT03441555).

About CDK9 Inhibition and MCL-1

MCL-1 is a member of the apoptosis-regulating BCL-2 family of proteins.2 In normal function, it is essential for early embryonic development and for the survival of multiple cell lineages, including lymphocytes and hematopoietic stem cells.3 MCL-1 inhibits apoptosis and sustains the survival of leukemic blasts, which may lead to relapse or resistance to treatment.2,4 The expression of MCL-1 in leukemic blasts is regulated by cyclin-dependent kinase 9 (CDK9).5,6 Because of the short half-life of MCL-1 (2-4 hours), the effects of targeting upstream pathways are expected to reduce MCL-1 levels rapidly.5 Inhibition of CDK9 has been shown to block MCL-1 transcription, resulting in the rapid downregulation of MCL-1 protein, thus restoring the normal apoptotic regulation.2

About Tolero Pharmaceuticals, Inc.

Tolero Pharmaceuticals is a clinical-stage biopharmaceutical company researching and developing treatments to improve and extend the lives of patients with hematological and oncological diseases. Tolero has a diverse pipeline that targets important biological drivers of blood disorders to treat leukemias, anemia, and solid tumors, as well as targets of drug resistance and transcriptional control.

Tolero Pharmaceuticals is based in the United States and is an indirect, wholly owned subsidiary of Sumitomo Dainippon Pharma Co., Ltd., a pharmaceutical company based in Japan. Tolero works closely with its parent company, Sumitomo Dainippon Pharma, and Boston Biomedical, Inc., also a wholly owned subsidiary, to advance a pipeline of innovative oncology treatments. The organizations apply their expertise and collaborate to achieve a common objective - expediting the discovery, development and commercialization of novel treatment options.

Additional information about the company and its product pipeline can be found at http://www.toleropharma.com.

Tolero Pharmaceuticals Forward-Looking Statements

This press release contains "forward-looking statements," as that term is defined in the Private Securities Litigation Reform Act of 1995 regarding the research, development and commercialization of pharmaceutical products. The forward-looking statements in this press release are based on management's assumptions and beliefs in light of information presently available, and involve both known and unknown risks and uncertainties, which could cause actual outcomes to differ materially from current expectations. Any forward-looking statements set forth in this press release speak only as of the date of this press release. We do not undertake to update any of these forward-looking statements to reflect events or circumstances that occur after the date hereof. Information concerning pharmaceuticals (including compounds under development) contained within this material is not intended as advertising or medical advice.

References

1 Zeidner, Joshua et al. "Alvocidib Followed by 7+3 Induction in Newly Diagnosed AML Achieves High Rates of MRD-Negative CR: Results of a Phase 1 Dose Escalation Study." 25th European Hematology Association (EHA) Virtual Annual Congress. 12 June 2020. Poster presentation 551

2 Thomas D, Powell JA, Vergez F, et al. Targeting acute myeloid leukemia by dual inhibition of PI3K signaling and Cdk9-mediated Mcl-1 transcription. Blood. 2013;122(5):738-748.

3 Perciavalle RM, Opferman JT. Delving deeper: MCL-1's contributions to normal and cancer biology. Trends Cell Biol. 2013;23(1):22-29.

4 Glaser SP, Lee EF, Trounson E, et al. Anti-apoptotic Mcl-1 is essential for the development and sustained growth of acute myeloid leukemia. Genes Dev. 2012;26(2):120-125.

5 Chen R, Keating MJ, Gandhi V, Plunkett W. Transcription inhibition by flavopiridol: mechanism of chronic lymphocytic leukemia cell death. Blood. 2005;106(7):2513-2519.

6 Ocana A, Pandiella A. Targeting oncogenic vulnerabilities in triple negative breast cancer: biological bases and ongoing clinical studies. Oncotarget. 2017;8(13):22218-22234

See the original post:
Tolero Pharmaceuticals Presents Findings from Phase 1 Zella 101 Clinical Study Evaluating Investigational Agent Alvocidib in Patients with Newly...

Autolus Therapeutics presents AUTO1 and AUTO3 data at the 2020 EHA25 Virtual Congress – BioSpace

Pivotal AUTO1 study in adult ALL patients enrolling

Conference Call and Webcast to be held Friday, June 12, 2020 at 7:30 am EDT / 12:30 pm BST

LONDON, June 12, 2020 (GLOBE NEWSWIRE) -- Autolus Therapeutics plc (Nasdaq: AUTL), a clinical-stage biopharmaceutical company developing next-generation programmed T cell therapies, today announced new data highlighting progress on its AUTO1 program, the companys CAR T cell therapy being investigated in the ongoing ALLCAR Phase 1 study of relapsed / refractory adult B-Acute Lymphocytic Leukemia (ALL), at the European Hematology Association EHA25 Virtual Congress beginning June 11.

AUTO1 in ALLAs of the data cut-off date of May 13, 2020, 19 patients had received AUTO1. AUTO1 was well tolerated, with no patients experiencing Grade 3 CRS. Three patients (16%) with high leukemia burden (>50% blasts) experienced Grade 3 neurotoxicity that resolved swiftly with the application of steroids. Of the 19 patients, 16 (84%) achieved MRD-negative CR. Two out of 16 patients received a transplant while in remission and CD19-negative relapse occurred in 3 (16%) patients. Durability of remissions is encouraging. Event Free Survival (EFS) and Overall Survival (OS) at 6 months are 62% and 72% respectively in all patients, and 76% and 92% respectively in the 13 patients treated with the closed (commercial) process. Median EFS and OS has not been reached, at a median follow up of 12.2 months (range up to 24.4 months).

I am very encouraged by the tolerable safety profile and high level of sustained CRs we have observed with AUTO1 in the ALLCAR19 study that was achieved without subsequent stem cell transplant, said Dr. Claire Roddie, Consultant Hematologist, UCL Cancer Institute and University College London Hospital.

Approximately 60% of adult ALL patients relapse or are refractory to first line therapy and there continues to exist a high unmet need, said Dr. Michael Bishop, MD, Professor of Medicine and Director of the Cellular Therapy Program at University of Chicago Medicine. AUTO1 is a novel CD19 CAR T candidate with a compelling activity and safety profile and has the potential to change standard of care as a curative therapy for r/r ALL.

The data update on AUTO1 presented at this years EHA meeting show an encouraging durability of response without subsequent stem cell transplant and confirm the positive safety profile, said Dr. Christian Itin, chairman and chief executive officer of Autolus. We have started enrolment of patients with r/r aALL in our pivotal Phase 1b/2 AUTO1-AL1 study.

AUTO3 in DLBCLDr. Wendy Osborne presented ALEXANDER Phase 1/2 clinical trial data for AUTO3. This data is consistent with our update on May 29, 2020, with a data cut-off date of April 27, 2020.

These data are very encouraging, in terms of safety and tolerability, with a high level of clinical activity, said Dr. Wendy Osborne, Consultant Hematologist, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust. We are looking forward to enrolling additional patients in the outpatient cohort.

Dr. Wendy Osborne of Newcastle upon Tyne Hospitals NHS Foundation Trust also discusses AUTO3 data during the American Society of Clinical Oncology (ASCO) Annual Meeting in this video courtesy of the Lymphoma Hub.

Investor call on Friday June 12, 2020Management will host a conference call and webcast at 7:30 am EDT/12:30 pm BST to discuss the EHA data. To listen to the webcast and view the accompanying slide presentation, please go to: https://www.autolus.com/investor-relations/news-and-events/events.

The call may also be accessed by dialing (866) 679-5407 for U.S. and Canada callers or (409) 217-8320 for international callers. Please reference conference ID 4838626. After the conference call, a replay will be available for one week. To access the replay, please dial (855) 859-2056 for U.S. and Canada callers or (404) 537-3406 for international callers. Please reference conference ID 4838626.

About Autolus Therapeutics plcAutolus is a clinical-stage biopharmaceutical company developing next-generation, programmed T cell therapies for the treatment of cancer. Using a broad suite of proprietary and modular T cell programming technologies, the company is engineering precisely targeted, controlled and highly active T cell therapies that are designed to better recognize cancer cells, break down their defense mechanisms and eliminate these cells. Autolus has a pipeline of product candidates in development for the treatment of hematological malignancies and solid tumors. For more information please visit http://www.autolus.com.

About AUTO1 AUTO1 is a CD19 CAR T cell investigational therapy designed to overcome the limitations in safety - while maintaining similar levels of efficacy - compared to current CD19 CAR T cell therapies. Designed to have a fast target binding off-rate to minimize excessive activation of the programmed T cells, AUTO1 may reduce toxicity and be less prone to T cell exhaustion, which could enhance persistence and improve the ability of the programmed T cells to engage in serial killing of target cancer cells. AUTO1 is currently being evaluated in two Phase 1 studies, one in pediatric ALL and one in adult ALL. The company has also now progressed the program to a potential pivotal study, AUTO1-AL1.

About AUTO1-AL1 pivotal studyThe AUTO1-AL1 study will enroll patients with relapsed / refractory ALL. The study will have a short Phase1b component prior to proceeding to a single arm Phase 2 study. The primary end point is overall response rate and the key secondary end points include duration of response MRD negative CR rate and safety. The study will enroll approximately 100 patients across 30 of the leading academic and non-academic centers in the US, UK and Europe.

About AUTO3AUTO3 is a programmed T cell therapy containing two independent chimeric antigen receptors targeting CD19 and CD22 that have each been independently optimized for single target activity. By simultaneously targeting two B cell antigens, AUTO3 is designed to minimize relapse due to single antigen loss in patients with B cell malignancies. AUTO3 is currently being tested in diffuse large B cell lymphoma in the ALEXANDER clinical trial, with a 20-patient cohort that was initiated in Q2 2020 to assess feasibility of treatment in an outpatient setting.

Forward-Looking StatementsThis press release contains forward-looking statements within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform Act of 1995. Forward-looking statements are statements that are not historical facts, and in some cases can be identified by terms such as "may," "will," "could," "expects," "plans," "anticipates," and "believes." These statements include, but are not limited to, statements regarding Autolus financial condition and results of operations, including its expected cash runway; the development of Autolus product candidates, including statements regarding the timing of initiation, completion and the outcome of pre-clinical studies or clinical trials and related preparatory work, and the periods during which the results of the studies and trials will become available; Autolus plans to research, develop, manufacture and commercialize its product candidates; the potential for Autolus product candidates to be alternatives in the therapeutic areas investigated; and Autolus manufacturing capabilities and strategy. Any forward-looking statements are based on management's current views and assumptions and involve risks and uncertainties that could cause actual results, performance or events to differ materially from those expressed or implied in such statements. For a discussion of other risks and uncertainties, and other important factors, any of which could cause our actual results to differ from those contained in the forward-looking statements, see the section titled "Risk Factors" in Autolus' Annual Report on Form 20-F filed with the Securities and Exchange Commission on March 3, 2020 as well as discussions of potential risks, uncertainties, and other important factors in Autolus' future filings with the Securities and Exchange Commission from time to time. All information in this press release is as of the date of the release, and the company undertakes no obligation to publicly update any forward-looking statement, whether as a result of new information, future events, or otherwise, except as required by law.

Contact:

Lucinda Crabtree, PhDVice President, Investor Relations and Corporate Communications+44 (0) 7587 372 619l.crabtree@autolus.com

Julia Wilson+44 (0) 7818 430877j.wilson@autolus.com

Susan A. NoonanS.A. Noonan Communications+1-212-966-3650susan@sanoonan.com

See the rest here:
Autolus Therapeutics presents AUTO1 and AUTO3 data at the 2020 EHA25 Virtual Congress - BioSpace

Genmab Announces European Marketing Authorization for the Subcutaneous Formulation of DARZALEX (daratumumab) for the Treatment of Patients with Multi…

Copenhagen, Denmark; June 4, 2020 Genmab A/S (Nasdaq: GMAB) announced today that the European Commission (EC) has granted marketing authorization for the subcutaneous (SC) formulation of DARZALEX (daratumumab), for the treatment of adult patients with multiple myeloma in all currently approved daratumumab intravenous (IV) formulation indications in frontline and relapsed / refractory settings. The approval follows a Positive Opinion by the CHMP of the European Medicines Agency (EMA) in April 2020. The SC formulation is administered as a fixed-dose over approximately three to five minutes, significantly less time than IV daratumumab, which is given over several hours. Patients currently on daratumumab IV will have the choice to switch to the SC formulation. In August 2012, Genmab granted Janssen Biotech, Inc. (Janssen) an exclusive worldwide license to develop, manufacture and commercialize daratumumab.

We are extremely pleased that patients in Europe with multiple myeloma will now, like patients in the U.S., have the opportunity for treatment with the subcutaneous formulation of daratumumab. With consistent efficacy, and greater convenience for patients and health care providers with dosing time reduced from hours to just minutes and fewer infusion-related reactions, this formulation provides significant benefits for patients, said Jan van de Winkel, Ph.D., Chief Executive Officer of Genmab

The approval was based on data from two studies: the Phase III non-inferiority COLUMBA (MMY3012) study, which compared the SC formulation of daratumumab to the IV formulation in patients with relapsed or refractory multiple myeloma, and data from the Phase II PLEIADES (MMY2040) study, which is evaluating SC daratumumab in combination with certain standard multiple myeloma regimens. The topline results from the COLUMBA study were announced in February 2019 and subsequently presented in oral sessions at the 2019 American Society of Clinical Oncology (ASCO) Annual Meeting and the 24th European Hematology Association (EHA) Annual Congress. Updated data of the COLUMBA and the PLEIADES studies were presented during poster sessions at the 61st American Society of Hematology (ASH) Annual Meeting in December 2019.

About the COLUMBA (MMY3012) studyThe Phase III trial (NCT03277105) is a randomized, open-label, parallel assignment study that included 522 adults diagnosed with relapsed and refractory multiple myeloma. Patients were randomized to receive either: SC daratumumab, as 1800 mg daratumumab with rHuPH20 2000 U/mL once weekly in Cycle 1 and 2, every two weeks in Cycles 3 to 6, every 4 weeks in Cycle 7 and thereafter until disease progression, unacceptable toxicity or the end of study; or 16 mg/kg IV daratumumab once weekly in Cycle 1 and 2, every two weeks in Cycles 3 to 6, every 4 weeks in Cycle 7 and thereafter until disease progression, unacceptable toxicity or the end of study. The co-primary endpoints of the study are overall response rate and Maximum trough concentration of daratumumab (Ctrough; defined as the serum pre-dose concentration of daratumumab on Cycle 3 Day 1).

About the PLEIADES (MMY2040) studyThe Phase II trial (NCT03412565) is a non-randomized, open-label, parallel assignment study that includes 265 adults either newly diagnosed or with relapsed or refractory multiple myeloma. Patients with newly diagnosed multiple myeloma are being treated with 1,800 mg SC daratumumab in combination with either bortezomib, lenalidomide and dexamethasone (D-VRd) or bortezomib, melphalan and prednisone (D-VMP). Patients with relapsed or refractory multiple myeloma are being treated with 1,800 mg SC daratumumab plus lenalidomide and dexamethasone (D-Rd). An additional cohort of patients with relapsed and refractory multiple myeloma treated with daratumumab plus carfilzomib and dexamethasone (D-Kd) was subsequently added to the study. The primary endpoint for the D-VMP, D-Kd and D-Rd cohorts is overall response rate. The primary endpoint for the D-VRd cohort is very good partial response or better rate.

About DARZALEX (daratumumab)DARZALEX (daratumumab) intravenous infusion is indicated for the treatment of adult patients in the United States: in combination with bortezomib, thalidomide and dexamethasone as treatment for patients newly diagnosed with multiple myeloma who are eligible for autologous stem cell transplant; in combination with lenalidomide and dexamethasone for the treatment of patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; in combination with bortezomib, melphalan and prednisone for the treatment of patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone, for the treatment of patients with multiple myeloma who have received at least one prior therapy; in combination with pomalidomide and dexamethasone for the treatment of patients with multiple myeloma who have received at least two prior therapies, including lenalidomide and a proteasome inhibitor (PI); and as a monotherapy for the treatment of patients with multiple myeloma who have received at least three prior lines of therapy, including a PI and an immunomodulatory agent, or who are double-refractory to a PI and an immunomodulatory agent.1 DARZALEX is the first monoclonal antibody (mAb) to receive U.S. Food and Drug Administration (U.S. FDA) approval to treat multiple myeloma.

DARZALEX is indicated for the treatment of adult patients in Europe via intravenous infusion or subcutaneous administration: in combination with bortezomib, thalidomide and dexamethasone as treatment for patients newly diagnosed with multiple myeloma who are eligible for autologous stem cell transplant; in combination with lenalidomide and dexamethasone for the treatment of patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; in combination with bortezomib, melphalan and prednisone for the treatment of adult patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; for use in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone, for the treatment of adult patients with multiple myeloma who have received at least one prior therapy; and as monotherapy for the treatment of adult patients with relapsed and refractory multiple myeloma, whose prior therapy included a PI and an immunomodulatory agent and who have demonstrated disease progression on the last therapy2. Daratumumab is the first subcutaneous CD38-directed antibody approved in Europe for the treatment of multiple myeloma. The option to split the first infusion of DARZALEX over two consecutive days has been approved in both Europe and the U.S.

In Japan, DARZALEX intravenous infusion is approved for the treatment of adult patients: in combination with lenalidomide and dexamethasone for the treatment of patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; in combination with bortezomib, melphalan and prednisone for the treatment of patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant; in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone for the treatment of relapsed or refractory multiple myeloma. DARZALEX is the first human CD38 monoclonal antibody to reach the market in the United States, Europe and Japan. For more information, visit http://www.DARZALEX.com.

DARZALEX FASPRO (daratumumab and hyaluronidase-fihj), a subcutaneous formulation of daratumumab, is approved in the United States for the treatment of adult patients with multiple myeloma: in combination with bortezomib, melphalan and prednisone in newly diagnosed patients who are ineligible for ASCT; in combination with lenalidomide and dexamethasone in newly diagnosed patients who are ineligible for ASCT and in patients with relapsed or refractory multiple myeloma who have received at least one prior therapy; in combination with bortezomib and dexamethasone in patients who have received at least one prior therapy; and as monotherapy, in patients who have received at least three prior lines of therapy including a PI and an immunomodulatory agent or who are double-refractory to a PI and an immunomodulatory agent.3 DARZALEX FASPRO is the first subcutaneous CD38-directed antibody approved in the U.S. for the treatment of multiple myeloma.

Daratumumab is a human IgG1k monoclonal antibody (mAb) that binds with high affinity to the CD38 molecule, which is highly expressed on the surface of multiple myeloma cells. Daratumumab triggers a persons own immune system to attack the cancer cells, resulting in rapid tumor cell death through multiple immune-mediated mechanisms of action and through immunomodulatory effects, in addition to direct tumor cell death, via apoptosis (programmed cell death).1,4,5,6,7

Daratumumab is being developed by Janssen Biotech, Inc. under an exclusive worldwide license to develop, manufacture and commercialize daratumumab from Genmab. A comprehensive clinical development program for daratumumab is ongoing, including multiple Phase III studies in smoldering, relapsed and refractory and frontline multiple myeloma settings. Additional studies are ongoing or planned to assess the potential of daratumumab in other malignant and pre-malignant diseases in which CD38 is expressed, such as amyloidosis and T-cell acute lymphocytic leukemia (ALL). Daratumumab has received two Breakthrough Therapy Designations from the U.S. FDA for certain indications of multiple myeloma, including as a monotherapy for heavily pretreated multiple myeloma and in combination with certain other therapies for second-line treatment of multiple myeloma.

About Genmab Genmab is a publicly traded, international biotechnology company specializing in the creation and development of differentiated antibody therapeutics for the treatment of cancer. Founded in 1999, the company is the creator of three approved antibodies: DARZALEX (daratumumab, under agreement with Janssen Biotech, Inc.) for the treatment of certain multiple myeloma indications in territories including the U.S., Europe and Japan, Arzerra (ofatumumab, under agreement with Novartis AG), for the treatment of certain chronic lymphocytic leukemia indications in the U.S., Japan and certain other territories and TEPEZZA (teprotumumab, under agreement with Roche granting sublicense to Horizon Therapeutics plc) for the treatment of thyroid eye disease in the U.S. A subcutaneous formulation of daratumumab, DARZALEX FASPRO (daratumumab and hyaluronidase-fihj), has been approved in the U.S. for the treatment of adult patients with certain multiple myeloma indications. Daratumumab is in clinical development by Janssen for the treatment of additional multiple myeloma indications, other blood cancers and amyloidosis. A subcutaneous formulation of ofatumumab is in development by Novartis for the treatment of relapsing multiple sclerosis. Genmab also has a broad clinical and pre-clinical product pipeline. Genmab's technology base consists of validated and proprietary next generation antibody technologies - the DuoBody platform for generation of bispecific antibodies, the HexaBody platform, which creates effector function enhanced antibodies, the HexElect platform, which combines two co-dependently acting HexaBody molecules to introduce selectivity while maximizing therapeutic potency and the DuoHexaBody platform, which enhances the potential potency of bispecific antibodies through hexamerization. The company intends to leverage these technologies to create opportunities for full or co-ownership of future products. Genmab has alliances with top tier pharmaceutical and biotechnology companies. Genmab is headquartered in Copenhagen, Denmark with sites in Utrecht, the Netherlands, Princeton, New Jersey, U.S. and Tokyo, Japan.

See more here:
Genmab Announces European Marketing Authorization for the Subcutaneous Formulation of DARZALEX (daratumumab) for the Treatment of Patients with Multi...

Having a good level of vitamin D may reduce the risk of certain cancers – Yahoo Singapore News

A new European review has found that having a good vitamin D level may be linked with a lower risk of certain cancers.

Carried out by researchers from the University of Eastern Finland and the Autonomous University of Madrid, the new review includes updated information on the molecular basis ofvitaminD and how it may play a role incancerprevention.

The article, which was published in the journalSeminars in Cancer Biology, says that studies which have looked into the effect of the vitamin on different types of cancers suggest that a 'good' vitamin D level, measured as the level of 25-hydroxyvitamin D in the blood, could be particularly beneficial for reducing the risk of colorectal cancer and blood cancers, such as leukemias and lymphomas. Low vitamin D status has also been linked with an increased risk of breast and prostate cancer.

Moreover, low vitamin D levels also appeared to be linked with a higher rate of cancer and a poorer prognosis.The researchers explain that vitamin D is important both for the functioning of blood cells and adult stem cells in rapidly regenerating tissues, such as colon or skin. If vitamin D levels are too low then the cells function at a suboptimal level, which may lead to them to turn into uncontrolled growing cancercells.

However, the team also note that randomized controlled trials which have looked into how vitamin D supplementation might be able to reduce cancer mortality have provided inconsistent results. The authors say the role of vitamin D might be clearer if studies took into account individual vitamin D responsiveness, which is an individual's molecular response or sensitivity to vitamin D supplementation. They add that high vitamin D responsiveness may be linked to a lower risk of cancer. Vitamin D responsiveness varies between individuals, and affects their need for vitamin D supplementation. For example, 25 percent of the Finnish population seem to be low responders, say the researchers, and therefore need a higher dose of vitamin D supplementation.

Vitamin D levels can be measured using a 25-hydroxyvitamin D blood test. Alevelof 20 nanograms/milliliter to 50 ng/mL is considered to be adequate for healthy people. Alevelless than 12 ng/mL indicatesvitamin Ddeficiency.

Read this article:
Having a good level of vitamin D may reduce the risk of certain cancers - Yahoo Singapore News

Major skin cancer research study to begin at The Hormel Institute – Austin Daily Herald – Austin Herald

Dr. Rebecca Morris, leader of the Stem Cells and Cancer lab at The Hormel Institute, received a multi-year grant to study stem cells originating in adult bone marrow and their possible effects on skin diseases, including cancer. The grant, from the Nation Institute of Healths National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), awarded Morris with $373,688 over two years for her research project Identification of Novel Epidermal Progenitors.

Morris said this research is significant because it will contribute new understanding of epithelial biology, and blood and bone marrow in general, provide possible new targets for epithelial cancer prevention and control, validate liquid biopsy of blood as a diagnostic tool, and help her and her team to achieve their goal of preventing and alleviating chronic skin diseases including cancer, psoriasis, and epidermolysis bullosa.

Many years ago, I contributed basic research on identification and isolation of adult tissue stem cells from skin epidermis, and demonstrated their role in skin cancer initiation and promotion, Morris said. Now, I am again thrilled to be on the edge of discovery of a new population of epithelial stem cells and have the opportunity to determine their roles in regeneration and cancer.

Cells in the body that cover surfaces (like the epidermis, or top layer of skin) or line spaces (like ducts in mammary gland or lining of the colon) are called epithelial cells. In adults, most cancers originate from these epithelial cells. However, new research has identified certain bone marrow derived epithelial cells (BMDECs) in normal, healthy human subjects.

Morris and her team do not believe anyone has yet described the features and nature of these cells, or analyzed their function.

The research team has hypothesized that the epithelial cells from the bone marrow are epithelial stem cells. They therefore hope to demonstrate that BMDECs include a novel population of adult tissue stem cells that can be gathered to chronically compromised epithelium, such as skin cancer or psoriasis, and regenerate it.

Skin cancer is by far the most common type of cancer in the United States, with millions of people diagnosed each year. As we enter summer, it is important to remember simple steps like staying out of the sun during the middle of the day, staying in the shade, and wearing sunscreen can help reduce your skin cancer risk.

Next steps for Morriss research include determining how these blood borne epithelial cells are recruited to the skin, the recruiting molecules, how the recruitment can be good or bad, and how to modulate their recruitment to alleviate disease.

See the original post here:
Major skin cancer research study to begin at The Hormel Institute - Austin Daily Herald - Austin Herald

Transforming the spleen into a liver-like organ in vivo – Science Advances

INTRODUCTION

The shortage of human organs suitable for transplantation is a global challenge (1). Tissue engineering has shown promise for growing tissues in vitro to replace dysfunctional organs in vivo (2). However, its application in regenerating large functional organs has achieved little clinical success (3). One major reason is that those organs have abundant, well-orchestrated blood vasculature, which is too complex for the current techniques (e.g., codelivering angiogenic agents) to recapitulate in engineered tissues (4). Poor vascularization has become the critical limitation accounting for the unsatisfactory performance of engineered tissue transplants in the body (5).

We proposed here the approach of transforming, instead of transplanting or engineering, an organ to solve the problem. It aimed to reconstruct an existing organ in the body to develop the function of another, dysfunctional one. An existing organ offers extensive, interconnected vasculature that is difficult to mimic in engineered tissues and provides a cell-adhesive microenvironment (6, 7). Furthermore, the organ to be transformed should be functionally dispensable and large enough so that it can adequately perform the function of a vital, large organ (like the liver). Evidence in multiple aspects suggested that the spleen would be such a suitable organ for transformation. First, as a large, peripheral lymphoid organ, the spleen is essentially a vestigial hematopoietic organ (8). Its functions could largely be compensated by the lymph nodes (for lymphoid) and the liver/kidney (for blood filtration). Second, the spleen is relatively large (>170 cm3 in adult humans) and has abundant blood supply from the hepatic artery (9). Third, patients who received splenectomies do not suffer from severe conditions (10), further confirming its dispensability.

To validate our hypothesis, and aimed at liver regeneration, we devised a two-step method to transform a mouse spleen into a functioning liver. In the first step (remodeling), through repeated injection of a tumor extract, the supernatant of a tumor tissue homogenate [solute tumor homogenate (STH)], we remodeled the tissue matrix of the spleen to support the growth of epithelium and establish an immunosuppressive microenvironment. In the second step (transplantation), on the basis of our previous findings that transspecies cell lines could growbut be confinedin the established immunosuppressive niche, we transplanted several auto-, allo-, and xenograft liver cells into the remodeled spleen. These cells grew and functioned in the host; in particular, the allograft primary cells exerted comprehensive liver-specific functions to support the survival of the mice suffering from 90% hepatectomy or drug-induced liver failure. Our results suggest that we have transformed the spleen into an organ that functions as a liver.

We transformed the spleen into a functioning liver in two steps: remodeling the spleen tissue and transplanting liver cells into the remodeled spleen. The transplanted liver cellsautograft, allograft, or xenograftshould survive, grow, and perform the physiological functions of the liver in the mouse body (hepatization).

Before these two steps, we performed a preparatory, but essential, surgical operation to translocate the spleen to facilitate subsequent repeated intraspleen injections. Specifically, we moved the murine spleen from its original site in the abdominal cavity to a subcutaneous location (Fig. 1A), with the surgery recorded in movie S1 and further illustrated in fig. S1A. Successful translocation of the spleen could be confirmed by both microcomputed tomography (micro-CT) (Fig. 1B) and direct observation in the euthanized mice (Fig. 1C). Seven days after the operation, the animals showed a satisfactory recovery, with no notable weight loss (fig. S1B) or physiological abnormities (table S1). The translocated spleen maintained its natural size, morphology (Fig. 1C), and weight [expressed as spleen weight/body weight (SW/BW); Fig. 1D], and its gene expression was the same as that of the normal spleen (Fig. 1E). Meanwhile, the weight, histology, and zonation of the liver, as well as the flow of the liver portal venous, were not altered after the spleen translocation (fig. S1, C to F). In accordance with our design, the translocated spleen could be easily accessed for subcutaneous injections (movie S2 and fig. S1G).

(A) Illustration of spleen remodeling. (B) The spleen (white dotted) before and after translocation (micro-CT). (C) Gross view of a normal and translocated spleen. (D) Mean SW/BW of normal and translocated spleens 7 days after translocation. (E) Microarray analysis of gene expression between normal and translocated spleens. IRCs, immune-related cytokines. (F to O) The translocated spleens injected with phosphate-buffered saline (PBS) or STH three times over 7 days with their (F) gross view; (G) SW/BW; (H) gene expression compared by microarray; (I) expression of typical genes of ECM, chemokines, growth factors (GFs), and cytokines; (J) contents of hydroxyproline, COL1, and COL4, plus the hardness of the spleens (n = 8 for COL4); (K) hematoxylin and eosin (H&E) and (L) Massons trichrome staining; (M) expression of COL1, COL4, and smooth muscle actin (-SMA) (inset scale bar, 200 m); (N) levels of growth factors and cytokines [enzyme-linked immunosorbent assay (ELISA); values normalized to PBS group]; and (O) three-dimensional (3D) reconstructed micro-CT images showing vascularization, with the vessel area measured. (P) Average proportion of different cell populations in the spleens treated with PBS or STH. Images are representative of three independent experiments. Results are shown as means SEM (n = 5 unless otherwise noted). Statistics: (D, G, J, N, and O) Students t test. TNF-, tumor necrosis factor; IFN-, interferon-; TGF-1, transforming growth factor1; EGF, epidermal growth factor; HGF, hepatocyte growth factor; VEGF, vascular endothelial growth factor. Photo credit: Lintao Wang, Nanjing University.

Then, we started to perform remodeling as the first step of the organ transformation. We used STH to remodel the tissue matrix of the translocated spleen for two purposes: (i) suppress immune rejection for accommodating allo-/xenograft liver cells and (ii) increase extracellular matrix (ECM) production (which is low in the spleen) to support epithelial development. We prepared STH from four different allograft tumor modelsS180 (sarcoma), Hepa1-6 (hepatoma), 4T1 (breast cancer), and B16-F10 (melanoma)which are all murine cancer cell lines implanted in mice. We compared the activity of all STH samples to promote interleukin-10 (IL-10) expression in mouse bone marrowderived macrophages and enhance type I collagen (COL1) production in mouse embryonic fibroblasts (MEFs). Among them, the STH generated from the S180 model outperformed other samples in inducing the expression of the two genes (fig. S2, A and B). Thus, we selected the S180-derived STH (termed as STH) in the following experiments.

We analyzed the protein content of STH by liquid chromatographymass spectrometry (LC-MS) and several cytokines by enzyme-linked immunosorbent assay (ELISA; table S5 and fig. S2C). The concentrations of IL-10 and transforming growth factor1 (TGF-1) were high according to the ELISA results. We injected STH into the spleen three times, at days 7, 10, and 14 after the translocation operation. Each time, we injected 50 l, which is the maximal value that can be injected to the spleen, to five separate sites (10 l at each site), as illustrated in fig. S1 (H and I). At day 17, the mice were euthanized, and the spleens were collected for analysis. The three injections of STH markedly changed the characteristics of the spleen tissue in various aspects. First, the size of the STH-treated spleens was enlarged (Fig. 1F), and their average weight was about twice of that of the phosphate-buffered saline (PBS)treated ones (Fig. 1G). Second, the expression of more than 4800 genes in the STH-treated spleen was far different from that in the normal spleen (Fig. 1H). Cluster analysis based on the variations highlighted in Fig. 1 (E and H) uncovered that most notable alterations occurred to the genes encoding ECM molecules, chemokines, growth factors, and immune-related cytokines (Fig. 1I). Third, the ECM composition was also changed, in agreement with the above outcomes of gene analysis. The hydroxyproline content of the STH-treated spleen was much higher than that of the control spleens because the content of COL1 in the spleens was increased, although that of COL4 remained unchanged, which made the STH-treated spleens considerably stiffer (Fig. 1J) (11).

We continued to examine more detailed histological changes in the remodeled spleen. As revealed by hematoxylin and eosin (H&E) staining, in the STH-treated spleen, the typical spleen structure of red and white pulps disappeared, the density of lymph cells decreased, and the deposition of matrix increased (Fig. 1K). Meanwhile, Massons trichrome staining illustrated a higher amount of tissue matrix, particularly collagens, in the transformed spleens (Fig. 1L). Further immunofluorescent staining for COL1, COL4, and smooth muscle actin (-SMA) in a whole spleen section showed that the STH treatment markedly increased the density of COL1 (but not COL4). The enhanced deposition of COL1 might be a result of the increased number of the -SMApositive fibroblasts (Fig. 1M), of which the distribution was heavily altered in the spleen tissue. Nevertheless, the hydroxyproline content, reflecting the concentration of collagen, in the STH-treated spleen was comparable to that of normal liver tissue (fig. S2D) (12).

Cytokine profiling further highlighted the radical changes in the matrix microenvironment in the remodeled spleen. Examination of key cytokines by ELISA (Fig. 1N) revealed that vascular endothelial growth factor (VEGF) was among the most up-regulated ones, indicating enhanced angiogenesis in the transformed spleen. This was further confirmed by micro-CT that showed a higher density of blood vessels in the STH-treated spleen than in the normal one (Fig. 1O). Meanwhile, the elevated variation of TGF-1, IL-10, and interferon- (IFN-; together with the histological changes) indicated a marked change in the cellular composition of the spleen. We observed a notable decrease in the number of both T and B cells, as well as an increase in that of fibroblasts, in the STH-treated spleen (Fig. 1P and fig. S2E). Macrophages, a key regulator of tissue microenvironment, not only increased in number but also expressed more CD206, an M2 polarization marker, in the STH-treated spleen (fig. S2E) (13). T regulatory (Treg) cells, one of the most important immune suppressors, also increased in number in the STH-treated spleen (fig. S2F) (14). Depletion of Treg cells or macrophages abolished the STHs protecting effect (fig. S2G). Meanwhile, any injections after the third did not further change the cell composition in the spleen (Fig. 1P), suggesting that three injections were enough for remodeling.

To validate the effect of STH in changing the phenotype of spleen cells, we isolated total spleen cells, treated them with STH ex vivo, and analyzed their gene expression profiles. We found that the STH treatment markedly changed the expression of key genes in spleen cells (fig. S2H); for instance, the levels of collagens, fibroblast growth factor (FGF), epidermal growth factor (EGF), VEGF, and IL-10 were significantly up-regulated, and the expression of IL-12 and tumor necrosis factor (TNF-) decreased. Furthermore, we purified the spleen macrophages for the same treatment and analysis; the outcomes showed a clear decrease in the expression of typical inflammation-related genes, such as IL-12a, nitric oxide synthase 2, TNF-, IL-6, and Toll-like receptor, as well as an increase in that of anti-inflammatory cytokines such as IL-10 and TGF-1 (fig. S2I). Further measurement of key cytokines by quantitative reverse transcription polymerase chain reaction (RT-PCR) (fig. S2J) demonstrated that more spleen macrophages switched from an M1 to an M2 phenotype after STH treatment for 18 hours (15). These results highlighted the direct effect of STH in inducing an immunosuppressive phenotypic change in spleen cells.

Such effect of STH, especially its capacity in inducing immunosuppression, led us to investigate its safety for use. To do so, first, we fluorescently labeled TGF-1 (the most abundant cytokine in STH), injected it into the spleen at the same dose as in STH, and traced its distribution. Most signals were located around the injection sites in the spleen (fig. S3A), suggesting that TGF-1 at this dose was immediately captured by the local cells. Second, we isolated macrophages from different organs of both normal mice and those whose spleen had been treated with STH for 24 weeks. The macrophages from the same organexcept the spleenof the treated or untreated mice expressed IL-10 and TGF-1 to a similar extent (fig. S3B). In addition, histological analysis of the other organs (the liver, lung, kidney, and heart) revealed no abnormalities (fig. S3C). Third, because M2 macrophages were reported to promote liver fibrosis, we examined the influence of STH on liver fibrosis in a mouse model induced by CCl4. Nevertheless, injection of STH into the spleen did not affect the process of fibrosis (fig. S3D). Fourth, the long-term (24 weeks) injection of STH did not cause any adverse changes in the main indexes of the blood routine examination or serum markers for multiple organ dysfunction (table S2). We kept monitoring the overall health of the mice, including their appearance, weight, and daily activity and did not observe any sign of discomfort or abnormality. However, we still explored the possibility of replacing STH with a cocktail of cytokines. We found that a mixture of murine TGF-1, IL-10, and VEGF-A, when the concentrations of these cytokines applied were about 1000 times higher than in STH, could induce immunosuppression (fig. S4). However, to exert an effect comparable to that of the STH in inducing ECM production, the cocktail needed to be injected more frequently (11 times for cocktail versus 3 times for STH) and for a longer period (4 weeks for cocktail versus 1 week for STH). Combined, these data suggested that STH, when applied at the described dose through intraspleen injection for remodeling the spleen, exerted no adverse effect on the recipient mice. In addition, it might be possible to devise a cytokine cocktail to mimic, and eventually replace, STH, but the formers efficiency was much lower and the cytokines had to be applied in a higher dose.

In summary, these data demonstrated that STH effectively remodeled the tissue compositions and cell populations of the translocated spleen, in favor of tissue development. STH did not cause any adverse effect to the recipient mice.

Transplantation of allogeneic or xenogeneic cells into immunocompetent hosts can trigger acute immune rejection leading to transplant failure (16). Before testing the transplantation of liver cells of various origins, we evaluated whether the STH-remodeled spleen could provide an immunosuppressive microenvironment to accommodate xenograft cells.

We injected HepG2 cells (1 106), a human cancer cell line, into the STH-remodeled spleen in C57BL/6 mice (Fig. 2A). First, we examined the gene expression profile in the spleens by microarray 8 hours after the transplantation, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The levels of key genes/pathways related to immune rejection were lower in STH-remodeled spleens than in PBS-treated ones, both having received HepG2 xenograft (Fig. 2B). In addition, the immunoglobulin G (IgG) and IgM concentrations in the blood from the mice with the remodeled spleen were unchanged in the first 3 days after transplantation and gradually increased in the next few days (Fig. 2C). This trend was very different from what was observed in the PBS-treated mice, where both IgG and IgM increased immediately upon cell transplantation. The change in the levels of TNF- and IFN-, two typical cytokine indicators for immune rejection, was consistent with that of the IgG and IgM in both animal groups (Fig. 2C).

(A) Scheme of xenografting HepG2 cells into the spleen. (B) Genome-wide microarray profiling of the spleen 8 hours after HepG2 transplantation. Signaling pathways (KEGG, left) and gene expression (heat map, right) related to immune rejection in STH/PBS-treated spleens. NOD, nucleotide oligomerization domain; JAK-STAT, Janus kinasesignal transducers and activators of transcription. (C) Serum concentration of immunoglobulins and cytokines and (D) CD4+/CD8+ T cell proportion in peripheral blood (statistics on the right) in mice 1 week after HepG2 transplantation. (E) Scheme of assessing long-term rejection of xenografted HepG2, with (F) the determination of growth factors and cytokines in the spleen. (G) Scheme of analyzing the response in OT-1 mice, whose (H) expression level of IFN- by spleen cells 4 days after stimulation with OVA-expressing HepG2 (OVA-HepG2) cells and (I) CD8+ T cell proportion expressing IFN- in the spleen (statistics on the right). Furthermore, OT-1 mice with OVA-HepG2 cell transplantation and long-term STH treatment were analyzed against the group with STH injection ceased for 14 days: (J) IFN- expression by the spleen cells and (K) CD8+ T cell proportion expressing IFN-. Images are representative of three independent experiments. Results are shown as means SEM (n = 5). Statistics: (F) one-way and (C and D) two-way analysis of variance (ANOVA) followed by Bonferronis multiple comparisons post hoc test and (H to K) Students t test.

Next, we assessed the change in CD4+ and CD8+ T cell proportions in the spleens 7 days after the transplantation of HepG2 cells. The fluorescence-activated cell sorting (FACS) data showed that CD4+ cells in the remodeled spleen maintained a much lower level compared with the control group; the proportion of CD8+ T cells, although starting to increase 1 day after transplantation, was still lower than that in the normal spleen most of the time (Fig. 2D). In addition, histological analysis of the spleen sections 4 days after transplantation indicated that inflammatory responses were minimal in the STH-transformed spleens but obvious in the PBS-treated control group (fig. S5A).

Then, we continued to monitor immunosuppression in the transformed spleen for a longer period of 30 days, with STH injected every 4 days (schemed in Fig. 2E). As a control group, injection of STH was ceased 14 days after cell implantation. We analyzed the T cell profiles (fig. S5B) and cytokine expression (Fig. 2F). We further evaluated CD8+ T cell activation in an OT-1 mouse model, in which the CD8+ T cells express a T cell receptor recognizing the SIINFEKL peptide of ovalbumin (OVA) (Fig. 2G) (17). Upon the stimulation of OVA-expressing HepG2 (OVA-HepG2) cells, the CD8+ T cell activation (as marked by the expression of IFN-) was significantly lower in the STH-remodeled spleen than in the PBS-treated one, as indicated by the expression of IFN- in spleen cells (Fig. 2H) and the ratio of IFN-+/CD8+ T cells (Fig. 2I). As long as STH was injected, the CD8+ T cells remained inactivated; once STH injection was ceased, these cells became activated (Fig. 2, J and K). These results suggested that continued STH injection was both effective and necessary to maintain the immunosuppressive environment in the spleen following the transplantation of the liver cells. In addition, the increased blood vessels in the spleen seemed unchanged after the end of STH injection (fig. S5C).

Together, these data demonstrated that the immunosuppressive niche established by STH treatment prepared the transformed spleen for accommodating xenograft cells. The transplanted human hepatoma did not trigger acute immune rejection.

As demonstrated above, continued STH injection (once every 4 days) could maintain an immunosuppressive niche in the remodeled spleen. We set out to comprehensively investigate the growth of autologous, allogeneic, and xenogeneic liver cells in this niche. As illustrated in Fig. 3A, after translocating and remodeling the spleen, we transplanted four different types of liver cells into the site and continued STH injection for another 8 weeks before sample collection and analysis. The transplanted cells included the following: (i) autograft liver cells (autoHEPs), (ii) allograft mouse liver cells (alloHEPs), (iii) xenograft human primary liver cells (hHEPs), and (iv) xenograft human induced pluripotent stem cell (iPSC)derived liver cells (hiPS-HEPs). For most groups, we monitored the cell growth for 8 weeks after their transplantation; for one group of alloHEPs, we continued STH injection for 24 weeks to evaluate their long-term growth.

(A) Implantation of liver cells to remodeled spleens. (B to E) Hepatocytes from green fluorescent protein (GFP) transgenic mice were transplanted to the remodeled spleens for 8 weeks, followed by (B) FACS counting of GFP+ cells, (C) SW/BW, (D) bioluminescence imaging and quantification, (E) H&E staining (asterisks, bile infarcts), (F) gross view, and (G) immunostaining for GFP (star, spleen-liver boundary). DAPI, 4,6-diamidino-2-phenylindole. (H to J) Long-term growth of the allograft hepatocytes (24 weeks), with (H) GFP signal in the spleen sections, (I) H&E staining and immunostaining for cytokeratin 7 (for bile duct), and (J) liver zonation analysis using glutamine synthetase (GS) immunostaining. (K to Q) Human primary hepatocytes alloHEPs or hiPS-HEPs were transplanted to the remodeled spleens in mouse for 8 weeks, followed by (K) human-specific genomic DNA (gDNA) detection, (L) SW/BW, (M) determination of human serum albumin (HSA), (N) gross view, (O) H&E staining (stars, bile infarcts), (P) immunostaining of HSA and Ku80 in the spleen (stars, spleen-liver boundary), and (Q) fluorescent in situ hybridization (FISH) analysis of the spleen sections for human GAPDH (glyceraldehyde-3-phosphate dehydrogenase). Images are representative of three independent experiments. Results are shown as means SEM (n = 5). Statistics: (M) one-way and (B, D, and K) two-way ANOVA followed by Bonferronis multiple comparisons post hoc test and (C and L) Students t test. Photo credit: Lintao Wang, Nanjing University.

For autoHEPs, the hepatocytes harvested from the same recipient mice, they grew larger tissue (gross view; fig. S6A) and proliferated faster [5-ethynyl-2-deoxyuridine (EdU) staining; fig. S6C] in the spleens remodeled by STH than in those treated with PBS.

For alloHEPs, we derived primary murine hepatocytes from green fluorescent protein (GFP) transgenic or luciferase transgenic mice for transplantation. These cells settled well and proliferated in the STH-remodeled spleen, as examined by cell counting (Fig. 3B), spleen weighing (expressed as SW/BW; Fig. 3C), EdU staining (fig. S6G), and bioluminescence imaging (Fig. 3D). In addition, they formed neotissue in the remodeled spleen, as evidenced by histological analysis (Fig. 3E and fig. S6D), morphology (Fig. 3F), and GFP immunostaining (Fig. 3G). Meanwhile, we traced the formation of bile duct (fig. S6E) and found that hepatic stellate cells took part in liver-like microstructure formation (fig. S6F). Furthermore, through the long-term (24 weeks) development, these alloHEPs expanded to occupy most space in the spleen (Fig. 3H) and developed the typical hepatic structure including the bile ducts (Fig. 3I) and liver zonation [indicated by the specific distribution of glutamine synthetase (GS); Fig. 3J].

For xenograft cells, both hHEPs and hiPS-HEPs showed desirable settlement and growth in the STH-remodeled (but not the PBS-treated) spleen, as validated by quantification of human genomic DNA (gDNA; Fig. 3K), SW/BW (Fig. 3L), as well as measurement of the human albumin content in the spleen (Fig. 3M). Meanwhile, both cells were proliferating in the remodeled spleen, while hiPS-HEP proliferated faster than the primary human liver cells, according to the outcomes from the EdU staining (fig. S6G). Moreover, they generated xenograft tissues in the remodeled spleen, as evidenced by morphological observation (Fig. 3N), histological staining (H&E; Fig. 3O and fig. S6D), human serum albumin (HSA) and Ku80 coimmunostaining (Fig. 3P), and fluorescent in situ hybridization (FISH) for human GAPDH (glyceraldehyde-3-phosphate dehydrogenase) (Fig. 3Q). Similar to the finding with alloHEPs, the tissue formed by human primary hepatocytes also contained bile infarcts (Fig. 3O). The above data indicated that the human hepatocytes were fully functioning in the STH-remodeled spleen in mice.

Continued STH injection after the transplantation was essential. We found in a separate experiment that, except for autoHEPs, all the implanted alloHEPs or xenograft hiPS-HEPs disappeared from the spleen only 2 weeks after we stopped STH injection (fig. S6H). Furthermore, STH treatment did not induce fibrosis in the implanted hepatocytes (fig. S7A), and long-term injection of STH did not change the properties of new hepatocytes (fig. S7B). Together, the above data validated that the STH-transformed spleens in mice supported the survival, proliferation, and development of auto-, allo-, and xenograft hepatic cells, including human primary liver cells.

On the basis of the above finding that transspecies hepatocytes could grow well in the STH-remodeled spleen with minimal rejection, we speculated on generating a hepatic tissue in the remodeled spleen with xenograft hepatoma cells, which would be valuable in solving the problem of the shortage of primary cells for transplantation. As indicated in Fig. 4A, we transplanted HepG2 cells into the translocated and STH-remodeled spleen and continued STH injection for another 8 weeks before sample collection and analysis. Our pilot experiment suggested 1 106 to be the minimal number of HepG2 cells that could steadily grow in the remodeled spleen (fig. S8A).

(A) Schematic diagram of intrasplenical injection and assay of the growth of transspecies HepG2 cells in the STH-remodeled or PBS-treated spleen. (B) Bioluminescence imaging and corresponding quantitative analysis based on photon counts gated over the spleen. (C to F, I, and K) The spleens treated as in (A) were analyzed at 8 weeks after HepG2 transplantation. (C) Representative gross view and (D) micro-CT scanning, with the section highlighting maximal splenic cross-sectional area; red dotted area indicated the liver-like region. (E) H&E staining of the spleens with EdU labeling identifying cells under proliferation. (F) Representative confocal microscopy images of the spleen stained for HSA or Ku80 and expressing GFP. The star indicated the boundary of spleen and liver tissue. (G) FACS-based quantitative analysis of GFP transgenic HepG2 cells in the spleen treated as in (A) at the indicated time. (H) Weight of the spleen treated as in (A) at the indicated time. (I) Representative immunostaining of HSA across the whole frozen spleen sections. (J) The levels of HSA in the spleens treated as in (A) at the indicated time. (K) Representative FISH of human GAPDH in the spleens. Images are representative of three independent experiments. Results are shown as means SEM (n = 5 per group). Statistics: (B, G, H, and J) two-way ANOVA followed by Bonferronis multiple comparisons post hoc test. Photo credit: Lintao Wang, Nanjing University.

We first used luciferase-labeled HepG2 cells, which enabled us to monitor their growth under a whole-body fluorescent imaging system. Through an 8-week observation, we found that the fluorescent signal steadily increased in the STH-remodeled spleens but quickly disappeared in the control group (Fig. 4B), suggesting that the transplanted cells proliferated only in the transformed spleens. Furthermore, the gross view (Fig. 4C) of the spleen 8 weeks after cell implantation illustrated notable changes in morphology and size of the transformed spleens accommodating HepG2, which appeared much larger than the ones in the control group. The micro-CT images obtained from the mice intravenously injected with the contrast agent further highlighted the outcomes of spleen transformation (Fig. 4D). An enlarged spleen was illustrated in the STH-remodeled, HepG2-implanted group. The radiolucent signals showed a generally homogeneous distribution, with substantial enhancement (red dotted) in certain zones that represented neonatal tissues with extensive angiogenesis. Consistently, H&E and EdU proliferation staining (Fig. 4E) revealed the formation of neotissue by HepG2 cells with notable proliferating signals.

We then used GFP-labeled HepG2 cells, which allowed us to quantify their proliferation by sorting the GFP-positive cells with FACS. Before sorting, we could already observe in the tissue sections a large number of GFP-positive cells (coimmunostained with HSA or Ku80) in the STH-transformed spleen (Fig. 4F). The sorting outcomes confirmed that the number of GFP-positive HepG2 cells in the STH-remodeled spleen increased approximately five times in 8 weeks; in contrast, these cells quickly diminished in the control group only 2 weeks after transplantation (Fig. 4G). This finding was consistent with the trend of the change in the spleen weights (Fig. 4H). The images from immunostaining of HSA illustrated abundant expression and wide distribution of HSA across the whole sectioning area of the transformed spleen 8 weeks after transplantation (Fig. 4I). An HSA-specific ELISA revealed a steady increase in the concentration of HSA along the time (Fig. 4J). In addition, immunofluorescent staining for CYP2D6, another specific marker of human hepatocytes (fig. S8B), and FISH for human GAPDH mRNA further confirmed the formation of neotissue by HepG2 cells (Fig. 4K).

In another experiment using rat hepatoma cell McA-RH7777, we found that McA-RH7777 cells proliferated faster and grew larger tissue in the mouse spleen remodeled by STH than in that treated with PBS. One injection of 1 106 McA-RH7777 cells could expand to 2.0 107 in 8 weeks (fig. S8, C to F).

It should be highlighted that these xenograft cells were confined in the spleen and did not spread to other tissues throughout the study (Fig. 4B), except for the signal temporarily present in the lungs and livers shortly after the injection. This finding was important evidence to emphasize the safety of this approach. We further traced GFP-labeled HepG2 cells and searched for human-specific gDNA in different organs, and the data suggested no HepG2 cells spreading to other organs than the spleen (fig. S8, G and H). In addition, after the termination of STH injection, the implanted HepG2 cells were rapidly eliminated (fig. S8I).

Together, the above data validated that the STH-remodeled spleens in mice could support the proliferation of human and rat hepatoma cells, which formed into new tissues that express hepatic markers of human or rat origin in the mouse body. The location of these xenograft cells was strictly confined within the spleen.

We lastly evaluated whether the allo-/xenograft liver cells growing in the transformed spleen could perform the functions of the liver. The alloHEPs, growing in the STH-transformed spleens for 8 weeks, exerted the typical hepatic functions of synthesizing lipids and glycogen, as illustrated by the Oil Red O staining and the periodic acidSchiff (PAS) staining (Fig. 5A). To more critically assess the hepatic function of the transformed spleen, we performed 90% hepatectomy (fig. S9, A and B) in the mice implanted with alloHEPs and evaluated whether the transformed spleens could recover the essential liver functions to save the animals from death. First, an indocyanine green (ICG) clearance test (Fig. 5B) and follow-up observation of the frozen sections of the spleen (Fig. 5C) revealed that the transformed spleen restored the livers uptaking capacity. Second, the mice, either with STH treatment but without cell transplantation or with PBS treatment followed by cell transplantation, all died within 48 hours after the hepatectomy. Only the mice with the STH-remodeled spleen and alloHEPs transplantation could all survive through the study period (Fig. 5D).

(A to D) Transplantation of alloHEPs into transformed spleens, followed by (A) Oil red O and PAS staining of the spleen, (B) ICG clearance test, (C) frozen spleen sections, and (D) mice survival after 90% hepatectomy. (E to H) Xenografting of human hepatocytes (HepG2 or hiPS-HEP) into transformed spleens: (E) liver-specific metabolic profiling (quantitative PCR; n = 3 for liver samples), (F) serum HSA concentration after HepG2 transplantation, (G) serum HSA concentration 15 days after hiPS-HEP transplantation, and (H) human-specific debrisoquine (DB) metabolite formations (n 5). AUC, area under the curve; 4OHDB, 4-hydroxydebrisoquine. (I) Oil red O and PAS staining of the spleen. (J) Frozen spleen sections after ICG intravenous injections. (K to P) Examination of McA-RH7777 cells in transformed spleens. (K) Coimmunostaining for CYP26A1 with prothrombin or GS in the spleens 30 days after transplantation. (L) H&E staining of the hepatized spleen with or without d-galatosamine hydrochloride (d-Gal) exposure. (M) The prothrombin time (PT) and (N) the level of blood ammonia 6 hours after exposure to d-Gal. (O) Mice survival after d-Gal treatment or (P) 90% hepatectomy (n = 10). Images are representative of three independent experiments. Results are shown as means SEM (n = 5 unless otherwise noted). Statistics: (B, G, H, M, and N) one-way and (F) two-way ANOVA followed by Bonferronis multiple comparisons post hoc test and (D, O, and P) log-rank test. HMG-CoA, hydroxymethylglutaryl-coenzyme A reductase; ACO, acyl-coenzyme A oxidase; PFK1, phosphofructokinase 1; GGT, gamma-glutamyltransferase.

We noticed that the hepatectomy procedure itself stimulated the growth of both the remnant liver and alloHEPs in the spleens. In the STH + alloHEPs group, the volume of the remnant 10% liver tissue reached about 30% size of a normal liver 7 days after the operation. The transformed spleen also doubled its size compared with that in the mice without hepatectomy (fig. S9, D and F). Meanwhile, the liver-like microstructures that included bile ducts, blood vessels, and hepatic stellate cells could be observed in the transformed spleen (fig. S9C). Although 30% liver tissue could support the animal to survive, we assumed that the mice needed to rely on the hepatic function provided by the transformed spleen to survive after the first 48 hours. To validate this assumption, we carried out small interfering RNA (siRNA) inhibition of c-Met after the hepatectomy, which suppressed hepatocyte proliferation in both the liver residue and the transformed spleen but found that it did not change the survival rate of the mice with alloHEPs (fig. S9, D to L). This outcome supported that the survival of these mice depended on the function of alloHEPs in the transformed spleen.

Xenograft human cells (HepG2 or hiPS-HEP) were then implanted to the STH-transformed spleen in mice. We first profiled the expression of the main genes involved in the metabolic pathways specific to the liver, in comparison with that of the normal liver and hepatoma tissue harvested from patients with hepatoma (table S6). The results demonstrated the expression of these genes in the transformed spleens with HepG2 cells and hiPS-HEP cells (Fig. 5E). The gene expression profile was closer to that in the normal human liver than in the hepatoma tissue even in the HepG2-derived tissue. Second, as albumin production is one key function of the hepatocytes to maintain the blood osmotic pressure and other important functions, we examined the serum HSA using human-specific ELISA. As shown in Fig. 5F, HSA was detected in the mouse serum, and its concentration increased as HepG2 cells grew. Although hiPS-HEP and hip proliferated slower than HepG2 cells in the spleens, we could detect HSA in the mouse serum (Fig. 5G and fig. S10A). Third, because human liver cells are more capable of modifying debrisoquine (DB) into 4-hydroxydebrisoquine (4OHDB) than mouse liver cells, we injected DB into the mice with human hepatocytegrafted transformed spleens (18). We observed a significantly higher serum level of 4OHDB/DB in these mice than in the control group (Fig. 5H), suggesting that the liver tissues generated by the human cell lines exhibited metabolic functions. Fourth, the Oil Red O and PAS staining (Fig. 5I) demonstrated that the hepatic tissue generated in the spleen had the typical liver function of synthesizing lipids and glycogen. Fifth, the transformed spleens transplanted with the human liver cells also developed the ability to take up ICG (Fig. 5J). Last, after 90% hepatectomy, 70% of the mice with the STH-remodeled spleen implanted with human primary hepatocytes from patients survived, whereas the control mice (i) with STH treatment and without hepatocyte transplantation or (ii) with PBS-treated spleen and grafted hepatocytes all died within 48 hours after the hepatectomy (fig. S10B).

To further test the ability of the transformed spleen in performing comprehensive functions of the liver, we again used the rat hepatic cell line McA-RH7777. One important feature of these cells is that they can resist the d-galatosamine hydrochloride (d-Gal)induced apoptosis (19, 20). We confirmed that they grew well and expressed hepatic markers in the transformed spleen; two genes were particularly obvious, prothrombin and GS, which are related to the key liver functions (prothrombin for the coagulation function and GS for the blood ammonia control) in maintaining the whole physiological homeostasis and the liver of the animals (Fig. 5K) (21, 22). We developed a fulminant liver failure model induced by d-Gal 8 weeks after the cell implantation (23). The induction was successful as indicated by the serum alanine aminotransferase (ALT) and aspartate transaminase (AST) level, histological examination of the liver tissue, and the massive animal death within 12 hours. First, both groups of mice with the McA-RH7777 cell hepatized spleens and the control mice developed massive liver injury, evidenced by the H&E staining of the liver tissue and the serum level of ALT (>150 U/liter) and AST (>90 U/liter) (fig. S10, C and D), while the tissue formed by the rat hepatic cells in the transformed spleen remained relatively intact under the challenge from d-Gal (Fig. 5L). Furthermore, the key parameters reflecting the liver functions, including coagulation time (Fig. 5M) and ammonia blood concentration (Fig. 5N), were better in the STH-transformed mice than in the control group (24). In addition, half of the mice (50%) with transformed spleen and xenograft liver tissue survived after the induction of liver failure, while all the animals in the other two groups died within 12 hours (Fig. 5O). Furthermore, we performed 90% hepatectomy in the mice implanted with McA-RH7777 and found that 80% of the animals in the STH + McA-RH7777 group remained alive (Fig. 5P), highlighting that these rat liver cells exerted hepatic functions to support the survival of the host.

As these different types of liver cells could grow and function in the transformed spleen, an interesting question emerged: How would the bile produced by these cells flow? To answer it, we injected a fluorescently labeled cholic acid into the spleen and traced the cholic acid in the liver, intestine, splenic vein, and common bile duct (fig. S11). This observation suggested that the bile, produced by the liver tissue growing in the transformed spleen, could be taken up by the biliary ducts in the liver.

The findings confirmed that transplantation of liver cellsbeing either allograft or xenograft; primary cells or cell linesinto the remodeled spleens could develop the histology and functions of the liver. Notably, the allograft primary liver cells and xenograft rat liver cell line protected the mice from death caused by drug-induced liver failure or 90% hepatectomy by restoring the liver functions that were lost or impaired in the host body. As shown in these and above data, the transformed spleen completed a hepatization procedure and adeptly performed the typical functions of the human (or rat) liver.

Cell-based approaches to regenerate large tissue/organs face several major challenges, including an insufficient number of therapeutic cells, immune rejection to allo-/xenografts, and poor vascularization of the transplants (16). In this study, we demonstrated a completely different strategy of tissue regeneration by transforming an existing organthe spleeninto a new one that performed the function of the liver in the mouse body.

This transformation builds on the remodeling of the spleen into an immune-privileged transplant bed to support xenogeneic liver cells to grow and function. We chose to remodel an existing organ, instead of delivering an engineered scaffold, because the former has inherent advantages such as highly organized vasculature, versatile stromal cells, sophisticated ECM structure, and well-formed communication with the host body, which remain hard to recapitulate in most current scaffolds (25). Our strategy resonates with other excellent studies demonstrating the benefits of using an existing organ for growing different tissuethe liver for pancreatic islets (26), lymph nodes for hepatocytes (27), and, notably, kidney for hepatocytes (2831). The present study has further extended the application of this concept, aimed at enhancing both the efficacy and feasibility of this approach. First, we chose the spleen for remodeling because this organ is functionally dispensable and well connected to the main circulation. Meanwhile, we emphasized on modifying the stromal tissue microenvironment in the spleen. This remodeling has proven effective, which increased fibroblasts and collagen content and made the organ larger, stiffer, and more suitable for anchorage-dependent epithelial cell growth. The remodeling also induced the macrophages into an anti-inflammatory, proregenerative phenotype and promoted vascularization in the spleen (32). As a result, the remodeled spleen facilitated the colonization, proliferation, and function of the xenografted liver cells.

Our findings further highlighted the importance of vascularization for tissue transplantation. Inadequate blood supply is a major hurdle for in vivo settlement of engineered tissue constructs, which encounter poor survival, function, and integration with the host. Previous studies delivered growth factors to induce angiogenesis and create a capillary bed that could support the extrahepatic engraftment of liver cells (33). These attempts provided valuable insights; however, those capillary vessels were far from the large, mature vasculature demanded for the development of most large tissue/organs (including the liver). Here, our method uses and enhances the naturally developed vasculature in the spleen, which is abundant, large, and well connected to the main circulation. As the data showed, the spleen vasculature enabled the implanted liver cells of different origins to grow and fully integrate with the host body. Such improvement is key to the overall efficacy of the transformed organ. An engineered or transplanted tissue is of physiological significance only when its volume, cell number, and functional capacity are comparable to those of its native counterpart (34). Our data showed that the transformed tissue was comparable to the native liver in size and accommodated enough functioning liver cells. It not only exhibited standard liver functions (e.g., albumin production and drug metabolism) but also, more importantly, rescued the mice from two types of extreme damages: fulminant liver failure and 90% hepatectomy. To the best of our knowledge, few previous studies have achieved this level of effectiveness.

Note that the spleen-transformed liver differs from the native liver in terms of blood supply, which may cause several consequences. The former harnesses the artery-rich splenic vasculature to support the transplanted hepatocytes, while the latter mainly relies on the portal blood with lower oxygen tension (35). Although our data showed that the transplanted hepatocytes could exert the main hepatic functions in the remodeled spleen, it is worth investigating whether the blood with possibly higher oxygen tension poses any subtle or long-term impact on the hepatocytes. Meanwhile, the native liver receives intestine-derived growth factors and other nutrients from the portal blood and filters harmful substances before they enter the main circulation. In the transformed organ, the spleen has no direct access to the portal blood; hence, the growth of the grafted hepatocytes is possibly supported by growth factors that are delivered from the main circulation or produced by the spleen (36). In addition, the protection effect of the native liver as a reticulo-endothelial organ may be weakened because the portal blood that may contain intestine-derived toxins is not immediately filtered; we speculate that these toxins can still be digested by the spleen-transformed liver tissue.

Although no abnormalities were observed in the animals, it is vital to assess the safety of this remodeling. Special attention has been paid to two parts of our approach. First, we invented a protocol of translocating the spleen to a subcutaneous site, which, as the preparatory step, brought great convenience to the whole practice. We performed comprehensive tests to confirm that the animals with translocated spleens were physiologically normal, suggesting that this procedure had no adverse effects. In addition, this procedure will not be performed if our approach is translated clinically because laparoscopy and other interventional technologies can provide surgeons with more convenience for operations on human patients. Thus, this procedure is unlikely to become a safety concern. Furthermore, although excessive tissue fibrosis might be tumorigenic, it is not a concern in our study because the extent of fibrosis induced by STH injection was much lower than that in a typical pathological scenario (e.g., liver fibrosis or pulmonary fibrosis). Neoplastic lesion-induced tumorigenesis in the spleen should be unlikely on the basis of our 24-week monitoring of these mice. Second, STH is used to remodel the spleen. Although no adverse effects of STH were found at all stages of our study, a replacement of clearer content will still be pursued. To address this issue, we demonstrated that a cocktail of cytokines might have the potential to replace STH in the future, although its efficacy needs substantial improvement and its safety requires long-term investigation.

Several factors must be considered for the future translation of this approach. First, liver diseases such as hepatitis and cirrhosis can cause substantial hypersplenism and other splenic changes, and they are among the major causes of liver failure. Therefore, further optimization of this approach in larger animals with representative kinds of liver diseases and splenic abnormalities will be of translational significance. In addition, inflammatory reactions are expected to be common under pathological circumstances and can lead to splenic enlargement; hence, anti-inflammatory treatment may be considered to aid the transformation of the spleen. Furthermore, strategies should be planned for more complex and specific clinical scenarios. For instance, for patients with immunodeficiency conditions or virus-infected diseases, the viability or function of the transplanted cells may be compromised. Customized manipulations, such as adjusting the extent of immunosuppression and exploiting genetic modification, need to be devised. Similarly, for patients with acute liver failure, the overall time of the spleen-transforming process should be shortened.

C57BL/6 mice were purchased from Vital River Laboratories (Beijing, China). C57BL/6 GFP+ mice were purchased from the Model Animal Research Center of Nanjing University (Nanjing, China). OT-1 T cell receptor transgenic mice [C57BL/6-Tg (TcraTcrb) 1100Mjb/J] were a gift from L. Li (Nanjing University, China). Luciferase transgenic mice [C57BL/6-Gt (ROSA) 26Sorem1(CAG-Luc-EGFP)Smoc] were purchased from Shanghai Model Organisms Center (Shanghai, China). The animals were fed in a specific pathogenfree animal facility with controlled light (12-hour light/12-hour dark), temperature, and humidity, with food and water available. Animal protocols were reviewed and approved by the Animal Care and Use Committee of Nanjing University and conformed to the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health.

Surgically resected paired hepatoma and normal adjacent tissues were collected from patients who had undergone surgery to remove localized liver tumors at the Affiliated Hospital of Jiangnan University (Wuxi, China). The surgically resected tissues were quickly frozen in liquid nitrogen until analysis or immediately digested and prepared for hepatocyte transplantation. The study was approved by the Human Research Ethics Committee of Nanjing University Affiliated Drum Tower Hospital (Nanjing, China) and Jiangnan University (Wuxi, China). The information about the clinical sample donors is listed in table S6.

Hepatocyte preparation. Mouse primary hepatocytes for allograft transplantation were obtained using a two-step collagenase perfusion assay as reported before (27). Mouse primary hepatocytes for autograft transplantation and human primary hepatocytes hHEPs were harvested from the digestion of liver tissues with collagenase IV and deoxyribonuclease I (both from Sigma-Aldrich, St. Louis, MO, USA). hiPS-HEPs (Y10050, Takara Bio Inc., Shiga, Japan) were thawed according to the manufacturers instruction. Two kinds of hepatoma cell lines, human hepatoma HepG2 (Institute of Biochemistry and Cell Biology, Shanghai, China) and rat hepatoma McA-RH7777 (Cobioer Technology Ltd., Nanjing, China), were respectively cultured in Dulbeccos modified Eagles medium (DMEM) and RPMI 1640 (both from Thermo Fisher Scientific, Waltham, MA, USA) containing 10% (v/v) fetal bovine serum (FBS; HyClone, Logan, UT, USA). For bioluminescent imaging, HepG2 cells were stably transfected by lentiviruses packed with the luciferase gene (GenePharma, Shanghai, China) to obtain the luciferase transgenic hepatocytes. For in vivo tracing, detection, and quantification, HepG2 cells and McA-RH7777 cells were transfected by lentiviruses packed with the GFP gene (GenePharma) to obtain the GFP-positive hepatocytes. For immune tolerance test in OT-1 transgenic mice, HepG2 cells expressing membrane-bound OVA (OVA-HepG2) were constructed by using a lentivirus to introduce the full-length OVA protein linked to the transmembrane region of H-2Db (GenePharma) (37). To isolate hepatocytes from hepatized spleens, the spleens were digested with Accumax solution (Sigma-Aldrich) to generate cell suspension. The cell suspension was filtered through 40-m cell strainers (BD Biosciences, San Diego, CA, USA) to obtain single cells. After red blood cell lysis, the pure hepatocytes were obtained using Percoll gradient method (38).

Nonhepatic cell preparation. Primary bone marrow cells obtained from 6-week-old C57BL/6 mice were cultured and differentiated into bone marrowderived macrophages in DMEM containing 10% (v/v) heat-inactivated FBS (HyClone), mice granulocyte-macrophage colony-stimulating factor (10 ng/ml) and mice macrophage colony-stimulating factor (5 ng/ml; both from PeproTech, Rocky Hill, NJ, USA), and penicillin (50 U/ml) and streptomycin (50 g/ml; both from Beyotime Biotechnology, Shanghai, China) (39). Primary embryonic fibroblasts (MEFs) were isolated, cultured, and purified from mouse fetus of C57BL/6 mice at the 13.5-day gestational age (40). The MEFs were cryopreserved at passage 1 and used from passages 3 to 5 for all experiments. Primary peripheral blood monocytes and macrophages from different organs were isolated following the manufacturers instructions (TBD Science, Tianjin, China). For isolation of splenocytes, the spleens were excised and placed in Hanks balanced salt solution (Thermo Fisher Scientific) and macerated using frosted glass slides. Cells were repeatedly aspirated with a sterile Pasteur pipette until a single-cell suspension was obtained. For isolation of splenic macrophages, after the single-splenocyte suspension was obtained, macrophages were separated using murine splenic macrophage isolation kit (TBD Science) according to the manufacturers instructions.

To acquire STH, four types of tumor cellsmouse sarcoma cell line S180 cells, mouse hepatoma cell line Hepa1-6 cells, mouse breast carcinoma cell line 4T1 cells, and mouse melanoma cell line B16-F10 cells (obtained from Institute of Biochemistry and Cell Biology)were prepared and transplanted into mice (41). Briefly, 1 106 cells were transplanted by subcutaneous injections into the left armpits of the animals. The tumors were harvested after 1 month and homogenized in equal volume of ice-cold PBS (pH 7.4) to obtain 50% (w/v) homogenate. The homogenate was then centrifuged at 12,000 rpm for 20 min at 4C to remove the insoluble tissue fragments, and the supernatant was collected. The steps of centrifugation and collection were repeated three times to obtain the final STH for subsequent use.

To analyze the composition of STH, the solution was subjected to label-free LC-MS through TripleTOF 5600 (AB SCIEX, Framingham, MA, USA) (42). Briefly, the samples were reduced with dl-dithiothreitol, alkylated with iodoacetamide, washed with tetramethylammonium bromide, digested with porcine sequencinggrade trypsin (LC-MS grade; all from Sigma-Aldrich), and subjected to LC-MS analysis. Samples were performed in three technical replicates. Identification of peptides and proteins from continuum LC-MS data was performed using the ProteinPilot 4.5 software (AB SCIEX).

For translocation of the spleen, the surgical operation illustrated in Fig. 1A, fig. S1, and movie S1 was designed by referring to a method of removing the spleen (43). The detailed process was as follows: (i) The mice were anesthetized with pentobarbital sodium (100 mg/kg of body weight) via intraperitoneal injection. (ii) The fur of mice was wetted with 70% ethanol and prevented from entering the peritoneum. (iii) A 2.5-cm-long skin incision midway between the last rib and the hip joint and a 1- to 2-cm incision in the peritoneal wall were made on the left side with the scissors. (iv) The spleen in the abdominal cavity was slightly pulled onto the exterior surface of the peritoneum and both the mesentery and connective tissue around blood vessels were cut away. (v) The peritoneal wall and the skin were closed with two separate sutures and three sutures, respectively, (vi) Any blood was removed from the skin. The mice were returned to a clean cage until the anesthesia wore off.

To remodel the spleen, the precooled PBS, STH, or cytokine cocktail containing mouse TGF-1, IL-10, and VEGF (all from PeproTech) was injected into the translocated spleen after the mouse was anesthetized. The detailed method of injection is shown in fig. S3 and movie S2. In short, PBS, STH, or cytokine cocktail (total of 50 l per mice) was injected into the spleen at five independent sites using a 31-gauge needle (BD Biosciences) every 4 days for the indicated times. After injection, the needle holes were pressed for 5 min with medical cotton ball to prevent leakage.

Before hepatocyte transplantation, the viability and number of hepatocytes were determined by trypan blue exclusion. Then, 1 106 viable hepatocytes suspended in 50 l of PBS or STH were injected directly into the remodeled spleen with a 31-gauge needle after the animals were anesthetized with pentobarbital sodium. After injection, the needle hole was pressed immediately (9).

For depletion of Treg cells, the mice with STH-remodeled spleens received intravenous injections of anti-CD25 monoclonal antibodies (0.25 mg per mouse per day; BD Biosciences) after hepatocyte transplantation. For depletion of macrophages, the mice received intrasplenic injections of clodronate liposomes (0.25 mg per mouse per day; FormuMax Scientific Inc., CA, USA) after hepatocyte transplantation.

For analysis of the influence of STH treatment on liver fibrosis, mice received intraperitoneal injections of a 25% solution carbon tetrachloride (CCl4; 4 ml/kg of body weight; Sigma-Aldrich) in sterile mineral oil twice per week for 4 weeks (44). Meanwhile, the mice received intrasplenic injections of 50 l of PBS or STH every 4 days for 4 weeks. The injections of STH started on the same day when the first dose of CCl4 was injected.

d-Gal (2 g/kg of body weight; Sangon, Shanghai, China) was injected intraperitoneally to female C57BL/6 mice (6 to 8 weeks old).

The surgical protocol of hepatectomy followed the literature (45). Briefly, the left lateral, left median, and right median lobes were removed using a single ligature (70%); afterward, the right lateral lobe (20%) was resected. Only the caudate lobes were left.

According to literature (46), to inhibit the liver regeneration after 90% hepatectomy, mice received an intravenous injection of a mixed solution of siRNAs (125 nmol/kg; Realgene, Nanjing, China) and Lipofectamine 2000 (Thermo Fisher Scientific) every 3 days for 1 week. siRNA sequences are listed in table S8.

All primary cells were washed and resuspended in medium at 2 106 cells/ml. The primary cells (1 106 cells/ml) were maintained in medium containing 50% PBS or STH (v/v) for 18 hours before further analysis.

H&E staining, Massons trichrome staining, and Sirius Red staining. Tissues were fixed in 4% paraformaldehyde (PFA) for 1 hour, processed, and embedded in paraffin. The sections (5 m thick) were prepared and stained according to the manufacturers instructions (Jiancheng Bioengineering Institute, Nanjing, China).

PAS and Oil Red O staining. Spleen tissues were immediately embedded in optimal cutting temperature (OCT; Leica Microsystems, Buffalo Grove, IL, USA) medium and sectioned. The sections were stained following the manufacturers instructions (Jiancheng Bioengineering Institute). The stained sections were photographed at different magnification times under a BX51 microscope (Olympus, Tokyo, Japan).

Fluorescence in situ hybridization. The sections (5 m) were deparaffinized, rehydrated, enzymatically disintegrated, and hybridized with human-specific GAPDH and rat-specific GAPDH (Abiocenter, Beijing, China; listed in table S9) according to the manufacturers instructions. Briefly, the sections were pretreated with pretreatment solution for 2 hours at 38C and hybridized at 40C overnight, followed by 4,6-diamidino-2-phenylindole (DAPI; Beyotime Biotechnology) for nuclear staining.

Immunostaining. Tissues were fixed in 4% PFA for 1 hour, processed, and embedded in paraffin or embedded in OCT medium. Sections (5 to 10 m) were mounted on poly-l-lysinecoated glass slides (Citotest, Haimen, China). The sections with antigen retrieval were blocked with 5% bovine serum albumin (BSA) containing 3 Triton X-100 for 1 hour and then stained with primary antibody at 4C overnight. Next, the sections were incubated with the corresponding fluorescent secondary antibody (Thermo Fisher Scientific) for 45 min at room temperature, followed by DAPI staining for nuclei. Fluorescence including GFP bioluminescence was visualized under a Leica TCS SP8 confocal microscope (Leica Microsystems).

Mice received an intraperitoneal injection of EdU (5 mg/kg of body weight; RiboBio, Guangzhou, China). After 96 hours, the spleen was harvested, embedded in OCT, and sectioned. Staining of the EdU-labeled spleen was performed using Cell-Light Apollo Stain Kit (RiboBio) according to the manufacturers instructions. Sections were counterstained with DAPI and viewed under a Leica TCS SP8 confocal microscope.

Mice received an intravenous injection of ICG (10 mg/kg of body weight) 12 hours after 90% hepatectomy. After 15 min, the blood samples were collected, allowed to coagulate at room temperature, and centrifuged to obtain serum. The concentration of ICG in serum was analyzed using Varioskan LUX (Thermo Fisher Scientific) at 800 nm. Besides, to further detect the ICG uptake by hepatic spleens, the spleens were quickly removed. The spleen was immediately embedded in OCT and sectioned. Followed by three washes with PBS, the sections were photographed under a BX51 microscope.

DB (Sigma-Aldrich), which was metabolized to 4OHDB (Enzo Life Sciences, Farmingdale, NY, USA) in humans but negligible in mice (18), was orally administrated (2 mg/kg of body weight) to the mice with remodeled spleens and transplanted human hepatocytes (HepG2 or hiPS-HEP) with untreated mice as control. Blood samples from the mice were collected 8 hours after drug administration. Plasma was separated by centrifugation from the blood. An internal standard (niflumic acid, Sigma-Aldrich; 1 M) in 100 l of methanol was added to 5 l of plasma and centrifuged (15,000 rpm, 4C, 5 min). The supernatant was subjected to LCtandem MS (1290 Infinity LC/6460 QQQ MS, Agilent Technologies, Santa Clara, CA, USA). The sampling volume was 10 l. The mobile phase was a water:acetonitrile (6:4) solution. In the first and second analysis of MS, the turbo gas was maintained at 300 and 250C and the flow rate was set as 10 and 7 liters/min, respectively. The ionspray voltage was 3500 V; the m/z (mass/charge ratio) transition (Q1/Q3) ratios for DB, 4OHDB, and internal standard were 176.5/134.2, 192.6/132.1, and 283.2/245.4, respectively. The metabolic ratios were determined by dividing AUC8h (the area under the curve at 8 hours) of 4OHDB by that of DB.

Five cell/spleen samples from three independent experiments were pooled according to individual groups. For most experiments, total RNA was extracted with TRIzol (Thermo Fisher Scientific). The quantity and quality of RNA were measured by NanoDrop ND-1000 (Thermo Fisher Scientific). RNA integrity was assessed by standard denaturing agarose gel electrophoresis. The RNA sample for gene expression profiling was hybridized on the Whole Mouse Genome Oligo Microarray (4x44 K, Agilent Technologies) and analyzed with KangCheng Biotechnology (Shanghai, China). The scatterplot with highlighted genes was produced using ggplot 2. Hierarchical clustering was performed using Cluster 3.0, and the result was viewed by using Java TreeView. The significantly differential genes were sorted by setting a twofold change. KEGG pathway analysis was performed in the standard enrichment computation method. After KEGG pathway analysis, the pathways of interest and related genes were displayed in a histogram and heat map.

gDNA was isolated from the spleen tissue using a DNA extraction kit (Generay, Shanghai, China). The total RNA was extracted with TRIzol. The quantity and quality of DNA/RNA were measured by NanoDrop Lite (Thermo Fisher Scientific). For RT-PCR, 1 g of RNA was reversely transcribed into cDNA with PrimeScript RT Master Mix (Takara Bio Inc.). PCR was performed using AmpliTaq Gold Fast PCR Master Mix (Thermo Fisher Scientific) on Applied Biosystems ProFlex PCR (Thermo Fisher Scientific). Quantitative real-time PCR (qPCR) was performed with SYBR Premix Ex Taq on an ABI 7500 fast real-time PCR system (Applied Biosystems, Waltham, MA, USA). Each sample was analyzed in triplicate within one test and repeated for three independent tests. Primer sequences are listed in table S7. For gDNA quantification, the standard curve was generated by qPCR analysis of commercial standard gDNA (Sangon). The quantity of gDNA was calculated by its interpolation in the standard curve. For qPCR analysis, the level of each target gene in each sample was normalized by subtracting the mean Ct value of -actin gene from the mean Ct value of the target gene (Ctmean target gene Ctmean -actin gene). The difference in the level of each normalized target gene was obtained (Ct experimental group Ct control group), and the fold difference was calculated using the equation 2CT.

To analyze blood samples, the whole blood was collected in K2EDTA collection tubes (Terumo Medical, Somerset, NJ, USA). To analyze spleen samples, the tissue was digested with Accumax solution to generate cell suspension. After red blood cell lysis, cells were blocked with 2% BSA and incubated with the fluorescence-conjugated antibodies in the dark for 30 min at 4C. For intracellular cytokine staining, splenocytes were incubated in a cell culture incubator for 10 hours with phorbol 12-myristate 13-acetate (50 ng/ml), ionomycin (750 ng/ml), and brefeldin A (10 mg/ml; all from Sigma-Aldrich). Surface staining was performed as described above. After surface staining, the cells were resuspended in a fixation/permeabilization solution (Cytofix/Cytoperm Kit, Multi Sciences, Hangzhou, China). The intracellular cytokine staining was performed according to the manufacturers protocol. To analyze the frequencies of Treg cells in the spleens, samples were prepared and detected following the manufacturers protocol (Mouse Regulatory T Cell Staining Kit, Multi Sciences). To determine the total number of GFP-positive hepatocytes in the spleen, total cells were resuspended in 1 ml of 2% BSA and mixed well after red blood cell lysing. Then, 100 l of the mixture was transferred to a new test tube to determine the number of GFP-positive hepatocytes. Each sample was measured three times, and the total number of GFP-positive hepatocytes was calculated by multiplying the average of the three results by 10. Flow cytometry data were acquired on FACSCalibur (BD Biosciences) and analyzed using FlowJo software (Tree Star, San Diego, CA, USA). For sorting the GFP-positive hepatocytes in the spleen, a FACS machine (FACSAria II, BD Biosciences) was used to separate the negative cells and the GFP-positive after red blood cell lysing.

Blood samples were collected, allowed to coagulate at room temperature, and centrifuged to obtain serum. The spleen was homogenized in equal volume of PBS to obtain 50% (w/v) homogenate and then centrifuged at 12,000 rpm for 10 min at 4C to harvest supernatants. The levels of HSA, TNF-, IL-1, IFN-, IL-10, IL-12p70, TGF-1, TGF-2, VEGF, EGF, hepatocyte growth factor, IgG, IgM, COL1, COL4, phosphorylatedextracellular signalregulated kinase 1/2 (P-ERK 1/2), and ERK 1/2 were measured using corresponding ELISA quantitation kits (Abcam, Cambridge, UK) according to the manufacturers instructions.

Proteins were extracted from tissue homogenate in radioimmunoprecipitation assay buffer (Beyotime Biotechnology) with a protease inhibitor (Sigma-Aldrich) and spun at 6000g for 10 min at 4C. The lysate was resolved by SDSpolyacrylamide gel electrophoresis, transferred to a polyvinylidene difluoride membrane using Mini-PROTEAN Tetra Electrophoresis System (Bio-Rad, CA, USA), and probed. Horseradish peroxidase (HRP)linked anti-rabbit (Jackson ImmunoResearch, West Grove, PA, USA) was used, and the HRP signal was visualized with 4200SF (Tanon, Shanghai, China).

The hardness of the spleen was measured using an elasticity-measuring instrument equipped with a coil spring (CL-150SL Type A Durometer, ASKER, Kyoto, Japan). To offer adequate thickness, five spleen samples were stacked together for the measurement. A constant force was applied to tissue samples, and the mean value representing three individual tests was recorded.

The content of hydroxyproline in tissue was measured using a hydroxyproline assay kit (Jiancheng Bioengineering Institute) to quantify collagen content following the manufacturers protocol. The data are expressed as hydroxyproline (in micrograms)/tissue wet weight (in grams).

Each mouse was examined in supine position after a rest of 15 min to avoid influence of posture and exercise. All subjects were scanned on a micro-ultrasound system with a 24-MHz transducer (Vevo 2100, VisualSonics, Canada). B-mode was used to display anatomical structures in two-dimensional (2D) grayscale image. Color Doppler mode was used to display blood flow in the portal vein. Pulse-wave Doppler mode was used to quantify the blood flow through the vessel.

To assess the biodistribution of TGF-1, TGF-1 (PeproTech) was labeled with fluorescein isothiocyanate (FITC; Sigma-Aldrich). Briefly, FITC (1 mg/ml) was dissolved in ethyl alcohol and then was added to 0.05 M PBS (pH 7.2) containing TGF-1 (1 mg/ml) at a ratio of 1:100 w/w (dye:protein). After 24 hours of stirring under light protection, the FITC-labeled TGF-1 was purified and concentrated to 25 mg/ml using Amicon Ultra-4 device (3000 molecular weight cutoff; Sigma-Aldrich). Thirty minutes after intrasplenic injection of the FITC-labeled TGF-1, the spleen was removed and immediately embedded in OCT for sectioning. The sections were photographed using a Leica TCS SP8 confocal microscope.

To trace the flow of bile from the spleen, a fluorescence-conjugated bile acid analog was prepared. Briefly, a green fluorescent compound, N-butyl-4-(2-hydroxyethylamino)-1,8-naphtalimide (NNOH), was synthesized with a reported method (47). NNOH (7.5 mg/ml) was dissolved in dimethylformamide. Then, cholic acid (20.4 mg/ml), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (9.55 mg/ml), and 4-dimethylaminopyridine (1.25 mg/ml; all from Sigma-Aldrich) were added. The mixture was stirred at room temperature overnight. The solution was evaporated under vacuum. The residue was dissolved in CH2Cl2, washed with distilled water and brine, dried with MgSO4, and purified by silica gel column chromatography (48).

IVIS Lumina XR System (Caliper Life Sciences, Hopkinton, MA, USA) was used to detect the donor hepatocytes (luciferase transgenic) and the direction of bile flow from the spleen in vivo. To test hepatocyte proliferation, the fluorescence intensity gated over the spleen was measured and analyzed using Living Image software (Caliper Life Sciences) after intraperitoneal injection of luciferin substrate (300 mg/kg of body weight; Promega, Madison, WI, USA). For tracing the bile flow from the spleen, the fluorescence intensity gated over the total body was detected and analyzed after intrasplenic injection of the fluorescence-conjugated bile acid (1 mg/kg of body weight).

The mice were examined with micro-CT for the regenerated tissue and vascular network in the spleen. The animals were perfused with 1 ml of iohexol (GE Healthcare AS, Munich, Germany) and euthanized 5 min after the injection of the contrast agent. The mice were scanned using the Skyscan 1176 (Bruker micro-CT, Brussels, Belgium) system at a resolution of 12.59 M and a rotation step of 0.9830. The system comprised two metalloceramic tubes equipped with a fixed Al filter (0.5 mm) and two digital x-ray cameras (1280 by 1024 pixels). Images were acquired at 50 kV and 455 A. The 3D images of the vessels were reconstructed using CTvol (Bruker micro-CT), and the blood vessel density in the spleen was determined using CTan (Bruker micro-CT).

Blood samples were obtained in a vacuum blood collection tube. Prothrombin time was measured with a Sysmex CS-5100 System (Siemens Healthineers, Erlangen, Germany).

Blood ammonia in serum was quantified using the corresponding kit according to the manufacturers instruction (Jiancheng Bioengineering Institute).

AST, ALT, blood urea nitrogen, creatine kinase, and lactate dehydrogenase in the serum were evaluated using corresponding kits according to the manufacturers instructions (Jiancheng Bioengineering Institute).

Blood samples were collected and subjected to the counting for platelets, leukocytes, erythrocytes, hemoglobin, hematocrit, mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, red cell distribution width, thrombocytocrit, and mean platelet volume on Sysmex xe-5000 (Siemens Healthineers).

Data are shown as means SEM. All data were normally distributed. Statistical analysis was performed using Prism Software (GraphPad, USA). Students t test and one-way and two-way analysis of variance (ANOVA) were performed, followed by Bonferronis multiple comparison test and log-rank test. Results were considered significant at P < 0.05.

Acknowledgments: Funding: The study was funded by the National Key Research and Development Program of China (2017YFC0909702), the National Natural Science Foundation of China (81973273, 81673380, 31971309, and 31671031), the Jiangsu Province Funds for Distinguished Young Scientists (BK20170015), the Fundamental Research Funds for the Central Universities (020814380115), The Science and Technology Development Fund, Macau SAR (File no. 0018/2019/AFJ and 0097/2019/A2), University of Macau (UM) (MYRG2017-00028-ICMS and MYRG2019-00080-ICMS), the International Cooperation and Exchange of the Natural Science Foundation of China and the Science and Technology Development Fund (31961160701), and the Jiangsu Province Postdoctoral Research Foundation (2019K147). L.W., Z.W., and Y.N. acknowledge UM Macao Postdoctoral Fellowship/Associateship (UMPF/UMPA). Author contributions: L.W. and L.D. designed, performed, and analyzed all experiments and wrote the manuscript. J.Z. designed the study. C.W. designed the study and wrote the manuscript. C.L. and S.X. performed the histopathological analysis. Z.W. wrote the manuscript. Y.N., J.G., and D.C. performed data analysis. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. Specifically, the microarray data have been submitted to the Gene Expression Omnibus under accession number GSE115107.

See the original post:
Transforming the spleen into a liver-like organ in vivo - Science Advances